1
|
Zeng F, Zhou M, Li Q, Hu H, Chen C. Sevoflurane promotes neuronal ferroptosis via upregulation of PLIN4 to modulate the hippo signaling pathway. Neurotoxicology 2024; 105:1-9. [PMID: 39182851 DOI: 10.1016/j.neuro.2024.08.001] [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: 05/29/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Sevoflurane is a widely used inhalation anesthetic associated with neuronal damage, cognitive impairment and neurodegenerative diseases, with iron overload reported to contribute to these adverse effects. However, the mechanisms of iron-dependent cell death (ferroptosis) in sevoflurane-induced neurotoxicity remain poorly understood. METHODS The role of PLIN4, a protein associated with neurodegeneration, in sevoflurane-induced neuronal damage was investigated using cultured mouse hippocampal neurons (HT22). PLIN4 knockdown or overexpression was performed through vector transfection, and PLIN4 transcription and expression levels after sevoflurane treatment and knockdown experiments were assessed via RT-qPCR, immunostaining, and western blot to evaluate its impact on ferroptosis. Transmission electron microscopy was used to assess cellular morphology and measure Fe2+ levels. RESULTS Sevoflurane treatment significantly increased PLIN4 expression in hippocampal neurons and induced ferroptosis. Silencing PLIN4 reduced ferroptosis and partially reversed sevoflurane's inhibition of the Hippo signaling pathway. Specifically, sevoflurane treatment led to a 2.9-fold increase in PLIN4 mRNA levels. Furthermore, higher PLIN4 levels upregulated ferroptosis in hippocampal neurons by inhibiting the Hippo pathway. CONCLUSION Our study indicates that sevoflurane promotes ferroptosis in neurons by upregulating PLIN4 and modulating the Hippo signaling pathway. These findings provide insights into the potential development of interventions to prevent anesthesia-related cognitive impairments and neurodegeneration.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu City, PR China
| | - Mingxia Zhou
- Wuhan Xinzhou District People's Hospital, Wuhan City, Hubei Province, PR China
| | - Qiang Li
- Department of Anesthesiology, The Third People's Hospital of Chengdu, Southwest Jiao Tong University, Chengdu City, Sichuan Province, PR China
| | - Huan Hu
- Department of Surgery Intensive Care Unit, People's Hospital of Sichuan Province, School of Medicine University of Electronic Science and Technology of China, Chengdu City, Sichuan Province, PR China
| | - Chen Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan City, Hubei Province 430022, PR China.
| |
Collapse
|
2
|
Iketani M, Hatomi M, Fujita Y, Watanabe N, Ito M, Kawaguchi H, Ohsawa I. Inhalation of hydrogen gas mitigates sevoflurane-induced neuronal apoptosis in the neonatal cortex and is associated with changes in protein phosphorylation. J Neurochem 2024; 168:2775-2790. [PMID: 38849977 DOI: 10.1111/jnc.16142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/03/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024]
Abstract
Inhalation of hydrogen (H2) gas is therapeutically effective for cerebrovascular diseases, neurodegenerative disorders, and neonatal brain disorders including pathologies induced by anesthetic gases. To understand the mechanisms underlying the protective effects of H2 on the brain, we investigated the molecular signals affected by H2 in sevoflurane-induced neuronal cell death. We confirmed that neural progenitor cells are susceptible to sevoflurane and undergo apoptosis in the retrosplenial cortex of neonatal mice. Co-administration of 1-8% H2 gas for 3 h to sevoflurane-exposed pups suppressed elevated caspase-3-mediated apoptotic cell death and concomitantly decreased c-Jun phosphorylation and activation of the c-Jun pathway, all of which are induced by oxidative stress. Anesthesia-induced increases in lipid peroxidation and oxidative DNA damage were alleviated by H2 inhalation. Phosphoproteome analysis revealed enriched clusters of differentially phosphorylated proteins in the sevoflurane-exposed neonatal brain that included proteins involved in neuronal development and synaptic signaling. H2 inhalation modified cellular transport pathways that depend on hyperphosphorylated proteins including microtubule-associated protein family. These modifications may be involved in the protective mechanisms of H2 against sevoflurane-induced neuronal cell death.
Collapse
Affiliation(s)
- Masumi Iketani
- Biological Process of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Mai Hatomi
- Biological Process of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Department of Life Sciences, Toyo University, Asaka, Japan
| | - Yasunori Fujita
- Biological Process of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Nobuhiro Watanabe
- Autonomic Neuroscience, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masafumi Ito
- Biological Process of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | | | - Ikuroh Ohsawa
- Biological Process of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
3
|
Sun H, Yisi Shan, Cao L, Wu X, Chen J, Yuan R, Qian M. Unveiling the hidden dangers: a review of non-apoptotic programmed cell death in anesthetic-induced developmental neurotoxicity. Cell Biol Toxicol 2024; 40:63. [PMID: 39093513 PMCID: PMC11297112 DOI: 10.1007/s10565-024-09895-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/21/2024] [Indexed: 08/04/2024]
Abstract
Anesthetic-induced developmental neurotoxicity (AIDN) can arise due to various factors, among which aberrant nerve cell death is a prominent risk factor. Animal studies have reported that repeated or prolonged anesthetic exposure can cause significant neuroapoptosis in the developing brain. Lately, non-apoptotic programmed cell deaths (PCDs), characterized by inflammation and oxidative stress, have gained increasing attention. Substantial evidence suggests that non-apoptotic PCDs are essential for neuronal cell death in AIDN compared to apoptosis. This article examines relevant publications in the PubMed database until April 2024. Only original articles in English that investigated the potential manifestations of non-apoptotic PCD in AIDN were analysed. Specifically, it investigates necroptosis, pyroptosis, ferroptosis, and parthanatos, elucidating the signaling mechanisms associated with each form. Furthermore, this study explores the potential relevance of these non-apoptotic PCDs pathways to the pathological mechanisms underlying AIDN, drawing upon their distinctive characteristics. Despite the considerable challenges involved in translating fundamental scientific knowledge into clinical therapeutic interventions, this comprehensive review offers a theoretical foundation for developing innovative preventive and treatment strategies targeting non-apoptotic PCDs in the context of AIDN.
Collapse
Affiliation(s)
- Haiyan Sun
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
| | - Liyan Cao
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
| | - Xiping Wu
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jiangdong Chen
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China
| | - Rong Yuan
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China.
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China.
| | - Min Qian
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China.
- Department of Anesthesiology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, China.
| |
Collapse
|
4
|
Xu L, Ma S, Qu M, Li N, Sun X, Wang T, Chen L, Zhu J, Ding Y, Gong Y, Hu F, Dong Z, Zhang R, Wang JH, Wang J, Zhou H. Parthanatos initiated by ROS-induced DNA damage is involved in intestinal epithelial injury during necrotizing enterocolitis. Cell Death Discov 2024; 10:345. [PMID: 39085218 PMCID: PMC11291915 DOI: 10.1038/s41420-024-02114-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/13/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Necrotizing enterocolitis (NEC) involves intestinal epithelial damage and inflammatory response and is associated with high morbidity and mortality in infants. To improve therapeutic prospects, elucidating underlying molecular mechanisms of intestinal epithelial damage during NEC is of the essence. Poly (ADP-ribose) polymerase 1 (PARP1)-dependent parthanatos is a programmed inflammatory cell death. In the present study, the presence of parthanatos-associated proteins PARP1 and poly (ADP-ribose) (PAR), along with high expression of DNA damage-associated biomarkers, 8-hydroxy-2'-deoxyguanosine (8-OHdG) and phosphorylation of histone H2AX (γH2AX), were discovered in the intestinal tissues of NEC infants. Additionally, the upregulated expression of PARP1 and PAR in NEC intestinal tissues correlated distinctly with clinical indices indicative of NEC incidence and severity. Furthermore, we demonstrated that inhibiting the expression of parthanatos-associated proteins, by either pharmacological blockage using 3-aminobenzamide (3-AB), an inhibitor of PARP1, or genetic knockout using Parp1-deficient mice, resulted in substantial improvements in both histopathological severity scores associated with intestinal injury and inflammatory reactions. Moreover, in an in vitro NEC model, reactive oxygen species (ROS)-induced DNA damage promoted the formation of PAR and nuclear translocation of apoptosis-inducing factor (AIF), thus activating PARP1-dependent parthanatos in Caco-2 cells and human intestinal organoids. Our work verifies a previously unexplored role for parthanatos in intestinal epithelial damage during NEC and suggests that inhibition of parthanatos may serve as a potential therapeutic strategy for intervention of NEC.
Collapse
Affiliation(s)
- Lingqi Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Shurong Ma
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Minhan Qu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xu Sun
- Department of Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Tingting Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lulu Chen
- Department of Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Jie Zhu
- Department of Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Yifang Ding
- Department of Pediatrics, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yuan Gong
- Department of Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Fangjie Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Zhenzhen Dong
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Rui Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
- Department of Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Huiting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Meng X, Wang Y, Zhao W, Chen Y, Li W, Peng K, Xu H, Yang Y, Shan X, Huo W, Liu H, Ji F. Identification of differential m6A RNA methylomes and ALKBH5 as a potential prevention target in the developmental neurotoxicity induced by multiple sevoflurane exposures. FASEB J 2024; 38:e23793. [PMID: 39003634 DOI: 10.1096/fj.202400664r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Sevoflurane, as a commonly used inhaled anesthetic for pediatric patients, has been reported that multiple sevoflurane exposures are associated with a greater risk of developing neurocognitive disorder. N6-Methyladenosine (m6A), as the most common mRNA modification in eukaryotes, has emerged as a crucial regulator of brain function in processes involving synaptic plasticity, learning and memory, and neurodevelopment. Nevertheless, the relevance of m6A RNA methylation in the multiple sevoflurane exposure-induced developmental neurotoxicity remains mostly elusive. Herein, we evaluated the genome-wide m6A RNA modification and gene expression in hippocampus of mice that received with multiple sevoflurane exposures using m6A-sequencing (m6A-seq) and RNA-sequencing (RNA-seq). We discovered 19 genes with differences in the m6A methylated modification and differential expression in the hippocampus. Among these genes, we determined that a total of nine differential expressed genes may be closely associated with the occurrence of developmental neurotoxicity induced by multiple sevoflurane exposures. We further found that the alkB homolog 5 (ALKBH5), but not methyltransferase-like 3 (METTL3) and Wilms tumor 1-associated protein (WTAP), were increased in the hippocampus of mice that received with multiple sevoflurane exposures. And the IOX1, as an inhibitor of ALKBH5, significantly improved the learning and memory defects and reduced neuronal damage in the hippocampus of mice induced by multiple sevoflurane exposures. The current study revealed the role of m6A methylated modification and m6A-related regulators in sevoflurane-induced cognitive impairment, which might provide a novel insight into identifying biomarkers and therapeutic strategies for inhaled anesthetic-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Xiaowen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Yichan Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Weiming Zhao
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Ying Chen
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wenting Li
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Hanbing Xu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Yufan Yang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Xisheng Shan
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Wenwen Huo
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Huayue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
- Ambulatory Surgery Center, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
6
|
Koo BW, Shin HJ, Jeon S, Bang JH, Do SH, Na HS. Neuroprotective effect of erythropoietin on anesthesia-induced neurotoxicity through the modulation of autophagy in Caenorhabditis elegans. Korean J Anesthesiol 2024; 77:384-391. [PMID: 38356139 PMCID: PMC11150124 DOI: 10.4097/kja.23789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The anti-oxidative, anti-inflammatory, and anti-apoptotic effects of erythropoietin may provide neuroprotective effects. Erythropoietin also modulates autophagy signaling that may play a role in anesthesia-induced neurotoxicity (AIN). Herein, we investigated whether AIN can be attenuated by the neuroprotective effect of erythropoietin in the Caenorhabditis elegans (C. elegans). METHODS Synchronized worms were divided into the control, Iso, EPO, and EPO-Iso groups. The chemotaxis index (CI) was evaluated when they reached the young adult stage. The lgg-1::GFP-positive puncta per seam cell were used to determine the autophagic events. The erythropoietin-mediated pathway of autophagy was determined by measuring the genetic expression level of let-363, bec-1, atg-7, atg-5, and lgg-3. RESULTS Increased lgg-1::GFP puncta were observed in the Iso, EPO, and EPO-Iso groups. In the Iso group, only the let-363 level decreased significantly as compared to that in the control group (P = 0.009). bec-1 (P < 0.001), atg-5 (P = 0.012), and lgg-3 (P < 0.001) were expressed significantly more in the EPO-Iso group than in the Iso groups. Repeated isoflurane exposure during development decreased the CI. Erythropoietin could restore the decreased CI by isoflurane significantly in the EPO-Iso group. CONCLUSIONS Erythropoietin showed neuroprotective effects against AIN and modulated the autophagic pathway in C. elegans. This experimental evidence of erythropoietin-related neuroprotection against AIN may be correlated with the induced autophagic degradation process that was sufficient for handling enhanced autophagy induction in erythropoietin-treated worms.
Collapse
Affiliation(s)
- Bon-Wook Koo
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Anesthesiology and Pain Medicine, Seoul National University, Seoul, Korea
| | - Hyun-Jung Shin
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Anesthesiology and Pain Medicine, Seoul National University, Seoul, Korea
| | - Sooyoung Jeon
- National Dental Care Center for Persons with Special Needs, Seoul National University Dental Hospital, Seoul, Korea
| | - Jung Hyun Bang
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang-Hwan Do
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Anesthesiology and Pain Medicine, Seoul National University, Seoul, Korea
| | - Hyo-Seok Na
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Anesthesiology and Pain Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Kang L, Piao M, Liu N, Gu W, Feng C. Sevoflurane Exposure Induces Neuronal Cell Ferroptosis Initiated by Increase of Intracellular Hydrogen Peroxide in the Developing Brain via ER Stress ATF3 Activation. Mol Neurobiol 2024; 61:2313-2335. [PMID: 37874483 DOI: 10.1007/s12035-023-03695-z] [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: 05/11/2023] [Accepted: 10/04/2023] [Indexed: 10/25/2023]
Abstract
Neuronal cell death is acknowledged as the primary pathological basis underlying developmental neurotoxicity in response to sevoflurane exposure, but the exact mechanism remains unclear. Ferroptosis is a form of programmed cell death characterized by iron-dependent lipid peroxidation that is driven by hydrogen peroxide (H2O2) and ferrous iron through the Fenton reaction and participates in the pathogenesis of multiple neurological diseases. As stress response factor, activating transcription factor 3 (ATF3) can be activated by the PERK/ATF4 pathway during endoplasmic reticulum (ER) stress, followed by increased intracellular H2O2, which is involved in regulation of apoptosis, autophagy, and ferroptosis. Here, we investigated whether ferroptosis and ATF3 activation were implicated in sevoflurane-induced neuronal cell death in the developing brain. The results showed that sevoflurane exposure induced neuronal death as a result of iron-dependent lipid peroxidation damage secondary to H2O2 accumulation and ferrous iron increase, which was consistent with the criteria for ferroptosis. Furthermore, we observed that increases in iron and H2O2 induced by sevoflurane exposure were associated with the upregulation and nuclear translocation of ATF3 in response to ER stress. Knockdown of ATF3 expression alleviated iron-dependent lipid peroxidation, which prevented sevoflurane-induced neuronal ferroptosis. Mechanistically, ATF3 promoted sevoflurane-induced H2O2 accumulation by activating NOX4 and suppressing catalase, GPX4, and SLC7A11 expression. Additionally, an increase in H2O2 was accompanied by the upregulation of TFR and TF and downregulation of FPN, which linked iron overload to ferroptosis induced by sevoflurane. Taken together, our results demonstrated that ER stress-mediated ATF3 activation contributed to sevoflurane-induced neuronal ferroptosis via H2O2 accumulation and the resultant iron overload.
Collapse
Affiliation(s)
- Liheng Kang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Wanping Gu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China.
| |
Collapse
|
8
|
Leonov G, Salikhova D, Shedenkova M, Bukharova T, Fatkhudinov T, Goldshtein D. Comparative Study of the Protective and Neurotrophic Effects of Neuronal and Glial Progenitor Cells-Derived Conditioned Media in a Model of Glutamate Toxicity In Vitro. Biomolecules 2023; 13:1784. [PMID: 38136654 PMCID: PMC10741670 DOI: 10.3390/biom13121784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/16/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Cell therapy represents a promising approach to the treatment of neurological diseases, offering potential benefits not only by cell replacement but also through paracrine secretory activities. However, this approach includes a number of limiting factors, primarily related to safety. The use of conditioned stem cell media can serve as an equivalent to cell therapy while avoiding its disadvantages. The present study was a comparative investigation of the antioxidant, neuroprotective and neurotrophic effects of conditioned media obtained from neuronal and glial progenitor cells (NPC-CM and GPC-CM) on the PC12 cell line in vitro. Neuronal and glial progenitor cells were obtained from iPSCs by directed differentiation using small molecules. GPC-CM reduced apoptosis, ROS levels and increased viability, expressions of the antioxidant response genes HMOX1 and NFE2L2 in a model of glutamate-induced oxidative stress. The neurotrophic effect was evidenced by a change in the morphology of pheochromocytoma cells to a neuron-like phenotype. Moreover, neurite outgrowth, expression of GAP43, TUBB3, MAP2, SYN1 genes and increased levels of the corresponding MAP2 and TUBB3 proteins. Treatment with NPC-CM showed moderate antiapoptotic effects and improved cell viability. This study demonstrated the potential application of CM in the field of regenerative medicine.
Collapse
Affiliation(s)
- Georgy Leonov
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (D.S.); (M.S.); (T.B.); (D.G.)
- Orekhovich Institute of Biomedical Chemistry of the Russian Academy of Sciences, 119121 Moscow, Russia
| | - Diana Salikhova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (D.S.); (M.S.); (T.B.); (D.G.)
- Research Institute of Molecular and Cellular Medicine, Medical Institute RUDN, 117198 Moscow, Russia;
| | - Margarita Shedenkova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (D.S.); (M.S.); (T.B.); (D.G.)
- Research Institute of Molecular and Cellular Medicine, Medical Institute RUDN, 117198 Moscow, Russia;
| | - Tatiana Bukharova
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (D.S.); (M.S.); (T.B.); (D.G.)
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Medical Institute RUDN, 117198 Moscow, Russia;
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (D.S.); (M.S.); (T.B.); (D.G.)
- Research Institute of Molecular and Cellular Medicine, Medical Institute RUDN, 117198 Moscow, Russia;
| |
Collapse
|
9
|
Boulos JC, Omer EA, Rigano D, Formisano C, Chatterjee M, Leich E, Klauck SM, Shan LT, Efferth T. Cynaropicrin disrupts tubulin and c-Myc-related signaling and induces parthanatos-type cell death in multiple myeloma. Acta Pharmacol Sin 2023; 44:2265-2281. [PMID: 37344563 PMCID: PMC10618500 DOI: 10.1038/s41401-023-01117-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/28/2023] [Indexed: 06/23/2023] Open
Abstract
The majority of blood malignancies is incurable and has unforeseeable remitting-relapsing paths in response to different treatments. Cynaropicrin, a natural sesquiterpene lactone from the edible parts of the artichoke plant, has gained increased attention as a chemotherapeutic agent. In this study, we investigated the effects of cynaropicrin against multiple myeloma (MM) cells in vitro and assessed its in vivo effectiveness in a xenograft tumor zebrafish model. We showed that cynaropicrin exerted potent cytotoxicity against a panel of nine MM cell lines and two leukemia cell lines with AMO1 being the most sensitive cell line (IC50 = 1.8 ± 0.3 µM). Cynaropicrin (0.8, 1.9, 3.6 µM) dose-dependently reduced c-Myc expression and transcriptional activity in AMO1 cells that was associated with significant downregulation of STAT3, AKT, and ERK1/2. Cell cycle analysis showed that cynaropicrin treatment arrested AMO1 cells in the G2M phase along with an increase in the sub-G0G1 phase after 24 h. With prolonged treatment times, cells accumulated more in the sub-G0G1 phase, implying cell death. Using confocal microscopy, we revealed that cynaropicrin disrupted the microtubule network in U2OS cells stably expressing α-tubulin-GFP. Furthermore, we revealed that cynaropicrin promoted DNA damage in AMO1 cells leading to PAR polymer production by PARP1 hyperactivation, resulting in AIF translocation from the mitochondria to the nucleus and subsequently to a novel form of cell death, parthanatos. Finally, we demonstrated that cynaropicrin (5, 10 µM) significantly reduced tumor growth in a T-cell acute lymphoblastic leukemia (T-ALL) xenograft zebrafish model. Taken together, these results demonstrate that cynaropicrin causes potent inhibition of hematopoietic tumor cells in vitro and in vivo.
Collapse
Affiliation(s)
- Joelle C Boulos
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Ejlal A Omer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany
| | - Daniela Rigano
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Carmen Formisano
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Manik Chatterjee
- University Hospital Würzburg, Translational Oncology, Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Ellen Leich
- Julius Maximilian University, Institute of Pathology, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Translational Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Le-Tian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128, Mainz, Germany.
| |
Collapse
|
10
|
Wang L, Ye B, Liu Y, Li J, Li C, Wen M, Li H. Xuebijing Injection Attenuates Heat Stroke-Induced Brain Injury through Oxidative Stress Blockage and Parthanatos Modulation via PARP-1/AIF Signaling. ACS OMEGA 2023; 8:33392-33402. [PMID: 37744847 PMCID: PMC10515343 DOI: 10.1021/acsomega.3c03084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/19/2023] [Indexed: 09/26/2023]
Abstract
Heat stroke (HS) is a potentially fatal acute condition caused by an interplay of complex events including inflammation, endothelial injury, and coagulation abnormalities that make its pharmacological treatment a challenging problem. The traditional Chinese medicine Xuebijing injection (XBJ) has been shown to reduce inflammatory responses and prevent organ injuries in HS-induced mice. However, the underlying mechanism of XBJ in HS-induced brain injury remains unclear. In this study, HS-induced rat models and cell models were established to elucidate the effects and underlying mechanisms of XBJ injection on HS-induced brain injury in vivo and in vitro. The results revealed that XBJ injection improved the survival outcome of HS rats and attenuated HS-induced brain injury in a concentration-dependent manner. Subsequently, the reduction in viability and proliferation of neurons induced by HS were reversed by XBJ treatment, while the HS-induced increased ROS levels and neuron death were also inhibited by XBJ injection. Mechanistically, HS activated PARP-1/AIF signaling in vitro and in vivo, inducing the translocation of AIF from the cytoplasm to the nucleus, leading to PARP-1-dependent cell death of neurons. Additionally, we compared XBJ injection effects in young and old age rats. Results showed that XBJ also provided protective effects in HS-induced brain injury in aging rats; however, the treatment efficacy of XBJ injection at the same concentration was more significant in the young age rats. In conclusion, XBJ injection attenuates HS-induced brain injury by inhibiting oxidative stress and Parthanatos via the PARP-1/AIF signaling, which might provide a novel therapeutic strategy for HS treatment.
Collapse
Affiliation(s)
- Lin Wang
- Department
of Emergency, The First Affiliated Hospital
of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Boxin Ye
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Yongrui Liu
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Jun Li
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Chunhe Li
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Minyong Wen
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Hongbo Li
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| |
Collapse
|
11
|
Hogarth K, Tarazi D, Maynes JT. The effects of general anesthetics on mitochondrial structure and function in the developing brain. Front Neurol 2023; 14:1179823. [PMID: 37533472 PMCID: PMC10390784 DOI: 10.3389/fneur.2023.1179823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
The use of general anesthetics in modern clinical practice is commonly regarded as safe for healthy individuals, but exposures at the extreme ends of the age spectrum have been linked to chronic cognitive impairments and persistent functional and structural alterations to the nervous system. The accumulation of evidence at both the epidemiological and experimental level prompted the addition of a warning label to inhaled anesthetics by the Food and Drug Administration cautioning their use in children under 3 years of age. Though the mechanism by which anesthetics may induce these detrimental changes remains to be fully elucidated, increasing evidence implicates mitochondria as a potential primary target of anesthetic damage, meditating many of the associated neurotoxic effects. Along with their commonly cited role in energy production via oxidative phosphorylation, mitochondria also play a central role in other critical cellular processes including calcium buffering, cell death pathways, and metabolite synthesis. In addition to meeting their immense energy demands, neurons are particularly dependent on the proper function and spatial organization of mitochondria to mediate specialized functions including neurotransmitter trafficking and release. Mitochondrial dependence is further highlighted in the developing brain, requiring spatiotemporally complex and metabolically expensive processes such as neurogenesis, synaptogenesis, and synaptic pruning, making the consequence of functional alterations potentially impactful. To this end, we explore and summarize the current mechanistic understanding of the effects of anesthetic exposure on mitochondria in the developing nervous system. We will specifically focus on the impact of anesthetic agents on mitochondrial dynamics, apoptosis, bioenergetics, stress pathways, and redox homeostasis. In addition, we will highlight critical knowledge gaps, pertinent challenges, and potential therapeutic targets warranting future exploration to guide mechanistic and outcomes research.
Collapse
Affiliation(s)
- Kaley Hogarth
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Doorsa Tarazi
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Jason T. Maynes
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Lu P, Liang F, Dong Y, Xie Z, Zhang Y. Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration. Int J Mol Sci 2023; 24:ijms24076746. [PMID: 37047719 PMCID: PMC10095407 DOI: 10.3390/ijms24076746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
Collapse
Affiliation(s)
- Pan Lu
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
13
|
The natural product dehydrocurvularin induces apoptosis of gastric cancer cells by activating PARP-1 and caspase-3. Apoptosis 2023; 28:525-538. [PMID: 36652130 DOI: 10.1007/s10495-023-01811-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
The natural product dehydrocurvularin (DSE2) is a fungal-derived macrolide with potent anticancer activity, but the mechanism is still unclear. We found that DSE2 effectively inhibited the growth of gastric cancer cells and induced the apoptosis by activating Poly(ADP-ribose) polymerase 1 (PARP-1) and caspase-3. Pharmacological inhibition and genetic knockdown with PARP-1 or caspase-3 suppressed DSE2-induced apoptosis. PARP-1 was previously reported to be cleaved into fragments during apoptosis. However, PARP-1 was barely cleaved in DSE2-induced apoptosis. DSE2 induced PARP-1 activation as indicated by rapid depletion of NAD+ and the concomitant formation of poly(ADP-ribosylated) proteins (PARs). Interestingly, the PARP-1 inhibitor (Olaparib) attenuated the cytotoxicity of DSE2. Moreover, the combination of Olaparib and Z-DEVD-FMK (caspase-3 inhibitor) further reduced the cytotoxicity. It has been shown that PARP-1 activation triggers cytoplasm-nucleus translocation of apoptosis-inducing factor (AIF). Caspase-3 inhibitors inhibited PARP-1 activation and suppressed PARP-1-induced AIF nuclear translocation. These results indicated that DSE2-induced caspase-3 activation may occur before PARP-1 activation. The ROS inhibitor, N-acetyl-cysteine, significantly inhibited the activation of caspase-3 and PARP-1, indicating that ROS overproduction contributed to DSE2-induced apoptosis. Using an in vivo approach, we further found that DSE2 significantly inhibited gastric tumor growth and promoted translocation of AIF to the nucleus. In conclusion, DSE2 induces gastric cell apoptosis by activating caspase-3 and PARP-1, and shows potent antitumor activity against human gastric carcinoma in vitro and in vivo.
Collapse
|
14
|
Liu LF, Hu Y, Liu YN, Shi DW, Liu C, Da X, Zhu SH, Zhu QY, Zhang JQ, Xu GH. Reactive oxygen species contribute to delirium-like behavior by activating CypA/MMP9 signaling and inducing blood-brain barrier impairment in aged mice following anesthesia and surgery. Front Aging Neurosci 2022; 14:1021129. [PMID: 36337710 PMCID: PMC9629746 DOI: 10.3389/fnagi.2022.1021129] [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: 08/17/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Postoperative delirium (POD) is common in the elderly and is associated with poor clinical outcomes. Reactive oxygen species (ROS) and blood-brain barrier (BBB) damage have been implicated in the development of POD, but the association between these two factors and the potential mechanism is not clear. Cyclophilin A (CypA) is a specifically chemotactic leukocyte factor that can be secreted in response to ROS, which activates matrix metalloproteinase 9 (MMP9) and mediates BBB breakdown. We, therefore, hypothesized that ROS may contribute to anesthesia/surgery-induced BBB damage and delirium-like behavior via the CypA/MMP9 pathway. To test these hypotheses, 16-month-old mice were subjected to laparotomy under 3% sevoflurane anesthesia (anesthesia/surgery) for 3 h. ROS scavenger (N-acetyl-cysteine) and CypA inhibitor (Cyclosporin A) were used 0.5 h before anesthesia/surgery. A battery of behavior tests (buried food test, open field test, and Y maze test) was employed to evaluate behavioral changes at 24 h before and after surgery in the mice. Levels of tight junction proteins, CypA, MMP9, postsynaptic density protein (PSD)-95, and synaptophysin in the prefrontal cortex were assessed by western blotting. The amounts of ROS and IgG in the cortex of mice were observed by fluorescent staining. The concentration of S100β in the serum was detected by ELISA. ROS scavenger prevented the reduction in TJ proteins and restored the permeability of BBB as well as reduced the levels of CypA/MMP9, and further alleviated delirium-like behavior induced by anesthesia/surgery. Furthermore, the CypA inhibitor abolished the increased levels of CypA/MMP, which reversed BBB damage and ameliorated delirium-like behavior caused by ROS accumulation. Our findings demonstrated that ROS may participate in regulating BBB permeability in aged mice with POD via the CypA/MMP9 pathway, suggesting that CypA may be a potential molecular target for preventing POD.
Collapse
Affiliation(s)
- Li-fang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Yun Hu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Yi-nuo Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - De-wen Shi
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Chang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Xin Da
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Si-hui Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Qian-yun Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Ji-qian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
| | - Guang-hong Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, China
- *Correspondence: Guang-hong Xu,
| |
Collapse
|
15
|
Shu L, Du C. PHLDA1 promotes sevoflurane-induced pyroptosis of neuronal cells in developing rats through TRAF6-mediated activation of Rac1. Neurotoxicology 2022; 93:140-151. [PMID: 36155068 DOI: 10.1016/j.neuro.2022.09.007] [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: 06/16/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Sevoflurane anesthesia induces neurocognitive impairment and pyroptosis in the developing brain. Pleckstrin homology-like domain, family A, member 1 (PHLDA1) was involved in neuronal apoptosis, oxidative stress and inflammation during ischemic stroke. The role of PHLDA1 in sevoflurane-induced pyroptosis in developing rats was investigated. Firstly, neonatal rats at day 7 was exposed to 2.0% sevoflurane for 6 h to induce neurotoxicity. Pathological analysis showed that sevoflurane anesthesia induced hippocampal injury and reduced the number of neurons. The expression of PHLDA1 was elevated in hippocampus of sevoflurane-treated rats. Secondly, sevoflurane anesthesia-treated neonatal rats were injected with adeno-associated virus serotype (AAV) to mediate knockdown of PHLDA1. Injection with AAV-shPHLDA1 ameliorated sevoflurane-induced hippocampal injury and neurocognitive impairment in rats. Moreover, knockdown of PHLDA1 increased the number of neurons in sevoflurane-treated rats. Silence of PHLDA1 suppressed neuronal apoptosis, and inhibited pyroptosis in sevoflurane-treated rats. Thirdly, PHLDA1 was also elevated in sevoflurane-treated primary neuronal cells. Loss of PHLDA1 also enhanced cell viability and suppressed pyroptosis of sevoflurane-treated primary neuronal cells. Lastly, silence of PHLDA1 reduced protein expression of TRAF6 and p-Rac1 in sevoflurane-treated rats and neuronal cells. Over-expression of TRAF6 attenuated PHLDA1 silence-induced increase of cell viability and decreased pyroptosis in neuronal cells. In conclusion, loss of PHLDA1 protected against sevoflurane-induced pyroptosis in developing rats through inhibition of TRAF6-mediated activation of Rac1.
Collapse
Affiliation(s)
- Lijuan Shu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, China; Department of ICU, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chunfu Du
- Department of Neurosurgery, Ya'an people's Hospital, Sichuan, Ya'an, Sichuan 625000, China.
| |
Collapse
|
16
|
Wang C, Jiang Q, Zhao P. Sevoflurane exposure during the second trimester induces neurotoxicity in offspring rats by hyperactivation of PARP-1. Psychopharmacology (Berl) 2022; 239:3031-3045. [PMID: 35859039 DOI: 10.1007/s00213-022-06188-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/07/2022] [Indexed: 11/27/2022]
Abstract
RATIONALE Fetal exposure to general anesthesia may cause noteworthy neurocognitive impairment, but the mechanisms are unclear. OBJECTIVES Our study designed to explore the potential mechanism of neurotoxicity in offspring rats after sevoflurane exposure to the pregnant rats during the second trimester. METHODS Pregnant rats (G14 day) were administrated with or without 3.5% sevoflurane, 40 mg/kg 3-aminobenzamide (3-AB), inhibitor of poly ADP ribose polymerase 1 (PARP-1), or 10 mg/kg TC-2153, inhibitor of striatal-enriched phosphatase 61 (STEP61). Afterwards, the effects on expression of β-tubulin (TUJ1), neurite outgrowth inhibitor A (Nogo-A), parthanatos-related and STEP61/proline-rich tyrosine kinase 2 (Pyk2) pathway-associated proteins, and reactive oxygen species (ROS) levels were examined by immunofluorescence staining, Western blot, and dihydroethidium (DHE) staining, respectively. Moreover, morphological changes in the hippocampal CA3 region and neuronal cell death were tested by glycine silver staining and TUNEL and immunofluorescence double staining, respectively. Furthermore, spatial learning and memory functions of rats on postnatal 28-33 days (PND 28-33) were evaluated by morris water maze (MWM). RESULTS Mid-pregnancy exposure to sevoflurane led to excessive PARP-1 activation, poly (ADP-ribose) (PAR) polymer accumulation, apoptosis-inducing factor (AIF) nuclear translocation, and Nogo-A accumulation. Besides, sevoflurane significantly inhibited neurite growth and increased cell death in the fetal rat brain. Additionally, sevoflurane activated STEP61/Pyk2 pathway and increased ROS levels. However, 3-AB or TC-2153 significantly alleviated cell death, promoted neurites growth, and improved sevoflurane-induced spatial learning and memory impairment. CONCLUSION This study proposes that sevoflurane exposure during the second trimester incudes neurotoxicity in offspring rats by hyperactivation of PARP-1 via STEP61/Pyk2 pathway.
Collapse
Affiliation(s)
- Cong Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No 36 Sanhao Street, Heping District Liaoning Province, 110004, Shenyang, China
| | - Qian Jiang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No 36 Sanhao Street, Heping District Liaoning Province, 110004, Shenyang, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, No 36 Sanhao Street, Heping District Liaoning Province, 110004, Shenyang, China.
| |
Collapse
|
17
|
Sun M, Xie Z, Zhang J, Leng Y. Mechanistic insight into sevoflurane-associated developmental neurotoxicity. Cell Biol Toxicol 2022; 38:927-943. [PMID: 34766256 PMCID: PMC9750936 DOI: 10.1007/s10565-021-09677-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023]
Abstract
With the development of technology, more infants receive general anesthesia for surgery, other interventions, or clinical examination at an early stage after birth. However, whether general anesthetics can affect the function and structure of the developing infant brain remains an important, complex, and controversial issue. Sevoflurane is the most-used anesthetic in infants, but this drug is potentially neurotoxic. Short or single exposure to sevoflurane has a weak effect on cognitive function, while long or repeated exposure to general anesthetics may cause cognitive dysfunction. This review focuses on the mechanisms by which sevoflurane exposure during development may induce long-lasting undesirable effects on the brain. We review neural cell death, neural cell damage, impaired assembly and plasticity of neural circuits, tau phosphorylation, and neuroendocrine effects as important mechanisms for sevoflurane-induced developmental neurotoxicity. More advanced technologies and methods should be applied to determine the underlying mechanism(s) and guide prevention and treatment of sevoflurane-induced neurotoxicity. 1. We discuss the mechanisms underlying sevoflurane-induced developmental neurotoxicity from five perspectives: neural cell death, neural cell damage, assembly and plasticity of neural circuits, tau phosphorylation, and neuroendocrine effects.
2. Tau phosphorylation, IL-6, and mitochondrial dysfunction could interact with each other to cause a nerve damage loop.
3. miRNAs and lncRNAs are associated with sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Mingyang Sun
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China 730000 ,Department of Anesthesiology and Perioperative Medicine, Center for Clinical Single Cell Biomedicine, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan People’s Republic of China 450003
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, Center for Clinical Single Cell Biomedicine, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan People’s Republic of China 450003
| | - Yufang Leng
- Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu People’s Republic of China 730000
| |
Collapse
|
18
|
Merighi A, Gionchiglia N, Granato A, Lossi L. The Phosphorylated Form of the Histone H2AX (γH2AX) in the Brain from Embryonic Life to Old Age. Molecules 2021; 26:7198. [PMID: 34885784 PMCID: PMC8659122 DOI: 10.3390/molecules26237198] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
The γ phosphorylated form of the histone H2AX (γH2AX) was described more than 40 years ago and it was demonstrated that phosphorylation of H2AX was one of the first cellular responses to DNA damage. Since then, γH2AX has been implicated in diverse cellular functions in normal and pathological cells. In the first part of this review, we will briefly describe the intervention of H2AX in the DNA damage response (DDR) and its role in some pivotal cellular events, such as regulation of cell cycle checkpoints, genomic instability, cell growth, mitosis, embryogenesis, and apoptosis. Then, in the main part of this contribution, we will discuss the involvement of γH2AX in the normal and pathological central nervous system, with particular attention to the differences in the DDR between immature and mature neurons, and to the significance of H2AX phosphorylation in neurogenesis and neuronal cell death. The emerging picture is that H2AX is a pleiotropic molecule with an array of yet not fully understood functions in the brain, from embryonic life to old age.
Collapse
Affiliation(s)
| | | | | | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, I-10095 Grugliasco, Italy; (A.M.); (N.G.); (A.G.)
| |
Collapse
|
19
|
Yang L, He K, Yao S, Zhang Y, Shen J. Sevoflurane inhibits neuroblastoma cell proliferation and invasion and induces apoptosis by miR-144-3p/YAP1 axis. Basic Clin Pharmacol Toxicol 2021; 129:297-307. [PMID: 34192826 DOI: 10.1111/bcpt.13629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sevoflurane (SEV) is a typical volatile anaesthetic and has an antitumour activity in various cancer cells. Here, we were curious whether SEV has tumour-suppressive effects in neuroblastoma (NB). METHODS NB cell lines (K-N-SH and SK-N-AS) were treated with SEV (1%, 2% and 4%). Cell Counting Kit-8 (CCK8) and Transwell assays were conducted to examine cell proliferation and invasion, respectively. The apoptosis was verified by flow cytometry, and the yes-associated protein 1 (YAP1), Bax, Bcl2 and cleaved caspase3 levels were detected by western blotting. Quantitative real-time PCR (qRT-PCR) was conducted to monitor the miR-144-3p level in SEV-treated NB cells. The targeted relationship between miR-144-3p and YAP1 was predicted by bioinformatics and testified by the dual-luciferase reporter assay. RESULTS SEV mitigated NB cell proliferation and invasion and strengthened apoptosis dose-dependently. SEV upregulated miR-144-3p. Moreover, the miR-144-3p inhibitor transfection significantly reduced the tumour-suppressive effect of SEV on NB cells. Furthermore, the dual-luciferase reporter assay confirmed that miR-144-3p targeted YAP1 and overexpressing YAP1 partially weakened the inhibitive effects of miR-144-3p on NB cells. CONCLUSION SEV abated NB cell proliferation and invasion and accelerated apoptosis through the miR-144-3p/YAP1 axis.
Collapse
Affiliation(s)
- Longqiu Yang
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Ke He
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Shudong Yao
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Yiqiang Zhang
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Jun Shen
- Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| |
Collapse
|
20
|
Çolak R, Celik A, Diniz G, Alkan Özdemir S, Yilmaz O, Calkavur S. Evaluation of the Neuroprotective Effect of Pycnogenol in a Hypoxic-Ischemic Brain Injury Model in Newborn Rats. Am J Perinatol 2021; 40:612-618. [PMID: 34044458 DOI: 10.1055/s-0041-1730349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study aimed to evaluate the efficacy of Pycnogenol (PYC) and its antioxidant and antiapoptotic effect in an experimental hypoxic-ischemic (HI) rat model. STUDY DESIGN A total of 24 Wistar albino rats who were on the seventh postnatal day were divided into three groups with developed HI brain injury model under the sevoflurane anesthesia: 40 mg/kg PYC was given to Group A, saline was given to Group B, and the sham group was Group C. Neuronal apoptosis was investigated by terminal deoxynucleotidyl transferase dUTP nick end labeling and immunohistochemically stained manually with primer antibodies of tumor necrosis factor-α and interleukin-1β. RESULTS The neuronal cell injury was statistically lower in the PYC treatment group. CONCLUSION This is the first study that investigates the role of PYC in the HI brain injury model. PYC reduces apoptosis and neuronal injury in the cerebral tissue of the rats. PYC may be a protective agent against hypoxic-ischemic encephalopathy. KEY POINTS · This is the first study that investigates the role of PYC in the HI brain injury model.. · PYC may be a protective agent against hypoxic-ischemic encephalopathy.. · Sevoflurane should not be preferred in rat studies where neuronal apoptosis will be investigated..
Collapse
Affiliation(s)
- Ruya Çolak
- Neonatal Intensive Care Unit, Division of Pediatric, Medical Park Bahcelievler Hospital, Altınbas University Medical Faculty, Istanbul, Turkey
| | - Aslı Celik
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, Izmir, Turkey
| | - Gulden Diniz
- Department of Pathology, Medical Faculty, Izmir Democracy University, Izmir, Turkey
| | - Senem Alkan Özdemir
- Neonatal Intensive Care Unit, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Osman Yilmaz
- Department of Laboratory Animal Science, Faculty of Health Sciences, Dokuz Eylül University, Izmir, Turkey
| | - Sebnem Calkavur
- Neonatal Intensive Care Unit, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| |
Collapse
|