1
|
Volberg C, Toussaint K, Politt K, Gschnell M, Wulf H. [Cancer Treatment and Anaesthesia - What are the Perioperative Considerations?]. Anasthesiol Intensivmed Notfallmed Schmerzther 2025; 60:229-242. [PMID: 40233784 DOI: 10.1055/a-2442-5977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Modern oncological treatment options are significantly extending patient survival. As a result, anaesthetists are increasingly faced with patients who have been pre-treated with immunotherapy or chemotherapy, are currently undergoing a cycle of therapy, or even need to receive chemotherapy intraoperatively. As the anaesthetic agents and perioperative analgesics may interfere with the oncological drugs, it is of interest for the anaesthetist to be aware of the spectrum of side effects and incompatibilities and to adapt the anaesthetic and perioperative treatment regimens accordingly. The aim of this review article is to present the relevant information and provide the clinician with recommendations on where problems may occur and how they can be avoided.
Collapse
|
2
|
Choi H, Hwang W. Anesthetic Approaches and Their Impact on Cancer Recurrence and Metastasis: A Comprehensive Review. Cancers (Basel) 2024; 16:4269. [PMID: 39766169 PMCID: PMC11674873 DOI: 10.3390/cancers16244269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/10/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer recurrence and metastasis remain critical challenges following surgical resection, influenced by complex perioperative mechanisms. This review explores how surgical stress triggers systemic changes, such as neuroendocrine responses, immune suppression, and inflammation, which promote the dissemination of residual cancer cells and circulating tumor cells. Key mechanisms, such as epithelial-mesenchymal transition and angiogenesis, further enhance metastasis, while hypoxia-inducible factors and inflammatory responses create a microenvironment conducive to tumor progression. Anesthetic agents and techniques modulate these mechanisms in distinct ways. Inhaled anesthetics, such as sevoflurane, may suppress immune function by increasing catecholamines and cytokines, thereby promoting cancer progression. In contrast, propofol-based total intravenous anesthesia mitigates stress responses and preserves natural killer cell activity, supporting immune function. Opioids suppress immune surveillance and promote angiogenesis through the activation of the mu-opioid receptor. Opioid-sparing strategies using NSAIDs show potential in preserving immune function and reducing recurrence risk. Regional anesthesia offers benefits by reducing systemic stress and immune suppression, though the clinical outcomes remain inconsistent. Additionally, dexmedetomidine and ketamine exhibit dual effects, both enhancing and inhibiting tumor progression depending on the dosage and context. This review emphasizes the importance of individualized anesthetic strategies to optimize long-term cancer outcomes. While retrospective studies suggest potential benefits of propofol-based total intravenous anesthesia and regional anesthesia, further large-scale trials are essential to establish the definitive role of anesthetic management in cancer recurrence and survival.
Collapse
Affiliation(s)
| | - Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
3
|
Huh J, Hwang W. The Role of Anesthetic Management in Lung Cancer Recurrence and Metastasis: A Comprehensive Review. J Clin Med 2024; 13:6681. [PMID: 39597826 PMCID: PMC11594908 DOI: 10.3390/jcm13226681] [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: 10/14/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Lung cancer remains a leading cause of cancer-related mortality worldwide. Although surgical treatment is a primary approach, residual cancer cells and surgery-induced pathophysiological changes may promote cancer recurrence and metastasis. Anesthetic agents and techniques have recently been shown to potentially impact these processes by modulating surgical stress responses, immune function, inflammatory pathways, and the tumor microenvironment. Anesthetics can influence immune-modulating cytokines, induce pro-inflammatory factors such as HIF-1α, and alter natural-killer cell activity, affecting cancer cell survival and spread. Preclinical studies suggest volatile anesthetics may promote tumor progression by triggering pro-inflammatory signaling, while propofol shows potential antitumor properties through immune-preserving effects and reductions in IL-6 and other inflammatory markers. Additionally, opioids are known to suppress immune responses and stimulate pathways that may support cancer cell proliferation, whereas regional anesthesia may reduce these risks by decreasing the need for systemic opioids and volatile agents. Despite these findings, clinical data remain inconclusive, with studies showing mixed outcomes across patient populations. Current clinical trials, including comparisons of volatile agents with propofol-based total intravenous anesthesia, aim to provide clarity but highlight the need for further investigation. Large-scale, well-designed studies are essential to validate the true impact of anesthetic choice on cancer recurrence and to optimize perioperative strategies that support long-term oncologic outcomes for lung cancer patients.
Collapse
Affiliation(s)
| | - Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
4
|
Loubet F, Robert C, Leclaire C, Theillière C, Saint-Béat C, Lenga Ma Bonda W, Zhai R, Minet-Quinard R, Belville C, Blanchon L, Sapin V, Garnier M, Jabaudon M. Effects of sevoflurane on lung alveolar epithelial wound healing and survival in a sterile in vitro model of acute respiratory distress syndrome. Exp Cell Res 2024; 438:114030. [PMID: 38583855 DOI: 10.1016/j.yexcr.2024.114030] [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: 01/14/2024] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a serious lung condition that often leads to hospitalization in intensive care units and a high mortality rate. Sevoflurane is a volatile anesthetic with growing interest for sedation in ventilated patients with ARDS. It has been shown to have potential lung-protective effects, such as reduced inflammation and lung edema, or improved arterial oxygenation. In this study, we investigated the effects of sevoflurane on lung injury in cultured human carcinoma-derived lung alveolar epithelial (A549) cells. We found that sevoflurane was associated with improved wound healing after exposure to inflammatory cytokines, with preserved cell proliferation but no effect on cell migration properties. Sevoflurane exposure was also associated with enhanced cell viability and active autophagy in A549 cells exposed to cytokines. These findings suggest that sevoflurane may have beneficial effects on lung epithelial injury by promoting alveolar epithelial wound healing and by influencing the survival and proliferation of A549 epithelial cells in vitro. Further research is needed to confirm these findings and to investigate the key cellular mechanisms explaining sevoflurane's potential effects on lung epithelial injury.
Collapse
Affiliation(s)
- Florian Loubet
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Cédric Robert
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Charlotte Leclaire
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Camille Theillière
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Cécile Saint-Béat
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | | | - Ruoyang Zhai
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Régine Minet-Quinard
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France; Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Corinne Belville
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Loic Blanchon
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Vincent Sapin
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France; Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Marc Garnier
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- iGReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France.
| |
Collapse
|
5
|
Tang Y, Tang L, Yao Y, Huang H, Chen B. Effects of anesthesia on long-term survival in cancer surgery: A systematic review and meta-analysis. Heliyon 2024; 10:e24791. [PMID: 38318020 PMCID: PMC10839594 DOI: 10.1016/j.heliyon.2024.e24791] [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: 09/09/2023] [Revised: 12/08/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Backgrounds The association between anesthesia and long-term oncological outcome after cancer surgery remains controversial. This study aimed to investigate the effect of propofol-based anesthesia and inhalation anesthesia on long-term survival in cancer surgery. Methods A comprehensive literature search was performed in PubMed, Medline, Embase, and the Cochrane Library until November 15, 2023. The outcomes included overall survival (OS) and recurrence-free survival (RFS). The hazard ratio (HR) and 95 % confidence interval (CI) were calculated with a random-effects model. Results We included forty-two retrospective cohort studies and two randomized controlled trials (RCTs) with 686,923 patients. Propofol-based anesthesia was associated with improved OS (HR = 0.82, 95 % CI:0.76-0.88, P < 0.00001) and RFS (HR = 0.80, 95 % CI:0.73-0.88, P < 0.00001) than inhalation anesthesia after cancer surgery. However, these positive results were only observed in single-center studies (OS: HR = 0.76, 95 % CI:0.68-0.84, P < 0.00001; RFS: HR = 0.76, 95 % CI:0.66-0.87, P < 0.0001), but not in multicenter studies (OS: HR = 0.98, 95 % CI:0.94-1.03, P = 0.51; RFS: HR = 0.95, 95 % CI:0.87-1.04, P = 0.26). The subgroup analysis revealed that propofol-based anesthesia provided OS and RFS advantages in hepatobiliary cancer (OS: HR = 0.58, 95 % CI:0.40-0.86, P = 0.005; RFS: HR = 0.62, 95 % CI:0.44-0.86, P = 0.005), gynecological cancer (OS: HR = 0.52, 95 % CI:0.33-0.81, P = 0.004; RFS: HR = 0.51, 95 % CI:0.36-0.72, P = 0.0001), and osteosarcoma (OS: HR = 0.30, 95 % CI:0.11-0.81, P = 0.02; RFS: HR = 0.32, 95 % CI:0.14-0.75, P = 0.008) surgeries. Conclusion Propofol-based anesthesia may be associated with improved OS and RFS than inhalation anesthesia in some cancer surgeries. Considering the inherent weaknesses of retrospective designs and the strong publication bias, our findings should be interpreted with caution. Well-designed multicenter RCTs are still urgent to further confirm these findings.
Collapse
Affiliation(s)
- Yaxing Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lele Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuting Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bing Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Fu B, Zhou F, Zhang J, Kong X, Ni B, Bu J, Xu S, He C. Sevoflurane attenuates proliferative and migratory activity of lung cancer cells via mediating the microRNA-100-3p/sterol O-Acyltransferase 1 axis. CHINESE J PHYSIOL 2023; 66:456-465. [PMID: 38149558 DOI: 10.4103/cjop.cjop-d-22-00124] [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] [Indexed: 12/28/2023] Open
Abstract
Recently, evidence has shown that microRNA-100-3p (miR-100-3p) has been revealed as a tumor suppressor in diverse human diseases, while its capability in lung cancer warrants further validation. In this work, we aimed to discuss the impact of sevoflurane on biological functions of lung cancer cells by modulating the miR-100-3p/sterol O-acyltransferase 1 (SOAT1) axis. Lung cancer cell lines (A549 and H460) were treated with various concentrations of sevoflurane. Cell viability, proliferation, migration, and invasion were evaluated using MTT, colony formation, wound healing, and transwell assays. Moreover, miR-100-3p and SOAT1 expressions were evaluated by reverse transcription-quantitative polymerase chain reaction in lung cancer cells. The target interaction between miR-100-3p and SOAT1 was predicted by bioinformatics analysis and verified by the dual-luciferase reporter gene assay. The findings of our work demonstrated that sevoflurane impeded the abilities on viability, proliferation, migration, and invasion of A549 and H460 cells. The expression of miR-100-3p was reduced, and SOAT1 expression was elevated in lung cancer cells. miR-100-3p targeted SOAT1. Besides, sevoflurane could lead to expressed improvement of miR-100-3p or limitation of SOAT1. Downregulation of miR-100-3p or upregulation of SOAT1 restored the suppression of sevoflurane on abilities of viability, proliferation, migration, and invasion in A549 and H460 cells. In the rescue experiment, downregulation of SOAT1 reversed the impacts of downregulation of miR-100-3p on sevoflurane on lung cancer cells. Collectively, our study provides evidence that sevoflurane restrained the proliferation and invasion in lung cancer cells by modulating the miR-100-3p/SOAT1 axis. This article provides a new idea for further study of the pathogenesis of lung cancer.
Collapse
Affiliation(s)
- Bicheng Fu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Fucheng Zhou
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jian Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xianglong Kong
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Boxiong Ni
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jianlong Bu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shidong Xu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Changjun He
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Yun S, Kim K, Shin K, Park H, Lee S, Shin Y, Paing AS, Choi S, Lim C. Effect of Sevoflurane on the Proliferation of A549 Lung Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59030613. [PMID: 36984614 PMCID: PMC10054785 DOI: 10.3390/medicina59030613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Background and Objectives: Sevoflurane has opposing effects on cancer progression, depending on its concentration and the cancer type. This study investigated the effects of sevoflurane on the proliferation of A549 lung cancer cells. Materials and Methods: In vitro, the number of A549 cells exposed to different concentrations of sevoflurane was counted. The size and weight of tumors from a xenograft mouse model exposed to air or sevoflurane were measured in vivo experiments. Additionally, hematoxylin and eosin staining and immunohistochemical detection of Ki-67 in the harvested tumor tissues were performed. Results: A total of 72 culture dishes were used and 24 dishes were assigned to each group: Air group; 2% Sevo group (air + 2% sevoflurane); and 4% Sevo group (air + 4% sevoflurane). The number of A549 cells in the 2% Sevo group was less than that in the Air and 4% Sevo groups (Air: 7.9 ± 0.5; 0.5, 2% Sevo: 6.8 ± 0.4, 4% Sevo: 8.1 ± 0.3; p = 0.000). The tumor size was not significantly different between the two groups (Air: 1.5 ± 0.7, 2% Sevo: 2.4 ± 1.9; p = 0.380). Conclusions: The in vitro data showed that sevoflurane inhibited the proliferation of A549 lung cancer cells in a concentration-specific manner. However, the in vivo data showed no correlation between sevoflurane exposure and A549 cell proliferation. Thus, further research is required to understand fully the effects of sevoflurane on cancer progression and to reconcile differences between the in vitro and in vivo experimental results.
Collapse
Affiliation(s)
- Sangwon Yun
- Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Kyongsik Kim
- Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Keuna Shin
- Research Institute for Medical Sciences, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hanmi Park
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Sunyeul Lee
- Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Yongsup Shin
- Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Aung Soe Paing
- Department of Surgery, 1000 Bedded Naypyitaw General Hospital, Naypyitaw 15011, Myanmar
| | - Songyi Choi
- Department of Pathology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Chaeseong Lim
- Department of Anesthesiology and Pain Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
- Research Institute for Medical Sciences, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| |
Collapse
|
8
|
Zhang C, Pan R, Ma S, Xu S, Wang B. Dezocine inhibits cell proliferation, migration, and invasion by targeting CRABP2 in ovarian cancer. Open Med (Wars) 2022; 17:2052-2061. [PMID: 36568517 PMCID: PMC9755696 DOI: 10.1515/med-2022-0541] [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: 04/20/2022] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 12/23/2022] Open
Abstract
Previous studies have shown that some anesthesia drugs can inhibit tumor growth and metastasis. As a clinical anesthetic drug, dezocine has been reported to play an important role in immune function. However, the effects of dezocine on ovarian cancer cell growth and metastasis are not fully understood. In this study, we found that dezocine dose-dependently inhibited the viability of ES-2 and SKOV3 cells. Dezocine suppressed the migration and invasion abilities of ovarian cancer cells, and promoted apoptosis. Moreover, the Akt/mTOR signaling pathway was also inhibited by dezocine. Furthermore, mechanism study showed that dezocine could significantly inhibit the expression of CRABP2, and CRABP2 overexpression reversed the inhibitory effects of dezocine on ovarian cancer cell proliferation and migration. In conclusion, dezocine has significant anti-tumor effects on the growth and metastatic potential of ovarian cancer cells, and CRABP2 functions as a downstream effector of dezocine.
Collapse
Affiliation(s)
- Chuanfeng Zhang
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ruirui Pan
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Shuangshuang Ma
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Shoucai Xu
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Baosheng Wang
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Jinan 250117, Shandong, China
| |
Collapse
|
9
|
Zeng X, Li J, Yang F, Xia R. The effect of narcotics on ferroptosis-related molecular mechanisms and signalling pathways. Front Pharmacol 2022; 13:1020447. [PMID: 36313359 PMCID: PMC9606818 DOI: 10.3389/fphar.2022.1020447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Ferroptosis is a novel programmed cell death form characterized by iron-mediated reactive oxygen species-induced lipid peroxidation and subsequent cell damage that is distinct from apoptosis, necroptosis, pyroptosis, and autophagy. Most studies on ferroptosis are based on its function and mechanism, but there have been relatively few studies on the effects of drugs, especially anaesthetics, on ferroptosis. Therefore, we summarized the recent literature on the effects of anaesthetics on ferroptosis to understand the underlying mechanism. In particular, we focused on the targets of various anaesthetics in different mechanisms of ferroptosis and the effects of ferroptosis induction or inhibition by narcotics on various diseases. The aims of this review are to provide a relatively reasonable drug regimen for clinicians, to explore potential ferroptosis protection drugs and targets, to reduce perioperative complications and to improve the postoperative performance of patients, especially those who are critically ill.
Collapse
Affiliation(s)
- Xiaoqin Zeng
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jingda Li
- College of Life Sciences, Yangtze University, Jingzhou, Hubei, China
| | - Fuyuan Yang
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| | - Rui Xia
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| |
Collapse
|
10
|
Zhang N, Gao Y, Bian Q, Wang Q, Shi Y, Zhao Z, Yu H. The role of fascin-1 in the pathogenesis, diagnosis and management of respiratory related cancers. Front Oncol 2022; 12:948110. [PMID: 36033434 PMCID: PMC9404296 DOI: 10.3389/fonc.2022.948110] [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: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Human cancer statistics report that respiratory related cancers such as lung, laryngeal, oral and nasopharyngeal cancers account for a large proportion of tumors, and tumor metastasis remains the major reason for patient death. The metastasis of tumor cells requires actin cytoskeleton remodeling, in which fascin-1 plays an important role. Fascin-1 can cross-link F-actin microfilaments into bundles and form finger-like cell protrusions. Some studies have shown that fascin-1 is overexpressed in human tumors and is associated with tumor growth, migration and invasion. The role of fascin-1 in respiratory related cancers is not very clear. The main purpose of this study was to provide an updated literature review on the role of fascin-1 in the pathogenesis, diagnosis and management of respiratory related cancers. These studies suggested that fascin-1 can serve as an emerging biomarker and potential therapeutic target, and has attracted widespread attention.
Collapse
Affiliation(s)
- Naibin Zhang
- Department of biochemistry, Jining Medical University, Jining, China
| | - Yankun Gao
- Department of biochemistry, Jining Medical University, Jining, China
| | - Qiang Bian
- Collaborative Innovation Center, Jining Medical University, Jining, China
- Department of Pathophysiology, Weifang Medical University, Weifang, China
| | - Qianqian Wang
- Department of biochemistry, Jining Medical University, Jining, China
| | - Ying Shi
- Department of biochemistry, Jining Medical University, Jining, China
| | - Zhankui Zhao
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Honglian Yu
- Department of biochemistry, Jining Medical University, Jining, China
- Collaborative Innovation Center, Jining Medical University, Jining, China
- *Correspondence: Honglian Yu,
| |
Collapse
|
11
|
Wang H, Cheng G, Quan L, Qu H, Yang A, Ye J, Feng Y, Li X, Shi X, Pan H. Sevoflurane inhibits the malignant phenotypes of glioma through regulating miR-146b-5p/NFIB axis. Metab Brain Dis 2022; 37:1373-1386. [PMID: 35386035 DOI: 10.1007/s11011-022-00959-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 03/10/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Sevoflurane is a common used inhaled anesthetic that was reported to regulate the progression of multiple cancers. Here, we aimed to investigate the function and regulatory mechanism underlying sevoflurane in glioma cells. METHODS A172 and U251 cells were treated with different concentrations of sevoflurane. Colony formation, EdU satining and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), flow cytometry, and transwell assays were performed to evaluate cell proliferation, apoptosis, migration and invasion, respectively. Circ_VCAN, microRNA-146b-5p (miR-146b-5p) and nuclear factor I B (NFIB) expression levels were assessed by real-time quantitative PCR (RT-qPCR) or western blot. Bioinformatics analysis and dual-luciferase reporter assay were applied to evaluate the correlation between miR-146b-5p and circ_VCAN or NFIB. A xenograft glioma mice model was established to verify the effect of sevoflurane on tumor growth in vivo. RESULTS Sevoflurane (Sev) inhibited proliferation, migration, invasion, and elevated apoptosis of A172 and U251 cells. Sevoflurane treatment inhibited the expression of circ_VCAN and NFIB, but elevated the expression of miR-146b-5p in glioma cells. Overexpression of circ_VCAN alleviated the inhibition effects of sevoflurane on the malignant phenotypes of glioma in vitro and in vivo. Besides, miR-146b-5p is a target of circ_VCAN and negatively regulated NFIB expression. Overexpression of miR-146b-5p partly reversed the effects of circ_VCAN in Sev-treated glioma cells. Furthermore, miR-146b-5p deletion enhanced glioma progression in sevoflurane treated glioma cells by targeting NFIB. Moreover, circ_VCAN could upregulate NFIB expression by sponging miR-146b-5p in Sev-treated glioma cells. CONCLUSION Sevoflurane alleviated proliferation, migration and invasion, but enhanced apoptosis of glioma cells through regulating circ_VCAN/miR-146b-5p/NFIB axis.
Collapse
Affiliation(s)
- Haili Wang
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Guofang Cheng
- Department of Orthopaedic, Sanmenxia Orthopaedic Hospital, Sanmenxia, Henan, China
| | - Lili Quan
- Department of Gynecology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Haibo Qu
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Ailing Yang
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Jiangge Ye
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Yuanbo Feng
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Xiaofang Li
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Xiaoli Shi
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China
| | - Hua Pan
- Department of Anesthesiology, Sanmenxia Central Hospital of Henan University of Science and Technology, Sanmenxia, Henan, China.
| |
Collapse
|
12
|
Miao L, Lv X, Huang C, Li P, Sun Y, Jiang H. Long-term oncological outcomes after oral cancer surgery using propofol-based total intravenous anesthesia versus sevoflurane-based inhalation anesthesia: A retrospective cohort study. PLoS One 2022; 17:e0268473. [PMID: 35559987 PMCID: PMC9106182 DOI: 10.1371/journal.pone.0268473] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/30/2022] [Indexed: 12/24/2022] Open
Abstract
Background Previous studies have shown that the anesthetic technique may influence long-term outcomes after cancer surgery. However, the association between the anesthetic technique and long-term oncological outcomes after oral cancer surgery remains unclear. Therefore, we conducted this study to address this gap. Methods We reviewed the electronic medical records of patients who underwent elective oral cancer surgery between January 2014 and December 2015. The patients were grouped based on the anesthesia maintenance: either propofol or sevoflurane. Propensity score matching in a 1:1 ratio was performed to deal with the potential confounding effects of baseline characteristics. Univariate and multivariate Cox regression analyses were performed to compare hazard ratios (HRs) and identify the risk factors for death and recurrence. Survival analysis was performed using the Kaplan–Meier method, and survival curves were constructed from the date of surgery to death. Results In total, 1347 patients were eligible for analysis, with 343 and 1004 patients in the propofol and sevoflurane groups, respectively. After propensity score matching, 302 patients remained in each group. Kaplan–Meier survival curves demonstrated the 5-year overall and recurrence-free survival rates of 59.3% and 56.0% and 62.7% and 56.5% in the propofol and sevoflurane groups, respectively. There was no significant difference in overall survival or recurrence-free survival between the groups. The multivariate Cox analysis verified this conclusion with HRs of 1.10 and 1.11 for overall survival and recurrence-free survival, respectively, in the sevoflurane group. Older age, advanced tumor-node-metastasis (TNM) stage, and American Society of Anesthesiologists class III were associated with poor overall survival. Patients with advanced TNM stage and poorly differentiated squamous cell carcinoma had a higher recurrence risk than their counterparts. Conclusion The overall and recurrence-free survival rates were similar between propofol-based intravenous anesthesia and sevoflurane volatile anesthesia in patients who underwent oral cancer surgery.
Collapse
Affiliation(s)
- Lingju Miao
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Lv
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Can Huang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Li
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YS); (HJ)
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YS); (HJ)
| |
Collapse
|
13
|
Xu Y, Zhang N, Chen C, Xu X, Luo A, Yan Y, Lu Y, Liu J, Ou X, Tan Y, Liang Y, Chen L, Song X, Liu X. Sevoflurane Induces Ferroptosis of Glioma Cells Through Activating the ATF4-CHAC1 Pathway. Front Oncol 2022; 12:859621. [PMID: 35372041 PMCID: PMC8969566 DOI: 10.3389/fonc.2022.859621] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
ObjectiveTo clarify the function and mechanisms of sevoflurane (Sev) on ferroptosis in glioma cells.MethodsDifferent concentrations of Sev were used to treat glioma cells U87 and U251. Ferroptosis inducer Erastin was used to incubate glioma cells combined with Sev and ATF4 siRNA transfection treatment. CCK-8 assay and colorimetric assay were performed to analyze cell viability and Fe+ concentration, respectively. The releases of reactive oxygen species (ROS) were determined by flow cytometry analysis. Transcriptional sequencing was used to screen the differential genes affected by Sev in U251 cells. The mRNA and protein expression of ferroptosis-associated genes was detected by qRT-PCR and Western blotting.ResultsSev could suppress cell viability, increase ROS levels and Fe+ concentration, downregulate the protein expression levels of GPX4, and upregulate transferrin, ferritin, and Beclin-1 in a dose-dependent manner in U87 and U251 cells. The expression of ferroptosis and mitophagy-related gene activating transcription factor 4 (ATF4) was identified to be enhanced by Sev analyzed by transcriptional sequencing. ChaC glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1), which is involved in ferroptosis, is a downstream gene of ATF4. Inhibition of ATF4 could interrupt the expression of CHAC1 induced by Sev in U87 and U251 cells. Ferroptosis inducer Erastin treatment obviously inhibited the cell viability, elevated the Fe2+ concentration, and promoted ROS generation in U87 and U251 cells. The protein level of ATF4 and CHAC1 was increased in Erastin-treated U87 and U251 cells. Moreover, the interruption of Sev-induced ferroptosis and CHAC1 activating induced by ATF4 suppression could be reversed by Erastin.ConclusionsIn summary, this study suggested that Sev exposure-induced ferroptosis by the ATF4-CHAC1 pathway in glioma cells.
Collapse
Affiliation(s)
- Yingyi Xu
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Na Zhang
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinke Xu
- Department of Neurosurgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ailing Luo
- Department of Hematology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Yan
- Department of Hematology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanhua Lu
- Operating Room, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jianhua Liu
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinxu Ou
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yonghong Tan
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yufeng Liang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatrics, Linzhi People’s Hospital, Linzhi, China
| | - Lihe Chen
- Medical Library, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xingrong Song
- Department of Anaesthesiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Liu
- Department of Hematology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xiaoping Liu,
| |
Collapse
|
14
|
Kang X, Li X, Li Y. Sevoflurane Suppresses the Proliferation, Migration and Invasion of Colorectal Cancer Through Regulating Circ_0000423/miR-525-5p/SGPP1 Network. Cell Mol Bioeng 2022; 15:219-230. [PMID: 35401845 PMCID: PMC8938590 DOI: 10.1007/s12195-021-00717-5] [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: 08/27/2021] [Accepted: 12/20/2021] [Indexed: 01/24/2023] Open
Abstract
Introduction Sevoflurane (SEV) has been shown to inhibit the malignant progression in many cancers, including colorectal cancer (CRC). However, it is not clear whether SEV regulates the progression of CRC by mediating the circular RNA (circRNA) axis. Methods Different concentrations of SEV were used to treat CRC cells. Cell proliferation, migration and invasion were determined by cell counting kit 8 assay, colony formation assay and transwell assay. The expression of circ_0000423, microRNA (miR)-525-5p and sphingosine-1-phosphate phosphatase 1 (SGPP1) mRNA was measured by quantitative real-time PCR. Cell apoptosis was assessed using flow cytometry, and protein expression was measured by western blot analysis. Dual-luciferase reporter assay and RIP assay were performed to confirm the interactions among circ_0000423, miR-525-5p and SGPP1. Animal experiments were performed to explore the effect of SEV and circ_0000423 on CRC tumorigenesis. Results SEV could inhibit CRC cell proliferation, migration and invasion. Circ_0000423 was upregulated in CRC and its expression could be reduced by SEV. Overexpressed circ_0000423 reversed the inhibitory effect of SEV on CRC cell proliferation, migration and invasion and the promotion effect on cell apoptosis. MiR-525-5p could be sponged by circ_0000423, and its overexpression also abolished the regulation of circ_0000423 on the progression of SEV-treated CRC cells. In addition, SGPP1 was confirmed to be a target of miR-525-5p, and its expression was positively regulated by circ_0000423. MiR-525-5p inhibitor promoted CRC cell progression under the treatment of SEV, while these effects could be overturned by SGPP1 silencing. Furthermore, the inhibition effect of SEV on CRC tumorigenesis also could be abolished by overexpressing circ_0000423. Conclusion Our results showed that SEV inhibited CRC progression through the regulation of circ_0000423/miR-525-5p/SGPP1 axis. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00717-5.
Collapse
Affiliation(s)
- Xiaofang Kang
- grid.488137.10000 0001 2267 2324Department of Anesthesiology, The 980 Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, No. 398, Zhongshan West Road, Shijiazhuang City, 050000 Hebei Province China
| | - Xiaocong Li
- grid.488137.10000 0001 2267 2324Department of Anesthesiology, The 980 Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, No. 398, Zhongshan West Road, Shijiazhuang City, 050000 Hebei Province China
| | - Yanli Li
- grid.488137.10000 0001 2267 2324Department of Anesthesiology, The 980 Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, No. 398, Zhongshan West Road, Shijiazhuang City, 050000 Hebei Province China
| |
Collapse
|
15
|
Song Z, Tan J. Effects of Anesthesia and Anesthetic Techniques on Metastasis of Lung Cancers: A Narrative Review. Cancer Manag Res 2022; 14:189-204. [PMID: 35046726 PMCID: PMC8763573 DOI: 10.2147/cmar.s343772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/20/2021] [Indexed: 01/21/2023] Open
Abstract
Purpose Tumor recurrence and metastasis are essential for the mortality and morbidity of cancer. Surgical resection of solid tumors is the conventional treatment approach for malignant tumors. However, even after undergoing radical surgery, certain patients develop local or distant metastasis, which may contribute to treatment failure. Anesthesia and anesthetic techniques are widely used in the perioperative period. Emerging evidence indicates that anesthetics influence tumor recurrence and metastasis. Therefore, the current review summarizes the effects of anesthesia and anesthetic techniques on tumor recurrence and lung metastasis. Methods Relevant literature was retrieved from the following databases: Medline/PubMed, CNKI and Wanfang. A total of 109 articles were selected and analyzed in this research. Results (1) A variety of intravenous anesthetics may affect metastasis or tumor growth, though the evidence is contradictory and inconsistent, and the clinical data are still inconclusive. (2) Volatile anesthetics have proinflammatory effects and may have direct and indirect effects on the survival of cancer cells. (3) Although the relevant clinical data are limited, there is strong evidence in vitro that local anesthetics have a protective effect on cancer recurrence. (4) No mode of anesthesia has been determined to be beneficial to patients with cancer, but clinical studies are currently recommended for anesthesia modality and composite use. Conclusion Available data suggest that anesthesia and anesthetic techniques might play an important role in tumor progression and lung metastasis, the understanding of which will help in designing more effective management of the tumor and attaining fewer side effects.
Collapse
Affiliation(s)
- Zhenghuan Song
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Jing Tan
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
- Correspondence: Jing Tan Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Xuanwu Section, Nanjing, Jiangsu Province, People’s Republic of ChinaTel +86-02583284765 Email
| |
Collapse
|
16
|
Zhu X, Peng C, Peng Z, Chang R, Guo Q. Sevoflurane Inhibits Metastasis in Hepatocellular Carcinoma by Inhibiting MiR-665-Induced Activation of the ERK/MMP Pathway. Cell Transplant 2022; 31:9636897221104447. [PMID: 35699095 PMCID: PMC9201366 DOI: 10.1177/09636897221104447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent evidence has indicated that inhalational anesthetics may affect the growth
and malignant potential of tumor cells and ultimately influence tumor recurrence
after surgery. Sevoflurane, a volatile anesthetic, is used extensively in
hepatectomy. However, the effect of sevoflurane on the growth of hepatocellular
carcinoma (HCC) cells remains unknown. The aim of this study was to explore the
effects of sevoflurane on HCC metastasis and its potential mechanisms in the
human HCC cell lines, HepG2 and SMMC7721. HepG2 and SMMC7721 cells were treated
with 1.7%, 3.4%, and 5.1 % sevoflurane for 6 h. Cell migration was analyzed
using invasion, migration, and scratch assays. Based on previous literature,
several microRNAs (miRNAs) were screened to determine regulatory miRNA targets
of sevoflurane in HepG2 and SMMC7721 cells; miR-665 was detected as a potential
target and overexpressed or inhibited in HepG2 and SMMC7721 cells by a
lentiviral system. The p-ERK/MMP pathway was also measured by western blotting.
Sevoflurane inhibited the migration and invasion of HCC cells in a
dose-dependent manner. It also inhibited miR-665 expression in HCC cells. We
further observed that sevoflurane inhibited HCC metastasis via miR-665.
Sevoflurane-induced downregulation of miRNA-665 led to phosphorylation of ERK and
matrix metalloproteinase (MMP-9) via suppression of SPRED1. These results
demonstrated that sevoflurane may inhibit invasion and migration via the
p-ERK/MMP-9 signaling pathway in HCC cells.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chuchu Peng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyong Peng
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Guangdong, China
| | - Ruimin Chang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Ramirez MF, Cata JP. Anesthesia Techniques and Long-Term Oncological Outcomes. Front Oncol 2021; 11:788918. [PMID: 34956903 PMCID: PMC8692375 DOI: 10.3389/fonc.2021.788918] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
Despite advances in cancer treatments, surgery remains one of the most important therapies for solid tumors. Unfortunately, surgery promotes angiogenesis, shedding of cancer cells into the circulation and suppresses anti-tumor immunity. Together this increases the risk of tumor metastasis, accelerated growth of pre-existing micro-metastasis and cancer recurrence. It was theorized that regional anesthesia could influence long-term outcomes after cancer surgery, however new clinical evidence demonstrates that the anesthesia technique has little influence in oncologic outcomes. Several randomized controlled trials are in progress and may provide a better understanding on how volatile and intravenous hypnotics impact cancer progression. The purpose of this review is to summarize the effect of the anesthesia techniques on the immune system and tumor microenvironment (TME) as well as to summarize the clinical evidence of anesthesia techniques on cancer outcomes.
Collapse
Affiliation(s)
- Maria F Ramirez
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, United States
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, United States
| |
Collapse
|
18
|
Liu D, Wan L, Gong H, Chen S, Kong Y, Xiao B. Sevoflurane promotes the apoptosis of laryngeal squamous cell carcinoma in-vitro and inhibits its malignant progression via miR-26a/FOXO1 axis. Bioengineered 2021; 12:6364-6376. [PMID: 34511023 PMCID: PMC8806578 DOI: 10.1080/21655979.2021.1962684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a laryngeal malignancy with a high mortality rates, and its treatment remains difficult. Sevoflurane is a surgical anesthesia which has anti-tumor effect. This investigation assessed the effects of LSCC cells treatment with Sevoflurane in vitro and in vivo. Hep-2 and Tu177 cells, human LSCC samples and BALB/C nude mice were used for result assessments. Cell viability, proliferation, migration and invasion were assessed via Cell Count Kit-8, wound healing assay and transwell invasion assay respectively. MiR-26a and FOXO1 expressions was examined by qRT-PCR. FOXO1, E-cadherin, N-cadherin and vimentin activities were examined by Western blotting. Moreover, animal experiments were performed to verify our findings in vitro. Lastly, miR-26a and FOXO1 expression levels in clinical samples were analyzed. According to the results, Sevoflurane decreased LSCC cells’ viability and even stimulated their apoptosis in vitro and in vivo. Moreover, it could reduce the migration, invasion and EMT. Mechanistically, sevoflurane could downregulate miR-26a expression and that miR-26a could negatively modulate FOXO1 activity. Thus, sevoflurane could increase FOXO1 activity. In the clinical samples, miR-26a expression was significantly upregulated, but FOXO1 was remarkably down-regulated and miR-26a expression in LSCC was linked with better prognosis. In conclusion, MiR-26a is increased and FOXO1 is reduced in human LSCC, Sevoflurane inhibits proliferation and mediates apoptosis of LSCC cells. Further, MiR-26a binds FOXO1 directly, and FOXO1 expression is down-regulated by Sevoflurane. Finally, Sevoflurane triggers LSCC cells apoptosis in vivo. Sevoflurane use to target miR-26a/FOXO1 may be a novel alternative for LSCC therapy.
Collapse
Affiliation(s)
- Dan Liu
- Department Of Otorhinolaryngology, Huangshi Central Hospital Of Edong Healthcare Group, Hubei Polytechnic University, Huangshi City, Hubei Province, China
| | - Lang Wan
- Department Of Otorhinolaryngology, Huangshi Central Hospital Of Edong Healthcare Group, Hubei Polytechnic University, Huangshi City, Hubei Province, China
| | - Hao Gong
- Department Of Anesthesiology, Huangshi Maternity And Children's Health Hospital, Huangshi City, Hubei Province, China
| | - Shiming Chen
- Department Of Otolaryngology Head And Neck Surgery, Renmin Hospital Of Wuhan University, Wuhan City, Hubei Province, China
| | - Yonggang Kong
- Department Of Otolaryngology Head And Neck Surgery, Renmin Hospital Of Wuhan University, Wuhan City, Hubei Province, China
| | - Bokui Xiao
- Otorhinolaryngology-Head And Neck Surgery Laboratory, Wuhan University School Of Medicine, Wuhan City, Hubei Province, China
| |
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: 7] [Impact Index Per Article: 1.8] [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
|
Zhan X, Lei C, Yang L. Sevoflurane inhibits cell proliferation and migration of glioma by targeting the miR‑27b/VEGF axis. Mol Med Rep 2021; 23:408. [PMID: 33786635 PMCID: PMC8025459 DOI: 10.3892/mmr.2021.12047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
Poor prognosis in patients with glioma is primarily due to rapid tumor growth and cell invasion and migration. In addition, microRNA (miR)‑27b is decreased in metastatic glioma. The present study investigated whether sevoflurane inhibited glioma cell progression by targeting miR‑27b. Cell proliferation was analyzed using a Cell Counting Kit‑8 assay and a wound healing assay was used to detect cell migration. Western blotting and reverse transcription‑quantitative PCR analysis were performed to determine the protein and mRNA expression levels. A dual luciferase assay was used to determine the relationship between vascular epithelial growth factor (VEGF) and miR‑27b. VEGF was identified to be a direct target of miR‑27b. Moreover, sevoflurane treatment increased the expression of miR‑27b and decreased the expression of VEGF in U251 and U87 cells. Compared with the control group, sevoflurane inhibited the proliferation and migration of U251 and U87 cells, as well as the expression of matrix metalloproteinase (MMP)‑2 and MMP‑9, which were subsequently abolished by pre‑treatment with an miR‑27b inhibitor. The present results indicated the potential use of sevoflurane by anesthesiologists for the surgical resection of glioma, which may improve patient outcomes in the clinical setting.
Collapse
Affiliation(s)
- Xi Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Changcheng Lei
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Linzhu Yang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
21
|
Gao C, Xu YJ, Qi L, Bao YF, Zhang L, Zheng L. CircRNA VIM silence synergizes with sevoflurane to inhibit immune escape and multiple oncogenic activities of esophageal cancer by simultaneously regulating miR-124/PD-L1 axis. Cell Biol Toxicol 2021; 38:825-845. [PMID: 34018092 DOI: 10.1007/s10565-021-09613-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/27/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Circular RNA of vimentin (circ-VIM) is a predictor for poor prognosis of acute myeloid leukemia, but we had little information on its function in esophageal cancer (EC). Here we examined the effects of circ-VIM together with sevoflurane on immune escape and multiple oncogenic activities of EC. METHODS Bioinformatic tools, luciferase assay, and RNA immunoprecipitation were used to examine regulations between circ-VIM, miR-124-3p (miR-124), and PD-L1. CCK-8, wound healing, and Transwell assays were used to measure cell proliferation, migration, and invasion, respectively. The impacts of EC cells on cytotoxicity, proliferation, and apoptosis of CD8+ T cells were examined using LDH assay, CFSE staining, and Annexin V/PI staining, respectively. The in vivo tumorigenesis and lung metastases were assessed using xenograft model and tail vein injection of EC cells. RESULTS Significant upregulation of circ-VIM and PD-L1 and downregulation of miR-124 were detected in EC tissues or cells. Circ-VIM sponged miR-124 and released its suppression on the downstream target PD-L1. Sevoflurane, independent of circ-VIM, also upregulated miR-124 to lower PD-L1 expression. By modulating miR-124/PD-L1 axis, silencing circ-VIM and applying sevoflurane both inhibited immune escape and multiple oncogenic activities of EC in vitro, and suppressed xenograft growth and lung metastases in vivo. The inactivation of Ras/ERK signaling pathway was involved in suppression of malignant phenotypes by silencing circ-VIM and sevoflurane treatment. CONCLUSIONS Silencing circ-VIM and applying sevoflurane, by separately regulating miR-124/PD-L1 axis, presented synergistic effects in inhibiting immune escape and multiple malignant phenotypes of EC cells.
Collapse
Affiliation(s)
- Cao Gao
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Yan-Jie Xu
- Departments of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Lei Qi
- Department of Gastroenterology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Ya-Fei Bao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Liang Zheng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
22
|
Ishikawa M, Iwasaki M, Zhao H, Saito J, Hu C, Sun Q, Sakamoto A, Ma D. Inhalational Anesthetics Inhibit Neuroglioma Cell Proliferation and Migration via miR-138, -210 and -335. Int J Mol Sci 2021; 22:ijms22094355. [PMID: 33919449 PMCID: PMC8122527 DOI: 10.3390/ijms22094355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023] Open
Abstract
Inhalational anesthetics was previously reported to suppress glioma cell malignancy but underlying mechanisms remain unclear. The present study aims to investigate the effects of sevoflurane and desflurane on glioma cell malignancy changes via microRNA (miRNA) modulation. The cultured H4 cells were exposed to 3.6% sevoflurane or 10.3% desflurane for 2 h. The miR-138, -210 and -335 expression were determined with qRT-PCR. Cell proliferation and migration were assessed with wound healing assay, Ki67 staining and cell count kit 8 (CCK8) assay with/without miR-138/-210/-335 inhibitor transfections. The miRNA downstream proteins, hypoxia inducible factor-1α (HIF-1α) and matrix metalloproteinase 9 (MMP9), were also determined with immunofluorescent staining. Sevoflurane and desflurane exposure to glioma cells inhibited their proliferation and migration. Sevoflurane exposure increased miR-210 expression whereas desflurane exposure upregulated both miR-138 and miR-335 expressions. The administration of inhibitor of miR-138, -210 or -335 inhibited the suppressing effects of sevoflurane or desflurane on cell proliferation and migration, in line with the HIF-1α and MMP9 expression changes. These data indicated that inhalational anesthetics, sevoflurane and desflurane, inhibited glioma cell malignancy via miRNAs upregulation and their downstream effectors, HIF-1α and MMP9, downregulation. The implication of the current study warrants further study.
Collapse
Affiliation(s)
- Masashi Ishikawa
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Correspondence: (M.I.); (D.M.)
| | - Masae Iwasaki
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Department of Anesthesiology, Graduate School of Medicine, Hirosaki University, Hirosaki, Aomori 036-8562, Japan
| | - Cong Hu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
| | - Atsuhiro Sakamoto
- Department of Anesthesiology and Pain medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan; (M.I.); (A.S.)
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK; (H.Z.); (J.S.); (C.H.); (Q.S.)
- Correspondence: (M.I.); (D.M.)
| |
Collapse
|
23
|
Yong F, Wang H, Li C, Jia H. Sevoflurane represses the migration and invasion of gastric cancer cells by regulating forkhead box protein 3. J Int Med Res 2021; 49:3000605211005936. [PMID: 33906525 PMCID: PMC8108091 DOI: 10.1177/03000605211005936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Previous studies suggested that sevoflurane exerts anti-proliferative, anti-migratory, and anti-invasive effects on cancer cells. To determine the role of sevoflurane on gastric cancer (GC) progression, we evaluated its effects on the proliferation, migration, and invasion of SGC7901, AGS, and MGC803 GC cells. METHODS GC cells were exposed to different concentrations of sevoflurane (1.7, 3.4, or 5.1% v/v). Cell viability, migration, and invasion were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Transwell assays. Immunohistochemical staining and immunoblotting were performed to analyze forkhead box protein 3 (FOXP3) protein expression in tissue specimens and cell lines, respectively. RESULTS FOXP3 was downregulated in human GC specimens and cell lines. Functionally, FOXP3 overexpression significantly inhibited the proliferation, migration, and invasion of GC cells and accelerated their apoptosis. Moreover, sevoflurane significantly blocked GC cell migration and invasion compared with the findings in the control group. However, FOXP3 silencing neutralized sevoflurane-induced apoptosis and the inhibition of GC cell migration and invasion. Sevoflurane-induced apoptosis and the suppression of migration and invasion might be associated with FOXP3 overactivation in GC cells. CONCLUSIONS Sevoflurane activated FOXP3 and prevented GC progression via inhibiting cell migration and invasion in vitro.
Collapse
Affiliation(s)
- Fangfang Yong
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Hemei Wang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Chao Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Huiqun Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
24
|
Ishikawa M, Iwasaki M, Sakamoto A, Ma D. Anesthetics may modulate cancer surgical outcome: a possible role of miRNAs regulation. BMC Anesthesiol 2021; 21:71. [PMID: 33750303 PMCID: PMC7941705 DOI: 10.1186/s12871-021-01294-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Background microRNAs (miRNAs) are single-stranded and noncoding RNA molecules that control post-transcriptional gene regulation. miRNAs can be tumor suppressors or oncogenes through various mechanism including cancer cell biology, cell-to-cell communication, and anti-cancer immunity. Main Body Anesthetics can affect cell biology through miRNA-mediated regulation of messenger RNA (mRNA). Indeed, sevoflurane was reported to upregulate miR-203 and suppresses breast cancer cell proliferation. Propofol reduces matrix metalloproteinase expression through its impact on miRNAs, leading to anti-cancer microenvironmental changes. Propofol also modifies miRNA expression profile in circulating extracellular vesicles with their subsequent anti-cancer effects via modulating cell-to-cell communication. Conclusion Inhalational and intravenous anesthetics can alter cancer cell biology through various cellular signaling pathways induced by miRNAs’ modification. However, this area of research is insufficient and further study is needed to figure out optimal anesthesia regimens for cancer patients.
Collapse
Affiliation(s)
- Masashi Ishikawa
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo, Tokyo, 113-8603, Japan. .,Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, 369 Fulham Rd, London, SW10 9NH, UK.
| | - Masae Iwasaki
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo, Tokyo, 113-8603, Japan.,Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, 369 Fulham Rd, London, SW10 9NH, UK
| | - Atsuhiro Sakamoto
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo, Tokyo, 113-8603, Japan
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, 369 Fulham Rd, London, SW10 9NH, UK
| |
Collapse
|
25
|
Chang CY, Wu MY, Chien YJ, Su IM, Wang SC, Kao MC. Anesthesia and Long-term Oncological Outcomes: A Systematic Review and Meta-analysis. Anesth Analg 2021; 132:623-634. [PMID: 33105278 DOI: 10.1213/ane.0000000000005237] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Whether propofol elicits a survival benefit over volatile anesthetics during cancer surgery remains inconclusive. The primary aim of this systematic review and meta-analysis is to compare the effects of propofol-based total intravenous anesthesia (TIVA) with any volatile anesthesia on long-term oncological outcomes. The secondary aim is to compare propofol-based TIVA with specific volatile agents on long-term oncological outcomes. METHODS We searched PubMed, Embase, Scopus, Web of Science, and Cochrane Library from inception through March 3, 2020. Randomized control trials and observational studies that compared the effects of propofol-based TIVA and volatile anesthesia on long-term oncological outcomes, which also reported hazard ratios (HR) as effect estimates, were considered eligible for inclusion. Using the inverse variance method with a random-effects model, HR and 95% confidence intervals (CI) were calculated. Trial sequential analysis was incorporated to test if the results were subject to a type I or type II error. RESULTS Nineteen retrospective observational studies were included. Patients who received propofol-based TIVA during cancer surgery were associated with significantly better overall survival than those who received volatile anesthesia (HR = 0.79, 95% CI, 0.66-0.94, P = .008, I2 = 82%). In contrast, no statistically significant difference was observed in recurrence-free survival between patients who received propofol-based TIVA and volatile anesthesia during cancer surgery (HR = 0.81, 95% CI, 0.61-1.07, P = .137, I2 = 85%). In the subgroup analysis by different volatile anesthetics, patients who received propofol-based TIVA were associated with better overall survival than those who received desflurane (HR = 0.54, 95% CI, 0.36-0.80, P = .003, I2 = 80%). In contrast, there was no statistically significant difference in overall survival between patients who received propofol-based TIVA and those who received sevoflurane (HR = 0.92, 95% CI, 0.74-1.14, P = .439, I2 = 70%). In the trial sequential analysis of overall survival, the cumulative Z curve reached the required heterogeneity-adjusted information size and crossed the traditional significance boundary. In contrast, in the trial sequential analysis of recurrence-free survival, the cumulative Z curve did not cross the traditional significance boundary. However, the required heterogeneity-adjusted information size has not yet been reached. CONCLUSIONS Propofol-based TIVA is generally associated with better overall survival than volatile anesthesia during cancer surgery. Further large-scaled, high-quality randomized control trials are warranted to confirm our findings.
Collapse
Affiliation(s)
- Chun-Yu Chang
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Meng-Yu Wu
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Emergency Medicine
| | - Yung-Jiun Chien
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Physical Medicine and Rehabilitation, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - I-Min Su
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Anesthesiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shih-Ching Wang
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ming-Chang Kao
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
26
|
Budi HS, Izadi S, Timoshin A, Asl SH, Beyzai B, Ghaderpour A, Alian F, Eshaghi FS, Mousavi SM, Rafiee B, Nikkhoo A, Ahmadi A, Hassannia H, Ahmadi M, Sojoodi M, Jadidi-Niaragh F. Blockade of HIF-1α and STAT3 by hyaluronate-conjugated TAT-chitosan-SPION nanoparticles loaded with siRNA molecules prevents tumor growth. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102373. [PMID: 33667724 DOI: 10.1016/j.nano.2021.102373] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/15/2020] [Accepted: 02/07/2021] [Indexed: 12/15/2022]
Abstract
HIF-1α and STAT3 are two of the critical factors in the growth, proliferation, and metastasis of cancer cells and play a crucial role in inhibiting anti-cancer immune responses. Therefore, we used superparamagnetic iron oxide (SPION) nanoparticles (NPs) coated with thiolated chitosan (ChT) and trimethyl chitosan (TMC) and functionalized with hyaluronate (H) and TAT peptide for delivery of siRNA molecules against STAT3 and HIF-1α to cancer cells both in vivo and in vitro. The results indicated that tumor cell transfection with siRNA-encapsulated NPs robustly inhibited proliferation and migration and induced apoptosis in tumor cells. Furthermore, simultaneous silencing of HIF-1α and STAT3 significantly repressed cancer development in two different tumor types (4T1 breast cancer and CT26 colon cancer) which were associated with upregulation of cytotoxic T lymphocytes and IFN-γ secretion. The findings suggest inhibiting the HIF-1α/STAT3 axis by SPION-TMC-ChT-TAT-H NPs as an effective way to treat cancer.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Anton Timoshin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of propaedeutics of dental diseases, Moscow, Russia
| | | | - Behzad Beyzai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaderpour
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Farzaneh Sadat Eshaghi
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Behnam Rafiee
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Afshin Nikkhoo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Ahmadi
- iepartment of Chemical and Materials Engineering, The University of Alabama in Huntsville, AL, USA
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozhdeh Sojoodi
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Qi Y, Guo L, Liu Y, Zhao T, Liu X, Zhang Y. Sevoflurane Limits Glioma Progression by Regulating Cell Proliferation, Apoptosis, Migration, and Invasion via miR-218-5p/DEK/β-Catenin Axis in Glioma. Cancer Manag Res 2021; 13:2057-2069. [PMID: 33664593 PMCID: PMC7924128 DOI: 10.2147/cmar.s265356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/09/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose Sevoflurane (SEV) is a frequently used volatile anesthetic in cancer surgery. Sevoflurane treatment has been shown to suppress the migration and invasion of several human cancer cells. However, the effect of sevoflurane on glioma remains largely unclear. Methods Glioma cell lines (U251 and U343) were treated by various concentrations of sevoflurane. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), flow cytometry assay, and transwell assay were performed to detect the cell viability, apoptosis, migration and invasion. Western blot assay was employed to detect the protein levels of β-catenin, c-Myc, CyclinD1, β-catenin, N-cadherin, vimentin, and DEK. Moreover, quantitative real-time polymerase chain reaction (qRT-PCR) was used to examine the expression level of miR-218-5p. The target interaction between miR-218-5p and DEK was predicted through bioinformatics analysis and verified by dual-luciferase reporter assay system. Results We found that sevoflurane aberrantly inhibited the abilities on viability, migration, invasion, EMT and β-catenin signaling and promoted cell apoptosis in U251 and U343 cells in a dose-dependent manner. MiR-218-5p strikingly suppressed the abilities of proliferation, migration, invasion rather than apoptosis and activation of β-catenin signaling. Sevoflurane could facilitate the miR-218-5p expression, and its suppressing effects on glioma cells were reversed by pre-treatment with miR-218-5p inhibitors or pcDNA3.1/DEK in vitro and in vivo. Silencing of miR-218-5p reverted sh-DEK and sevoflurane-induced repression on proliferation, migration, invasion, and β-catenin signaling, and promotion on apoptosis in the glioma cells. Conclusion Our data showed that sevoflurane inhibited the proliferation, migration, invasion, and enhanced the apoptosis in glioma cells through regulating miR-218-5p/DEK/β-catenin axis.
Collapse
Affiliation(s)
- Yingying Qi
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Lina Guo
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Yanchao Liu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Tonghang Zhao
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Xianwen Liu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| | - Yang Zhang
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, Shandong, People's Republic of China
| |
Collapse
|
28
|
Yu Q, Dai H, Jiang Y, Zha Y, Zhang J. Sevoflurane alleviates oxygen-glucose deprivation/reoxygenation-induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway. Exp Ther Med 2021; 21:376. [PMID: 33732349 PMCID: PMC7903476 DOI: 10.3892/etm.2021.9807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 07/07/2020] [Indexed: 12/30/2022] Open
Abstract
Sevoflurane (Sev), a volatile anesthetic, has been reported to exhibit beneficial effects on different ischemia/reperfusion (I/R)-injured organs. However, the neuroprotective effect of Sev on cerebral I/R injury is poorly understood. In the present study, the effects of Sev on HT22 cells exposed to oxygen-glucose deprivation/reperfusion (OGD/R) injury are investigated. The present study demonstrated that OGD/R suppressed the cell viability and increased lactate dehydrogenase (LDH) release from the cells, and these effects were attenuated by Sev treatment. The results also demonstrated that Sev alleviated OGD/R-induced cell apoptosis via flow cytometry and caspase-3 activity determination. Biochemical analysis results revealed that Sev significantly protected against OGD/R-induced oxidative stress by reducing ROS generation and improving antioxidant defense markers. Western blot analysis demonstrated that Sev reactivated the PI3K/AKT/glycogen synthase kinase-3β (GSK3β) signaling pathway, which was inhibited by OGD/R. In addition, wortmannin, a selective PI3K inhibitor was used to investigate the underlying pathways. Notably, the neuroprotective effect of Sev on apoptosis and reactive oxygen species production was found to be suppressed by wortmannin. Collectively, these results demonstrated that Sev may protect neuronal cells against OGD/R-induced injury through the activation of the PI3K/AKT/GSK3β signaling pathway. The findings from the present study provide a novel insight into understanding the neuroprotective effect of Sev on cerebral I/R injury.
Collapse
Affiliation(s)
- Qiong Yu
- Department of Anesthesia, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Haofei Dai
- Department of Nursing, Huashan Hospital-North, Fudan University, Shanghai 201907, P.R. China
| | - Yinan Jiang
- Department of Anesthesia, Huashan Hospital-North, Fudan University, Shanghai 201907, P.R. China
| | - Yifeng Zha
- Department of Anesthesia, Huashan Hospital-North, Fudan University, Shanghai 201907, P.R. China
| | - Jie Zhang
- Department of Anesthesia, Huashan Hospital-North, Fudan University, Shanghai 201907, P.R. China
| |
Collapse
|
29
|
Jeon S, Kim HK, Kwon JY, Baek SH, Ri HS, Choi HJ, Cho HR, Lee YS, Kim JY, Kim J, Bae J, Lee HJ. Role of Sevoflurane on Natural Killer Group 2, Member D-Mediated Immune Response in Non-Small-Cell Lung Cancer: An In Vitro Study. Med Sci Monit 2020; 26:e926395. [PMID: 33139690 PMCID: PMC7648410 DOI: 10.12659/msm.926395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background The purpose of this study was to investigate the effects of sevoflurane on cancer immunosurveillance and metastasis in non-small-cell lung cancer (NSCLC). Material/Methods NCI-H23 cells, a human NSCLC cell line, were incubated with or without sevoflurane at the concentrations of 0, 12.5, 25, 50, 100, and 200 μM for 6 h. Cell viability, the expression of natural killer group 2, member D ligands (NKG2D ligands: UL16-binding proteins 1–3 [ULBP1–3] and major histocompatibility complex class I chain-related molecules A/B [MICA/B]), the expression of matrix metalloproteinases (MMPs), NK cell-mediated cytotoxicity, and cancer cell migration were measured. Results At 12.5, 25, 50, and 100 μM, sevoflurane increased the expression of NKG2D ligands (ULBP2–3 and MICA, ULBP1–3, ULBP1–3, and ULBP1, respectively). Sevoflurane decreased the expression of NKG2D ligands at 200 μM (MICA/B). NK cell-mediated lysis of NCI-H23 cells at 200 μM sevoflurane was significantly reduced compared with the control (P=0.025; target cell: effect cell=1: 10). Sevoflurane increased the expression of MMP-1, -2, and -9 and increased cell migration in NCI-H23 cells at 50, 100, and 200 μM (P=0.001, 0.035, and 0.039, respectively, compared with the control after 18 h of wound formation). Conclusions Sevoflurane could suppress NKG2D-mediated NK cell cytotoxicity and increased expression of MMPs and migration in NCI-H23 cells. Further research is needed to determine the effects of sevoflurane on cancer immunosurveillance and metastasis in NSCLC.
Collapse
Affiliation(s)
- Soeun Jeon
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Pusan National University, School of Medicine, Busan, South Korea
| | - Hae-Kyu Kim
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea
| | - Jae-Young Kwon
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea
| | - Seung-Hoon Baek
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea
| | - Hyun-Su Ri
- Department of Anesthesia and Pain Medicine, Pusan National University Yangsan Hospital, Pusan National University, School of Medicine, Yangsan, South Korea
| | - Ho Jung Choi
- Department of Biochemistry, Pusan National University, School of Medicine, Yangsan, South Korea.,PNU BK21 Plus Biomedical Science Education Center, Pusan National University, School of Medicine, Yangsan, South Korea
| | - Hae-Ryung Cho
- Department of Biochemistry, Pusan National University, School of Medicine, Yangsan, South Korea.,PNU BK21 Plus Biomedical Science Education Center, Pusan National University, School of Medicine, Yangsan, South Korea
| | - Young Shin Lee
- Department of Biochemistry, Pusan National University, School of Medicine, Yangsan, South Korea.,PNU BK21 Plus Biomedical Science Education Center, Pusan National University, School of Medicine, Yangsan, South Korea
| | - Joo-Young Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jinsil Kim
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea
| | - Jaeho Bae
- Department of Biochemistry, Pusan National University, School of Medicine, Yangsan, South Korea.,PNU BK21 Plus Biomedical Science Education Center, Pusan National University, School of Medicine, Yangsan, South Korea
| | - Hyeon-Jeong Lee
- Department of Anesthesia and Pain Medicine, Pusan National University, School of Medicine, Busan, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Pusan National University, School of Medicine, Busan, South Korea
| |
Collapse
|
30
|
He J, Zhao H, Liu X, Wang D, Wang Y, Ai Y, Yang J. Sevoflurane suppresses cell viability and invasion and promotes cell apoptosis in colon cancer by modulating exosome‑mediated circ‑HMGCS1 via the miR‑34a‑5p/SGPP1 axis. Oncol Rep 2020; 44:2429-2442. [PMID: 33125091 PMCID: PMC7610314 DOI: 10.3892/or.2020.7783] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/16/2020] [Indexed: 12/24/2022] Open
Abstract
As a novel halogenated hydroxyl ether-inhaled general anesthetic, sevoflurane has been reported to affect the progression of diverse human cancers. In the present study, we aimed to explore the functions and underlying mechanisms of sevoflurane in colon cancer. MTT assay, flow cytometric analysis and Transwell assay were conducted to evaluate cell viability, apoptosis and invasion, respectively. Western blot analysis was performed to determine the protein level of sphingosine-1-phosphate phosphatase 1 (SGPP1). The morphology and size of exosomes were analyzed by TEM and NTA. The levels of circular RNA 3-hydroxy-3-methylglutaryl-CoA synthase 1 (circ-HMGCS1), microRNA (miR)-34a-5p and SGPP1 mRNA were examined by RT-qPCR. Dual-luciferase reporter and RNA RIP assays were utilized to explore the interaction between miR-34a-5p and circ-HMGCS1 or SGPP1. A murine xenograft model was established to investigate the effect of circ-HMGCS1 in vivo. As a result, it was determined that sevoflurane suppressed cell viability and invasion and induced apoptosis in colon cancer in a dose-dependent way. Exosomal circ-HMGCS1 was increased in the serums and cells of colon cancer patients. Circ-HMGCS1 was downregulated by sevoflurane treatment in colon cancer cells and circ-HMGCS1 overexpression could restore the effect of sevoflurane on colon cancer cell development. miR-34a-5p was a target of circ-HMGCS1 and miR-34a-5p inhibition reversed the effect of circ-HMGCS1 silencing on colon cancer cell progression. Moreover, circ-HMGCS1 knockdown suppressed SGPP1 expression via sponging miR-34a-5p. Knockdown of circ-HMGCS1 blocked tumor growth in vivo. In conclusion, sevoflurane inhibited colon cancer progression by modulating the exosome-transmitted circ-HMGCS1/miR-34a-5p/SGPP1 axis.
Collapse
Affiliation(s)
- Juan He
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Huaping Zhao
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Xing Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Dongmei Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yong Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yanqiu Ai
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Jianjun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
31
|
Sevoflurane downregulates insulin-like growth factor-1 to inhibit cell proliferation, invasion and trigger apoptosis in glioma through the PI3K/AKT signaling pathway. Anticancer Drugs 2020; 30:e0744. [PMID: 31305291 DOI: 10.1097/cad.0000000000000744] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sevoflurane is a new type of inhalation anesthetic used widely in the clinic. It has the characteristics of rapid induction, rapid recovery, and less irritative to the airway. Studies have shown that sevoflurane can affect the invasion and migration of a variety of malignant tumors. However, its effects on human glioma cells and related mechanisms are not clear. Cultured U251 and U87 cells were pretreated with sevoflurane. The effect of sevoflurane on proliferation was evaluated by MTT, and cell migration assay, cell apoptosis, and invasion ability were evaluated by wound-healing assay, cell apoptosis, and Transwell assays. Insulin-like growth factor-1 (IGF-1) and PI3K/AKT signaling pathway gene expression in sevoflurane-treated cell lines was measured by western blotting analysis, respectively. 5% sevoflurane significantly inhibited proliferation ability in both U251 and U87 cells. Sevoflurane inhibited glioma cells invasion and migration, and promoted apoptosis. Sevoflurane inhibited IGF-1 and promoted the expression of apoptosis-related proteins in glioma cells. In addition, sevoflurane inhibited the PI3K/AKT signaling pathway in glioma cells. This study clarifies that sevoflurane inhibits proliferation, invasion, and migration, and promotes apoptosis in glioma cells. These effects are regulated by IGF-1, an upstream gene of the PI3K/AKT signaling pathway. These findings may be significant for the selection of anesthetic agents in glioma surgery to improve the prognosis of patients.
Collapse
|
32
|
Codelivery of HIF-1α siRNA and Dinaciclib by Carboxylated Graphene Oxide-Trimethyl Chitosan-Hyaluronate Nanoparticles Significantly Suppresses Cancer Cell Progression. Pharm Res 2020; 37:196. [PMID: 32944844 DOI: 10.1007/s11095-020-02892-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/24/2020] [Indexed: 01/07/2023]
Abstract
PURPOSE Hypoxia-inducible factor (HIF) is one of the critical components of the tumor microenvironment that is involved in tumor development. HIF-1α functionally and physically interacts with CDK1, 2, and 5 and stimulates the cell cycle progression and Cyclin-Dependent Kinase (CDK) expression. Therefore, hypoxic tumor microenvironment and CDK overexpression lead to increased cell cycle progression and tumor expansion. Therefore, we decided to suppress cancer cell expansion by blocking HIF-1α and CDK molecules. METHODS In the present study, we used the carboxylated graphene oxide (CGO) conjugated with trimethyl chitosan (TMC) and hyaluronate (HA) nanoparticles (NPs) loaded with HIF-1α-siRNA and Dinaciclib, the CDK inhibitor, for silencing HIF-1α and blockade of CDKs in CD44-expressing cancer cells and evaluated the impact of combination therapy on proliferation, metastasis, apoptosis, and tumor growth. RESULTS The results indicated that the manufactured NPs had conceivable physicochemical properties, high cellular uptake, and low toxicity. Moreover, combination therapy of cancer cells using CGO-TMC-HA NPs loaded with HIF-1α siRNA and Dinaciclib (SCH 727965) significantly suppressed the CDKs/HIF-1α and consequently, decreased the proliferation, migration, angiogenesis, and colony formation in tumor cells. CONCLUSIONS These results indicate the ability of CGO-TMC-HA NPs for dual drug/gene delivery in cancer treatment. Furthermore, the simultaneous inhibition of CDKs/HIF-1α can be considered as a novel anti-cancer treatment strategy; however, further research is needed to confirm this treatment in vivo. Graphical Abstract The suppression of HIF-1α and CDKs inhibits cancer growth. HIF-1α is overexpressed by the cells present in the tumor microenvironment. The hypoxic environment elevates mitochondrial ROS production and increases p38 MAP kinase, JAK/STAT, ERK, JNK, and Akt/PI3K signaling, resulting in cyclin accumulation and aberrant cell cycle progression. Furthermore, the overexpression of HIF-1α/CDK results in increased expression of genes such as BCL2, Bcl-xl, Ki-67, TGFβ, VEGF, FGF, MMP2, MMP9, and, HIF-1α and consequently raise the survival, proliferation, angiogenesis, metastasis, and invasion of tumor cells. In conclusion, HIF-1α-siRNA/Dinaciclib-loaded CGO-TMC-HA NPs can inhibit the tumor expansion by blockage of CDKs and HIF-1α (JAK: Janus kinase, STAT: Signal transducer and activator of transcription, MAPK: mitogen-activated protein kinase, ERK: extracellular signal-regulated kinase, JNK: c-Jun N-terminal kinase, PI3K: phosphatidylinositol 3-kinase).
Collapse
|
33
|
Zhao H, Xing F, Yuan J, Li Z, Zhang W. Sevoflurane inhibits migration and invasion of glioma cells via regulating miR-34a-5p/MMP-2 axis. Life Sci 2020; 256:117897. [PMID: 32502543 DOI: 10.1016/j.lfs.2020.117897] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/26/2020] [Accepted: 05/31/2020] [Indexed: 11/19/2022]
Abstract
Glioma is the most common brain malignancy and surgical resection is the primary option for patient with glioma. Anesthetics could be used to inhibit cancer dissemination and metastasis during surgery. This study aims to assess the function of volatile anesthetic sevoflurane in glioma migration and invasion and explore the potential mechanism. Twenty-five patients with glioma were recruited in this study. LN229 and U251 cells were used in vitro experiments. Cell viability was analyzed by MTT analysis. Cell migration and invasion were examined via transwell analysis. microRNA-34a-5p (miR-34a-5p) and matrix metalloproteinase-2 (MMP-2) levels were measured via quantitative real-time polymerase chain reaction. The relationship of miR-34a-5p and MMP-2 was tested via bioinformatics analysis, luciferase reporter analysis, RNA immunoprecipitation and RNA pull-down. Sevoflurane decreased glioma cell migration and invasion. In glioma cells, sevoflurane up-regulated miR-34a-5p abundance and down-regulated MMP-2 level. Overexpression of miR-34a-5p contributed to sevoflurane-caused suppression of migration and invasion, while its knockdown played an opposite effect. MMP-2 was targeted via miR-34a-5p and MMP-2 silence reversed the influence of miR-34a-5p knockdown under sevoflurane. Sevoflurane exposure represses cell migration and invasion, which might be related to inhibition of MMP-2 by up-regulating miR-34a-5p. This study provides a novel mechanism for understanding the pharmacological effects of sevoflurane on glioma.
Collapse
Affiliation(s)
- Huaping Zhao
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Fei Xing
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jingjing Yuan
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhisong Li
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wei Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
34
|
Sevoflurane Enhances Proliferation, Metastatic Potential of Cervical Cancer Cells via the Histone Deacetylase 6 Modulation In Vitro. Anesthesiology 2020; 132:1469-1481. [DOI: 10.1097/aln.0000000000003129] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract
Background
Sevoflurane is commonly used for cervical cancer surgery, but its effect on cervical cancer cell biology remains unclear. This mechanistic study explores how sevoflurane affects the proliferation and metastatic potential of immortalized cervical cancer cell lines.
Methods
Cultured cervical cancer Caski and HeLa lines were exposed to 1, 2, or 3% sevoflurane for 2 or 4 h. Cell proliferation was determined through the Kit-8 assay and Ki-67 immunofluorescent staining. Cell migration and invasion were evaluated with the Transwell assay. Immunofluorescent staining and Western blot analysis were used to identify sevoflurane-induced morphological and biochemical changes.
Results
Sevoflurane exposure for either 2 or 4 h significantly increased HeLa cell proliferation in a time- and concentration-dependent manner to be 106 ± 2.7% and 107 ± 1.4% relative to the controls (n = 10; P = 0.036; P = 0.022) at 24 h after exposure and to be 106 ± 2.2% and 106 ± 1.7% relative to the controls (n = 10; P = 0.031; P = 0.023) at the highest concentration of 3% sevoflurane studied, respectively, but not Caski cells. Sevoflurane promoted invasion ability (1.63 ± 0.14 and 1.92 ± 0.12 relative to the controls) and increased cell size (1.69 ± 0.21 and 1.76 ± 0.13 relative to the controls) of Caski and HeLa cells (n = 6; all P < 0.001), respectively. Sevoflurane increased histone deacetylase 6 expression in both cells, and histone deacetylase 6 knockdown abolished the prometastatic effects of sevoflurane. Sevoflurane also induced deacetylation of α-tubulin in a histone deacetylase 6–dependent manner. The protein kinase B (AKT) or extracellular regulated protein kinase (ERK1/2) phosphorylation inhibition attenuated sevoflurane-induced histone deacetylase 6 expression.
Conclusions
Sevoflurane enhanced proliferation, migration, and invasion of immortalized cervical cancer cells, which was likely associated with increasing histone deacetylase 6 expression caused by phosphatidylinositide 3-kinase/AKT- and ERK1/2-signaling pathway activation.
Editor’s Perspective
What We Already Know about This Topic
What This Article Tells Us That Is New
Collapse
|
35
|
Wang X, Yao Y, Gao J. Sevoflurane inhibits growth factor-induced angiogenesis through suppressing Rac1/paxillin/FAK and Ras/Akt/mTOR. Future Oncol 2020; 16:1619-1627. [PMID: 32479124 DOI: 10.2217/fon-2020-0221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We investigated the direct effects of sevoflurane on angiogenesis and a variety of tumor cells. Materials & methods: The antiangiogenic activity of sevoflurane was determined using angiogenesis and biochemical assays. Results: Sevoflurane at low doses inhibits capillary network formation. Sevoflurane inhibited VEGF- and bFGF-stimulated migration, adhesion and growth in endothelial cells and induced apoptosis. Sevoflurane only at high doses inhibited growth and migration of tumor cells, suggesting differential effects of sevoflurane between endothelial and tumor cells. Mechanistically, sevoflurane decreased growth factors-induced Ras and Rac1 activation, and suppressed Ras and Rac1 signaling. Conclusion: We demonstrate the antiangiogenic effects of sevoflurane and provide preclinical evidence into the potential mechanisms by which sevoflurane may negatively affect cancer growth and metastasis.
Collapse
Affiliation(s)
- Xuesong Wang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| | - Yao Yao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| | - Jin Gao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| |
Collapse
|
36
|
Han XC, Zhang YJ, Dong X, Xing QZ, Li KH, Zhang L. Sevoflurane modulates the cancer stem cell-like properties and mitochondrial membrane potential of glioma via Ca 2+-dependent CaMKII/JNK cascade. Life Sci 2020; 253:117675. [PMID: 32360621 DOI: 10.1016/j.lfs.2020.117675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/01/2020] [Accepted: 04/11/2020] [Indexed: 12/24/2022]
Abstract
AIMS Gliomas are responsible for the majority of deaths from primary brain tumours. Sevoflurane showed inhibition effects on the tumor progression in vitro. However, whether sevoflurane could affect the stemness of glioma stem cells (GSCs) and the potential molecular mechanism have not been well elucidated. MAIN METHODS Effects of sevoflurane on cell viability, proliferation and invasion ability of glioma cells as well as tumor growth in vivo were assessed. Sphere formation assay was performed to evaluate the effect of sevoflurane on the stemness of GSCs. Effects of sevoflurane on mitochondrial function was evaluated by intracellular/mitochondrial reactive oxygen species (ROS) level and mitochondrial membrane potential. Expression levels of proliferation-related proteins, stemness markers and proteins in CaMKII/JNK cascade were measured by Western blot. KEY FINDINGS Sevoflurane inhibited the viability, proliferation and invasion ability of glioma cells (U87MG and U373MG). Western blot showed that sevoflurane decreased the expression levels of proliferation and invasion-related proteins. Sphere formation ability of GSCs, expression levels of stemness markers and mitochondrial function were significantly suppressed by sevoflurane. Moreover, sevoflurane treatment significantly increased the Ca2+ concentration and stimulated phosphorylation of CaMKII, JNK and IRS1. Ca2+ chelator BAPTA-AM combined with sevoflurane synergistically inhibited colony forming ability and the expression levels of proliferation-related proteins and stemness markers. In addition, the in vivo study further confirmed that sevoflurane inhibited tumor growth via Ca2+-dependent CaMKII/JNK cascade. SIGNIFICANCE The present study demonstrated that sevoflurane inhibited glioma tumorigenesis and modulated the cancer stem cell-like properties and mitochondrial membrane potential via activation of Ca2+-dependent CaMKII/JNK cascade.
Collapse
Affiliation(s)
- Xue-Chang Han
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China.
| | - Ya-Jie Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China
| | - Xu Dong
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China
| | - Qun-Zhi Xing
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China
| | - Ke-Han Li
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China
| | - Lu Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 JingHua Road, Jianxi District, Luoyang 471003, Henan Province, China
| |
Collapse
|
37
|
Cao Y, Lv W, Ding W, Li J. Sevoflurane inhibits the proliferation and invasion of hepatocellular carcinoma cells through regulating the PTEN/Akt/GSK‑3β/β‑catenin signaling pathway by downregulating miR‑25‑3p. Int J Mol Med 2020; 46:97-106. [PMID: 32319540 PMCID: PMC7255470 DOI: 10.3892/ijmm.2020.4577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 01/09/2020] [Indexed: 12/25/2022] Open
Abstract
Sevoflurane (Sevo) is one of the most frequently used volatile anesthetic agents in surgical oncology and has various effects on tumors, including inhibiting tumor growth, recurrence, and metastases; however, the molecular mechanisms are unknown. This study tried to investigate the influence of Sevo on hepatocellular carcinoma (HCC) cells and its possible mechanisms of action. The present study found that Sevo suppressed both the proliferative and invasive capabilities of both HCCLM3 and Huh7 cells in a dose-dependent manner. Moreover, 53 differentially expressed microRNAs (miRNAs/miRs) in HCC cells that resulted from Sevo were screened out using miRNA microarray assay. In particular, miR-25-3p displayed a significant decrease in response to Sevo treatment. Further studies showed that Sevo's inhibitory actions on HCC cells were attenuated by overexpression of miR-25-3p but enhanced by its inhibitor. Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN (PTEN), a tumor suppressor gene, was directly targeted by miR-25-3p and its expression was upregulated by Sevo. In addition, Sevo suppressed the expression of phosphorylated-protein kinase B (p-Akt) (S473), glycogen synthase kinase (GSK) 3β (p-GSK3β) (S9), β-catenin, c-Myc and matrix metalloproteinase 9; whereas these inhibitory effects were reversed by miR-25-3p overexpression. More importantly, Sevo's tumor-suppressive effects were enhanced by LY294002 (a PI3-kinase inhibitor) but weakened by insulin growth factor-1 (an agonist of the Akt signaling pathway). These data suggest that Sevo's antitumor effects on HCC could be explained, in part, by Sevo inhibiting the miR-25-3p/PTEN/Akt/GSK-3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yinghao Cao
- Department of Anesthesiology, Beijing Youan Hospital, Capital Medical University, Beijing 100048, P.R. China
| | - Wenfei Lv
- Department of Anesthesiology, Beijing Youan Hospital, Capital Medical University, Beijing 100048, P.R. China
| | - Wan Ding
- Department of Anesthesiology, No. 6 Medical Center, General Hospital of PLA, Beijing 100048, P.R. China
| | - Jun Li
- Department of Anesthesiology, No. 6 Medical Center, General Hospital of PLA, Beijing 100048, P.R. China
| |
Collapse
|
38
|
Su G, Yan Z, Deng M. Sevoflurane Inhibits Proliferation, Invasion, but Enhances Apoptosis of Lung Cancer Cells by Wnt/β-catenin Signaling via Regulating lncRNA PCAT6/miR-326 Axis. Open Life Sci 2020; 15:159-172. [PMID: 33987473 PMCID: PMC8114787 DOI: 10.1515/biol-2020-0017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
Sevoflurane was frequently used as a volatile anesthetic in cancer surgery. However, the potential mechanism of sevoflurane on lung cancer remains largely unclear. In this study, lung cancer cell lines (H446 and H1975) were treated by various concentrations of sevoflurane. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assessment and colony formation assay were performed to detect the cell viability and proliferation, separately. Also, transwell assay or flow cytometry assay was applied as well to evaluate the invasive ability or apoptosis in lung cancer cells, respectively. Western blot assay was employed to detect the protein levels of β-catenin and Wnt5a. Moreover, quantitative real-time polymerase chain reaction (qRT-PCR) was used to examine the expression level of prostate cancer-associated transcript 6 (PCAT6) and miR-326 in lung cancer tissues and cells. The target interaction between miR-326 and PCAT6 or Wnt5a was predicted by bioinformatics analysis and verified by the dual-luciferase reporter gene assay. Sevoflurane inhibited the abilities on viability, proliferation, invasion, and activation of Wnt/β-catenin signaling, but promoted apoptosis of H446 and H1975 cells in a dose-dependent manner. The expression of PCAT6 was increased in lung cancer tissues and cells, except for that of miR-326. Besides, sevoflurane could lead to expressed limitation of PCAT6 or improvement of miR-326. This process presented a stepwise manner. Up-regulation of PCAT6 restored the suppression of sevoflurane on abilities of proliferation, invasion, rather than apoptosis, and re-activated the Wnt5a/β-catenin signaling in cells. Moreover, the putative binding sites between miR-326 and PCTA6 or Wnt5a were predicted by starBase v2.0 software online. PCAT6 suppressing effects on cells could be reversed by pre-treatment with miR-326 vector. The promotion of Wnt5a inverted effects led from miR-326 or sevoflurane. Our study indicated that sevoflurane inhibited the proliferation, and invasion, but enhanced the apoptosis in lung cancer cells by regulating the lncRNA PCAT6/miR-326/Wnt5a/β-catenin axis.
Collapse
Affiliation(s)
- Guoning Su
- Department of Anesthesiology, Yunnan Second People's Hospital, No.176 Qingnian Road, Kunming, Yunan, 652600, China
| | - Zhibing Yan
- Department of Anesthesiology, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, China
| | - Min Deng
- Department of Anesthesiology, Yunnan Second People's Hospital, No.176 Qingnian Road, Kunming, Yunan, 652600, China
| |
Collapse
|
39
|
Xu Y, Jiang W, Xie S, Xue F, Zhu X. The Role of Inhaled Anesthetics in Tumorigenesis and Tumor Immunity. Cancer Manag Res 2020; 12:1601-1609. [PMID: 32184663 PMCID: PMC7061426 DOI: 10.2147/cmar.s244280] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
Inhaled anesthetics are widely used for induction and maintenance of anesthesia during surgery, including isoflurane, sevoflurane, desflurane, haloflurane, nitrous oxide (N2O), enflurane and xenon. Nowadays, it is controversial whether inhaled anesthetics may influence the tumor progression, which urges us to describe the roles of different inhaled anesthetics in human cancers. In the review, the relationships among the diverse inhaled anesthetics and patient outcomes, immune response and cancer cell biology were discussed. Moreover, the mechanisms of various inhaled anesthetics in the promotion or inhibition of carcinogenesis were also reviewed. In summary, we concluded that several inhaled anesthetics have different immune functions, clinical outcomes and cancer cell biology, which could contribute to opening new avenues for selecting suitable inhaled anesthetics in cancer surgery.
Collapse
Affiliation(s)
- Yichi Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shangdan Xie
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Fang Xue
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
40
|
Zhang L, Wang J, Fu Z, Ai Y, Li Y, Wang Y, Wang Y. Sevoflurane suppresses migration and invasion of glioma cells by regulating miR-146b-5p and MMP16. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3306-3314. [PMID: 31385537 DOI: 10.1080/21691401.2019.1648282] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background: Glioma is the most common brain tumor with poor prognosis all over the world. Anesthetics have been demonstrated to have important impacts on cell migration and invasion in different cancers. However, the underlying mechanism that allows anesthetics-mediated progression of glioma cells remains elusive. Methods: Sevoflurane (Sev), a class of common anesthetics, was used to expose to U87-MG and U251 cells. The expressions of microRNA-146b-5p (miR-146b-5p) and matrix metallopeptidase 16 (MMP16)were measured by quantitative real-time polymerase chain reaction or western blot. Transfection was performed in glioma cells with miR-146b-5p inhibitor, inhibitor negative control, MMP16 overexpression vector, empty vector, small interfering RNA against MMP16 or scramble. Cell migration and invasion were analyzed by the trans-well assay. The interaction between miR-146b-5p and MMP16 was explored by luciferase activity and RNA immunoprecipitation assays. Results: Sev treatment inhibited migration and invasion of glioma cells. The expression of miR-146b-5p was enhanced and MMP16 protein was decreased in glioma cells after exposure of Sev. Knockdown of miR-146b-5p or overexpression of MMP16 reversed Sev-induced inhibition of migration and invasion of glioma cells. Moreover, MMP16 was indicated as a target of miR-146b-5p and its silencing attenuated the regulatory role of miR-146b-5p abrogationin Sev-treated glioma cells. Conclusion: Sev impeded cell migration and invasion through regulating miR-146b-5p and MMP16 in glioma, indicating a novel theories foundation for the application of anesthetics like Sev in glioma.
Collapse
Affiliation(s)
- Le Zhang
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Jun Wang
- b Department of Operating Room, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Zhijie Fu
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - YanQiu Ai
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Yanrong Li
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Ying Wang
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Yanping Wang
- a Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
41
|
Zhang C, Wang B, Wang X, Sheng X, Cui Y. Sevoflurane inhibits the progression of ovarian cancer through down-regulating stanniocalcin 1 (STC1). Cancer Cell Int 2019; 19:339. [PMID: 31889892 PMCID: PMC6916020 DOI: 10.1186/s12935-019-1062-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Background Ovarian cancer is one of the leading causes of female death worldwide, with a poor prognosis of advanced patients. Sevoflurane, a volatile anesthetic commonly used in clinical operations, has been reported to have anti-cancer activity against some tumors. In the present study, we aimed to investigate the effects of sevoflurane on the progression of ovarian cancer and its potential mechanism. Methods The effects of sevoflurane on ovarian cancer cell viability, proliferation, migration, invasion, cell cycle, and apoptosis were determined by functional experiments in vitro. Gelatin zymography assay was performed to examine MMP9 activity. In vivo, sevoflurane was injected into mice of transplantation tumor with SKOV3 cells or with pcDNA-STC1 treated SKOV3 cells. Results We found that sevoflurane inhibited the viability of SKOV3 and OVCAR3 cells in a dose-dependent manner, and colony formation assay revealed that sevoflurane inhibited ovarian cancer cell colony-formation abilities. Additionally, sevoflurane could induce cell cycle arrest and promote cell apoptosis in SKOV3 and OVCAR3 cells. Moreover, sevoflurane reduced the migration and invasion abilities of SKOV3 and OVCAR3 cells, as well as the MMP-9 activity. Furthermore, sevoflurane down-regulated the expression of stanniocalcin 1 (STC1), and up-regulation of STC1 could reverse the inhibitory effects of sevoflurane on cell proliferation and invasion. In vivo, sevoflurane significantly inhibited the tumor growth, which was be reversed by STC1 overexpression. Conclusion These data reveal an anti-cancer activity of sevoflurane on the growth and invasion of ovarian cancer, which may be through down-regulating STC1. Sevoflurane may serve as a potential anti-cancer agent in ovarian cancer therapy.
Collapse
Affiliation(s)
- Chuanfeng Zhang
- 1Shandong Cancer Hospital Affiliated to Shandong University, Jinan, 250117 China.,2Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| | - Baosheng Wang
- 2Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| | - Xiuqin Wang
- 2Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| | - Xiugui Sheng
- 2Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China.,National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116 China
| | - Yongchun Cui
- 2Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| |
Collapse
|
42
|
Qi J, Wu Q, Zhu X, Zhang S, Chen X, Chen W, Sun Z, Zhu M, Miao C. Propofol attenuates the adhesion of tumor and endothelial cells through inhibiting glycolysis in human umbilical vein endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1114-1122. [PMID: 31650167 DOI: 10.1093/abbs/gmz105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Indexed: 12/19/2022] Open
Abstract
Propofol is one of the most commonly used intravenous anesthetics and plays an important role in tumor suppression. In the present study, we aimed to investigate the mechanism by which propofol attenuates tumor endothelial cells (TECs) and tumor cell adhesion to inhibit tumor metastasis in vitro. Human umbilical vein endothelial cells (HUVECs) cultured in Dulbecco's modified Eagle's medium were treated with tumor conditioned medium for 24 h, followed by 4 h of treatment with or without 25 μM of propofol, 10 μM of KN93, 500 μM of MK801, or 20 μM of rapastinel. It was found that propofol inhibited TEC adhesion and the glycolysis level of TECs. Consistently, propofol inhibited the expressions of adhesion molecules (E-selectin, ICAM-1, and VCAM-1) and glycolysis proteins (GLUT1, HK2, and LDHA) in TECs. Moreover, propofol attenuated the expression of HIF-1α, the phosphorylation of AKT and Ca2+/calmodulin-dependent protein kinase II (CaMKII), and the Ca2+ concentration in TECs. MK801, an inhibitor of NMDA receptor, and KN93, an inhibitor of CaMKII, both inhibited the expressions of adhesion molecules and glycolysis proteins, in a manner similar to propofol. Additionally, rapastine, an activator of NMDA receptor, could counteract the effects of propofol. Our results indicated that propofol attenuates intracellular Ca2+ concentration, CaMKII and AKT phosphorylation, and HIF-1α expression, probably via inhibiting the NMDA receptor, thus inhibiting glycolysis and adhesion of tumor and endothelial cells.
Collapse
Affiliation(s)
- Jie Qi
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qichao Wu
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xuqin Zhu
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shan Zhang
- Department of Anaesthesia, Critical Care and Pain Medicine, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiangyuan Chen
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wankun Chen
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhirong Sun
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Minmin Zhu
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Changhong Miao
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
43
|
Xue F, Xu Y, Song Y, Zhang W, Li R, Zhu X. The Effects Of Sevoflurane On The Progression And Cisplatinum Sensitivity Of Cervical Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3919-3928. [PMID: 31819366 PMCID: PMC6873969 DOI: 10.2147/dddt.s219788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/11/2019] [Indexed: 12/25/2022]
Abstract
Objective To investigate the effect of sevoflurane on the progression of cervical cancer cells, and to explore its effect on the cisplatinum (DDP) sensitivity in cervical cancer cells and underlying mechanism. Methods Siha and Hela cervical cancer cells were cultured and treated with 3% sevoflurane, 10 μmol/L DDP, or the co-treatment of sevoflurane and DDP, respectively. Cell proliferation was evaluated by the CCK8 assay. Cell apoptosis was assessed by flow cytometry. Cell migration was detected by wound healing assay. The expression of B-cell lymphoma-2 (BCL-2), B-cell lymphoma-2 associated X (BAX), Ezrin, matrix metalloproteinase 2 (MMP2), lung resistance-related protein (LRP), multiple drug resistance protein 1 (MRP1), glutathione-S-transferase-π (GST-π), and P glycoprotein (P-gp) protein was determined by Western blotting. Results After treated with sevoflurane, cell proliferation and migration rate in Siha and Hela cells were significantly elevated, while cell apoptosis was decreased. In addition, the expression of migration-related protein Ezrin and MMP2 was increased accordingly, apoptotic-related protein BCL-2 expression was also increased while BAX protein expression was decreased after sevoflurane treatment. The proliferation, migration rate, and apoptosis of Siha and Hela cells in sevoflurane plus DDP group were not significantly different with those in DDP group. There was no significant difference in apoptotic-related protein, migration-related protein, and drug resistance-associated proteins expression between DDP treatment group and combined treatment group. Conclusion Sevoflurane promotes the progression but has no effect on the cisplatinum sensitivity in cervical cancer cells.
Collapse
Affiliation(s)
- Fang Xue
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Yichi Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Yizuo Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Wenwen Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Ruyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| |
Collapse
|
44
|
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]
|
45
|
Yang X, Zheng YT, Rong W. Sevoflurane induces apoptosis and inhibits the growth and motility of colon cancer in vitro and in vivo via inactivating Ras/Raf/MEK/ERK signaling. Life Sci 2019; 239:116916. [PMID: 31626792 DOI: 10.1016/j.lfs.2019.116916] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 12/21/2022]
Abstract
AIMS To investigate the effects of sevoflurane on proliferation, cell cycle, apoptosis, autophagy, invasion and epithelial-mesenchymal transition of colon cancer cell line SW480, and to explore its possible mechanism. MATERIALS AND METHODS SW480 and SW620 cells were treated with a mixture of 95% O2+5% CO2 containing different concentrations of sevoflurane (1.7% SAV, 3.4% SAV and 5.1% SAV) for 6 h. Meanwhile, we performed a rescue experiment by treating cells with the ERK pathway activator LM22B-10 prior to treatment of cells with 5.1% sevoflurane。 KEY FINDINGS: High concentration (5.1%) of sevoflurane significantly inhibited the proliferation and invasion of cells, causing G0/G1 phase arrest and promoted apoptosis and autophagy. 5.1% sevoflurane can participate in the regulation of EMT by regulating the expression of E-cadherin, Vimentin and N-cadherin proteins. LM22B-10 promoted proliferation and invasion of cancer cells and inhibited apoptosis and autophagy, while 5.1% sevoflurane could reverse the effect of LM22B-10 on the biological characteristics of cells. Sevoflurane can significantly inhibit tumor growth in SW480 cells transplanted nude mice. Moreover, 5.1% sevoflurane significantly increased the expression of p-Raf, p-MEK1/2, and p-ERK1/2 in SW480 cells and tumor tissues without affecting p-JNK and p-p38 proteins, meanwhile, 5.1% sevoflurane can inhibit the activation of ERK signaling pathway by LM22B-10 in vitro and in vivo. SIGNIFICANCE Sevoflurane can inhibit the proliferation and invasion of colon cancer cells, induce apoptosis and autophagy, and participate in the regulation of epithelial-mesenchymal transition, which may be related to its inhibition of the ERK signaling pathway.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China
| | - Yao-Tun Zheng
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China
| | - Wei Rong
- Department of Anesthesiology, Weihai Central Hospital, Weihai, 264400, Shandong, China.
| |
Collapse
|
46
|
Song G, Tian L, Cheng Y, Liu J, Wang K, Li S, Li T. Antitumor activity of sevoflurane in HCC cell line is mediated by miR‐29a‐induced suppression of Dnmt3a. J Cell Biochem 2019; 120:18152-18161. [PMID: 31190353 DOI: 10.1002/jcb.29121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Guangming Song
- Department of Anesthesiology Dongying People's Hospital Dongying Shandong China
| | - Ling Tian
- Department of Operating Room Dongying People's Hospital Dongying Shandong China
| | - Yi Cheng
- Department of Anesthesiology Dongying People's Hospital Dongying Shandong China
| | - Jinshan Liu
- Department of Anesthesiology Dongying People's Hospital Dongying Shandong China
| | - Kun Wang
- Department of Operating Room Dongying People's Hospital Dongying Shandong China
| | - Shuai Li
- Department of Anesthesiology Dongying People's Hospital Dongying Shandong China
| | - Tianhua Li
- Department of Anesthesiology Dongying People's Hospital Dongying Shandong China
| |
Collapse
|
47
|
Gao K, Su Z, Liu H, Liu Y. RETRACTED: Anti-proliferation and anti-metastatic effects of sevoflurane on human osteosarcoma U2OS and Saos-2 cells. Exp Mol Pathol 2019; 108:121-130. [PMID: 30974101 DOI: 10.1016/j.yexmp.2019.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 01/17/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief and the authors. The journal was initially contacted by the corresponding author to request the retraction of the article as the results were reportedly not reproducible post-publication. Further investigation by the journal revealed also that the author name Hailin Liu was added to the authorship without editorial approval, after the article was accepted by the handling Editor. Other authorship issues as well as data realiability issues with the article were further revealed by the institutional investigation: http://med.china.com.cn/content/pid/291148/tid/1013/iswap/1. Given the concerns raised regarding panels from Figure 3A and also the comments of Dr Elisabeth Bik regarding this article “This paper belongs to a set of over 400 papers (as per February 2020) that share very similar Western blots with tadpole-like shaped bands, the same background pattern, and striking similarities in title structures, paper layout, bar graph design, and - in a subset - flow cytometry panels”, the journal requested the authors to provide the raw data. However, the authors were not able to fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Ke Gao
- Department of Cardiovascular, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Zhen Su
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Hailin Liu
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Yan Liu
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China.
| |
Collapse
|
48
|
Clinically relevant concentration of sevoflurane suppresses cervical cancer growth and migration through targeting multiple oncogenic pathways. Biochem Biophys Res Commun 2019; 514:1179-1184. [PMID: 31103261 DOI: 10.1016/j.bbrc.2019.05.082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023]
Abstract
The biological effects of sevoflurane, a volatile anesthetics, on cancer cells seem to be contradictory and are not fully understood. While some studies demonstrate that sevoflurane promotes tumor growth, other studies report that sevoflurane displays anti-cancer activities. In this work, we systematically investigated the effects of sevoflurane at clinically relevant dose on the multiple biological aspects of cervical cancer cells and analyzed the underlying mechanism. Using a panel of cell lines, we found that sevoflurane significantly inhibited proliferation and migration of cervical cancer cells regardless of cellular origin and genetic background. In contrast, sevoflurane did not affect cervical cancer survival. Additionally, sevoflurane significantly enhanced chemosensitivity of cervical cancer cells. Mechanistically, we show that sevoflurane inhibits Ras and RhoA GTPase activities, leading to the blockade of their downstream signaling pathways, such as Ras/Erk/Akt and Rho/MYPT1/MLC. The rescue studies using Rho activator calpeptin or constitutively active Ras further confirm that Ras and RhoA are the targets of sevoflurane in cervical cancer. Interestingly, we found that the anti-proliferative effect of sevoflurane was via targeting Ras whereas the anti-migratory effect of sevoflurane was mediated via targeting RhoA. Our data clearly demonstrates the anti-cancer effects of sevoflurane. These findings provide preclinical evidence into the potential mechanisms by which sevoflurane may negatively affect cervical cancer growth and metastasis.
Collapse
|
49
|
Fan L, Wu Y, Wang J, He J, Han X. Sevoflurane inhibits the migration and invasion of colorectal cancer cells through regulating ERK/MMP-9 pathway by up-regulating miR-203. Eur J Pharmacol 2019; 850:43-52. [PMID: 30685432 DOI: 10.1016/j.ejphar.2019.01.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 02/08/2023]
Abstract
Surgery resection is the primary treatment for colorectal cancer (CRC) patients with the risk of cancer dissemination and metastasis. Sevoflurane is one inhalational anesthesia which regulates migration and invasion in varying cancers. However, the effect of sevoflurane on CRC cells and its mechanism remain poorly understood. In this study, SW620 and HCT116 cells were treated with different concentrations of sevoflurane for 6 h in vitro. We measured the effect of sevoflurane on cell survival, migration and invasion by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide or trans-well assays. Moreover, we explored the interaction between sevoflurane and miR-203 and Roundabout1 (Robo1) as well as the extracellular signal-regulated kinase (ERK) and matrix metalloproteinase-9 (MMP-9) pathway. Results showed that sevoflurane inhibited cell migration and invasion in SW620 and HCT116 cells in a concentration dependent manner. Moreover, different concentrations of sevoflurane suppressed the phosphorylation of ERK. miR-203 expression was impaired while sevoflurane reversed the expression of miR-203 in CRC cells. In addition, inhibition of miR-203 attenuated the inhibitory effect of sevoflurane on cell migration, invasion and phosphorylated ERK level. Notably, MMP-9, as a downstream of ERK, was involved in sevoflurane-mediated processes in CRC cells. Besides, Robo1 was indicated as a target of miR-203 and inhibited by sevoflurane treatment. These results indicated that sevoflurane suppressed cell migration and invasion through regulating ERK/MMP-9 pathway via miR-203/Robo1 in CRC cells, indicating important clinical implications for anesthetic agents to prevent metastasis in CRC.
Collapse
Affiliation(s)
- Lihua Fan
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China.
| | - Yini Wu
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| | - Jianping Wang
- Department of Anorectal Surgery, Wenzhou Medical University, The Fifth Affiliated Hospital, Lishui, Zhejiang 323000, China.
| | - Jiaqun He
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| | - Xin Han
- Department of Anesthesiology, Wenzhou Medical University, The Sixth Affiliated Hospital, Lishui, Zhejiang 323000, China
| |
Collapse
|
50
|
Gao C, Shen J, Meng ZX, He XF. Sevoflurane Inhibits Glioma Cells Proliferation and Metastasis through miRNA-124-3p/ROCK1 Axis. Pathol Oncol Res 2019; 26:947-954. [PMID: 30915607 DOI: 10.1007/s12253-019-00597-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022]
Abstract
Malignant glioma is the most common primary malignancy in the brain. It is aggressive, highly invasive, and destructive. Studies have shown that sevoflurane can affect the invasion and migration of a variety of malignant tumors. However, its effects on human glioma cells and related mechanisms are not clear. Cultured U251 and U87 cells were pretreated with sevoflurane. The effect of sevoflurane on cell proliferation, migration, apoptosis and invasion ability were evaluated by MTT, wound healing assay, cell apoptosis and transwell assays, respectively. miRNA-124-3p and ROCK1 signaling pathway genes expression in sevoflurane treated cell lines was measured by quantitative real-time PCR (qRT-PCR) and western blotting analysis. The potential target genes of miRNA were predicted by online software. Luciferase reporter assay was employed to validate the direct targeting of ROCK1 by miRNA-124-3p. In present studies, sevoflurane inhibits glioma cells proliferation, invasion and migration. Additionally, inversely correlation between miR-124-3p and ROCK1 expression in sevoflurane treated glioma cells was observed. Furthermore, sevoflurane inhibits glioma cells proliferation, migration and invasion through miR-124-3p/ROCK1 axis. Taken together, our study revealed that sevoflurane can inhibit glioma cell proliferation, invasion and migration. Its mechanism may be related to the upregulation of miR-124-3p, which suppresses ROCK1 signaling pathway. The results of the study will help to understand the pharmacological effects of inhaled general anesthetics more comprehensively and help to provide an experimental basis for selecting more reasonable anesthetics for cancer patients.
Collapse
Affiliation(s)
- Cao Gao
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213002, People's Republic of China
| | - Jiang Shen
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213002, People's Republic of China
| | - Zhi-Xiu Meng
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213002, People's Republic of China
| | - Xiao-Feng He
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213002, People's Republic of China.
| |
Collapse
|