1
|
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
|
2
|
Prajapat M, Kaur G, Choudhary G, Pahwa P, Bansal S, Joshi R, Batra G, Mishra A, Singla R, Kaur H, Prabha PK, Patel AP, Medhi B. A systematic review for the development of Alzheimer's disease in in vitro models: a focus on different inducing agents. Front Aging Neurosci 2023; 15:1296919. [PMID: 38173557 PMCID: PMC10761490 DOI: 10.3389/fnagi.2023.1296919] [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: 09/19/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disease and is associated with dementia. Presently, various chemical and environmental agents are used to induce in-vitro models of Alzheimer disease to investigate the efficacy of different therapeutic drugs. We screened literature from databases such as PubMed, ScienceDirect, and Google scholar, emphasizing the diverse targeting mechanisms of neuro degeneration explored in in-vitro models. The results revealed studies in which different types of chemicals and environmental agents were used for in-vitro development of Alzheimer-targeting mechanisms of neurodegeneration. Studies using chemically induced in-vitro AD models included in this systematic review will contribute to a deeper understanding of AD. However, none of these models can reproduce all the characteristics of disease progression seen in the majority of Alzheimer's disease subtypes. Additional modifications would be required to replicate the complex conditions of human AD in an exact manner. In-vitro models of Alzheimer's disease developed using chemicals and environmental agents are instrumental in providing insights into the disease's pathophysiology; therefore, chemical-induced in-vitro AD models will continue to play vital role in future AD research. This systematic screening revealed the pivotal role of chemical-induced in-vitro AD models in advancing our understanding of AD pathophysiology and is therefore important to understand the potential of these chemicals in AD pathogenesis.
Collapse
Affiliation(s)
| | - Gurjeet Kaur
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | | - Paras Pahwa
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Seema Bansal
- MM College of Pharmacy, Maharishi Markandeshwar (DU) University, Mullana, Ambala, India
| | - Rupa Joshi
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Gitika Batra
- Department of Neurology, PGIMER, Chandigarh, India
| | - Abhishek Mishra
- Department of Biomedical Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Rubal Singla
- Department of Pharmacology, PGIMER, Chandigarh, India
| | | | | | | | - Bikash Medhi
- Department of Pharmacology, PGIMER, Chandigarh, India
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
Expression Profiles of Immune Cells after Propofol or Sevoflurane Anesthesia for Colorectal Cancer Surgery: A Prospective Double-blind Randomized Trial. Anesthesiology 2022; 136:448-458. [PMID: 35051263 DOI: 10.1097/aln.0000000000004119] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The antitumor effects of natural killer cells, helper T cells, and cytotoxic T cells after cancer surgery were reported previously. This study hypothesized that propofol-based anesthesia would have fewer harmful effects on immune cells than volatile anesthetics-based anesthesia during colorectal cancer surgery. METHODS In total, 153 patients undergoing colorectal cancer surgery were randomized and included in the analysis. The primary outcome was the fraction of circulating natural killer cells over time in the propofol and sevoflurane groups. The fractions of circulating natural killer, type 1, type 17 helper T cells, and cytotoxic T cells were investigated. The fractions of CD39 and CD73 expressions on circulating regulatory T cells were investigated, along with the proportions of circulating neutrophils, lymphocytes, and monocytes. RESULTS The fraction of circulating natural killer cells was not significantly different between the propofol and sevoflurane groups until 24 h postoperatively (20.4 ± 13.4% vs. 20.8 ± 11.3%, 17.9 ± 12.7% vs. 20.7 ± 11.9%, and 18.6 ± 11.6% vs. 21.3 ± 10.8% before anesthesia and after 1 and 24 h after anesthesia, respectively; difference [95% CI], -0.3 [-4.3 to 3.6], -2.8 [-6.8 to 1.1], and -2.6 [-6.2 to 1.0]; P = 0.863, P = 0.136, and P = 0.151 before anesthesia and after 1 and 24 h, respectively). The fractions of circulating type 1 and type 17 helper T cells, cytotoxic T cells, and CD39+ and CD73+ circulating regulatory T cells were not significantly different between the two groups. The neutrophil to lymphocyte ratio in both groups remained within the normal range and was not different between the groups. CONCLUSIONS Propofol-based anesthesia was not superior to sevoflurane-based anesthesia in terms of alleviating suppression of immune cells including natural killer cells and T lymphocytes during colorectal cancer surgery. EDITOR’S PERSPECTIVE
Collapse
|
5
|
Wu T, Sun L, Wang C, Yu P, Cheng L, Chen Y. Sevoflurane Suppresses the Migration, Invasion, and Epithelial-Mesenchymal Transition of Breast Cancer Cells Through the miR-139-5p/ARF6 Axis. J Surg Res 2020; 258:314-323. [PMID: 33317757 DOI: 10.1016/j.jss.2020.08.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 08/05/2020] [Accepted: 08/25/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Breast cancer (BC) is common cancer in female globally. Sevoflurane (SEV) has been reported to inhibit the metastasis of multiple cancers, including glioma, colorectal cancer, and hepatocellular carcinoma. However, the role of SEV in the metastasis of BC cells remains poorly understood. METHODS Transwell migration and invasion assays were performed to detect the migration and invasion of BC cells. Western blot assay was carried out to measure epithelial-mesenchymal transition (EMT)-related proteins in BC cells, including E-cadherin, N-cadherin, and fibronectin. Quantitative real-time polymerase chain reaction was conducted to determine the enrichment of miR-139-5p and ADP-ribosylation factor 6 (ARF6) in BC tissues and cells. The protein expression of ARF6 in BC tissues and cells was measured by western blot assay. The target of miR-139-5p was predicted by starBase software, and the target relationship between miR-139-5p and ARF6 in BC cells was confirmed by dual-luciferase reporter assay. RESULTS SEV suppressed the migration, invasion, and EMT of BC cells, especially in the high-concentration SEV group. The level of miR-139-5p was lower in BC tissues and cells than that in paired normal tissues and normal mammary epithelial cells MCF-10A. MiR-139-5p was upregulated in BC cells treated with SEV. ARF6 was upregulated in BC tissues and cells compared with that in corresponding normal tissues and normal mammary epithelial cells MCF-10A. SEV reduced the mRNA and protein expression of ARF6 in BC cells. The accumulation of ARF6 or the interference of miR-139-5p reversed the suppressive effects of SEV treatment on the migration, invasion, and EMT of BC cells. MiR-139-5p bound to ARF6 and inversely modulated the level of ARF6 in BC cells. The transfection of si-ARF6 attenuated the promoting effects of miR-139-5p depletion on the migration, invasion, and EMT of BC cells treated with SEV. CONCLUSIONS SEV suppressed the migration, invasion, and EMT of BC cells through downregulating the abundance of ARF6 by upregulating miR-139-5p. The miR-139-5p/ARF6 axis might be a promising target for the treatment of BC.
Collapse
Affiliation(s)
- Tongle Wu
- Department of Anesthe Siology, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China
| | - Luwei Sun
- Department of General Surgery, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China
| | - Chuantao Wang
- Department of Thoracic Surgery, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China
| | - Peng Yu
- Department of General Surgery, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China
| | - Long Cheng
- Department of General Surgery, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China
| | - Yongmin Chen
- Department of Anesthe Siology, People's Hospital of Weifang Binhai Economic and Technological Development Zone, Weifang, Shandong, China.
| |
Collapse
|
6
|
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: 7.4] [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
|
7
|
Action of copper(II) complex with β-diketone and 1,10-phenanthroline (CBP-01) on sarcoma cells and biological effects under cell death. Biomed Pharmacother 2019; 112:108586. [PMID: 30784909 DOI: 10.1016/j.biopha.2019.01.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/20/2018] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
This work reports the biological evaluation of a copper complex of the type [Cu(O-O)(N-N)ClO4], in which O-O = 4,4,4-trifluoro-1-phenyl-1,3-butanedione (Hbta) and N-N = 1,10-phenanthroline (phen), whose generic name is CBP-01. The cytotoxic effect of CBP-01 was evaluated by resazurin assay and cell proliferation was determined by MTT assay. DNA fragmentation was analyzed by gel electrophoresis. Cell cycle progression was detected through propidium iodide (PI) staining. Apoptosis and autophagy were determined by, respectively, Annexin V and 7-AAD staining and monodansylcadaverine (MDC) staining. The changes in intracellular reactive oxygen species levels were detected by DCFDA analysis. The copper complex CBP-01 showed in vitro antitumor activity with IC50s values of 7.4 μM against Sarcoma 180 and 26.4 against murine myoblast cells, displaying selectivity toward the tumor cell tested in vitro (SI > 3). An increase in reactive oxygen species (ROS) generation was observed, which may be related to the action mechanism of the complex. The complex CBP-01 may induce DNA damage leading cells to accumulate at G0/G1 checkpoint where, apparently, cells that are not able to recover from the damage are driven to cell death. Evidence has shown that cell death is initiated by autophagy dysfunction, culminating in apoptosis induction. The search for new metal-based drugs is focused on overcoming the drawbacks of already used agents such as acquired resistance and non-specificity; thus, the results obtained with CBP-01 show promising effects on cancer cells.
Collapse
|
8
|
Liu J, Yang L, Guo X, Jin G, Wang Q, Lv D, Liu J, Chen Q, Song Q, Li B. Sevoflurane suppresses proliferation by upregulating microRNA-203 in breast cancer cells. Mol Med Rep 2018; 18:455-460. [PMID: 29750301 DOI: 10.3892/mmr.2018.8949] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/20/2018] [Indexed: 11/05/2022] Open
Abstract
Rapid proliferation is one of the critical characteristics of breast cancer. However, the underlying regulatory mechanism of breast cancer cell proliferation is largely unclear. The present study indicated that sevoflurane, one of inhalational anesthetics, could significantly suppress breast cancer cell proliferation by arresting cell cycle at G1 phase. Notably, the rescue experiment indicated that miR-203 was upregulated by sevoflurane and mediated the function of sevoflurane on suppressing the breast cancer cell proliferation. The present study indicated the function of the sevoflurane/miR-203 signaling pathway on regulating breast cancer cell proliferation. These results provide mechanistic insight into how the sevoflurane/miR-203 signaling pathway supresses proliferation of breast cancer cells, suggesting the sevoflurane/miR-203 pathway may be a potential target in the treatment of breast cancer.
Collapse
Affiliation(s)
- Jiaying Liu
- Department of Anesthesiology, The Second Clinical College of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Longqiu Yang
- Department of Anesthesiology, Huangshi Central Hospital, Huangshi, Hubei 435000, P.R. China
| | - Xia Guo
- Department of Ophthalmology, Third People's Hospital of Jinan, Jinan, Shandong 250100, P.R. China
| | - Guangli Jin
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Qimin Wang
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Dongdong Lv
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Junli Liu
- Department of Anesthesiology, The Second Clinical College of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Qiu Chen
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Qiong Song
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| | - Baolin Li
- Department of Anesthesiology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan 450007, P.R. China
| |
Collapse
|
9
|
Ciechanowicz S, Zhao H, Chen Q, Cui J, Mi E, Mi E, Lian Q, Ma D. Differential effects of sevoflurane on the metastatic potential and chemosensitivity of non-small-cell lung adenocarcinoma and renal cell carcinoma in vitro. Br J Anaesth 2018; 120:368-375. [DOI: 10.1016/j.bja.2017.11.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2017] [Indexed: 01/05/2023] Open
|
10
|
Zhang Y, Li HJ, Wang DX, Jia HQ, Sun XD, Pan LH, Ye QS, Ouyang W, Jia Z, Zhang FX, Guo YQ, Ai YQ, Zhao BJ, Yang XD, Zhang QG, Yin N, Tan HY, Liu ZH, Yu JB, Ma D. Impact of inhalational versus intravenous anaesthesia on early delirium and long-term survival in elderly patients after cancer surgery: study protocol of a multicentre, open-label, and randomised controlled trial. BMJ Open 2017; 7:e018607. [PMID: 29187413 PMCID: PMC5719291 DOI: 10.1136/bmjopen-2017-018607] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Elderly patients who have solid organ cancer often receive surgery. Some of them may develop delirium after surgery and delirium development is associated with worse outcomes. Furthermore, despite all of the advances in medical care, the long-term survival in cancer patients is far from optimal. Evidences suggest that choice of anaesthetics during surgery, that is, either inhalational or intravenous anaesthetics, may influence outcomes. However, the impact of general anaesthesia type on the occurrence of postoperative delirium is inconclusive. Although retrospective studies suggest that propofol-based intravenous anaesthesia was associated with longer survival after cancer surgery when compared with inhalational anaesthesia, prospective studies as such are still lacking. The purposes of this randomised controlled trial are to test the hypotheses that when compared with sevoflurane-based inhalational anaesthesia, propofol-based intravenous anaesthesia may reduce the incidence of early delirium and prolong long-term survival in elderly patients after major cancer surgery. METHODS AND ANALYSIS This is a multicentre, open-label, randomised controlled trial with two parallel arms. 1200 elderly patients (≥65 years but <90 years) who are scheduled to undergo major cancer surgery (with predicted duration ≥2 hours) are randomised to receive either sevoflurane-based inhalational anaesthesia or propofol-based intravenous anaesthesia. Other anaesthetics and supplemental drugs including sedatives, opioids and muscle relaxants are administered in both arms according to routine practice. The primary early outcome is the incidence of 7-day delirium after surgery and the primary long-term outcome is the duration of 3-year survival after surgery. ETHICS AND DISSEMINATION The study protocol has been approved by the Clinical Research Ethics Committees of Peking University First Hospital (2015[869]) and all participating centres. The results of early and long-term outcomes will be analysed and reported separately. TRIAL REGISTRATION NUMBER ChiCTR-IPR-15006209; NCT02662257; NCT02660411.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Hui-Juan Li
- Project Development and Project Management Department, Peking University Clinical Research Institute, Beijing, China
| | - Dong-Xin Wang
- Department of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Hui-Qun Jia
- Department of Anesthesiology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xu-De Sun
- Department of Anesthesiology, Tang-Du Hospital Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ling-Hui Pan
- Department of Anesthesiology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qing-Shan Ye
- Department of Anesthesiology, Ningxia People’s Hospital, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Wen Ouyang
- Department of Anesthesiology, The Third Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Zhen Jia
- Department of Anesthesiology, Qinghai University Affiliated Hospital, Xining, Qinghai, China
| | - Fang-Xiang Zhang
- Department of Anesthesiology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yong-Qing Guo
- Department of Anesthesiology, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Yan-Qiu Ai
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-Jiang Zhao
- Department of Anesthesiology, Beijing Shijitan Hospital, Beijing, China
| | - Xu-Dong Yang
- Department of Anesthesiology, Peking University Hospital of Stomatology, Beijing, China
| | - Qin-Gong Zhang
- Department of Anesthesiology, Shanxi Provincial Cancer Hospital, Taiyuan, Shanxi, China
| | - Ning Yin
- Department of Anesthesiology, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| | - Hong-Yu Tan
- Department of Anesthesiology, Peking University Cancer Hospital&Institute, Key Laboratory of Carcinogenesis and Translational Research(Ministry of Education), Beijing, Beijing, China
| | - Zhi-Heng Liu
- Department of Anesthesiology, Shenzhen Second People’s Hospital, Shenzhen, Guangzhou, China
| | - Jian-Bo Yu
- Department of Anesthesiology, Tianjin Nankai Hospital, Tianjin, China
| | - Daqing Ma
- Department of Surgery and Cancer, Anaesthetics, Pain Medicine and Intensive Care Section, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
11
|
Bellanti F, Mirabella L, Mitarotonda D, Blonda M, Tamborra R, Cinnella G, Fersini A, Ambrosi A, Dambrosio M, Vendemiale G, Serviddio G. Propofol but not sevoflurane prevents mitochondrial dysfunction and oxidative stress by limiting HIF-1α activation in hepatic ischemia/reperfusion injury. Free Radic Biol Med 2016; 96:323-33. [PMID: 27154980 DOI: 10.1016/j.freeradbiomed.2016.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/16/2016] [Accepted: 05/01/2016] [Indexed: 02/05/2023]
Abstract
Mitochondrial dysfunction, reactive oxygen species (ROS) production and oxidative stress during reperfusion are determinant in hepatic ischemia/reperfusion (I/R) injury but may be impacted by different anesthetic agents. Thus, we aimed at comparing the effects of inhaled sevoflurane or intravenous propofol anesthesia on liver mitochondria in a rodent model of hepatic I/R injury. To this, male Wistar rats underwent I/R surgery using sevoflurane or propofol. In the I/R model, propofol limited the raise in serum aminotransferase levels as compared to sevoflurane. Mitochondrial oxygen uptake, respiratory activity, membrane potential and proton leak were altered in I/R; however, this impairment was significantly prevented by propofol but not sevoflurane. In addition, differently from sevoflurane, propofol limited hepatic I/R-induced mitochondria H2O2 production rate, free radical leak and hydroxynonenal-protein adducts levels. The I/R group anesthetized with propofol also showed a better recovery of hepatic ATP homeostasis and conserved integrity of mitochondrial PTP. Moreover, hypoxia-inducible factor 1 alpha (HIF-1α) expression was limited in such group. By using a cell model of desferoxamine-dependent HIF activation, we demonstrated that propofol was able to inhibit apoptosis and mitochondrial depolarization associated to HIF-1α action. In conclusion, hepatic I/R injury induces mitochondrial dysfunction that is not prevented by inhaled sevoflurane. On the contrary, propofol reduces liver damage and mitochondrial dysfunction by preserving respiratory activity, membrane potential and energy homeostasis, and limiting free radicals production as well as PTP opening. These hepatoprotective effects may involve the inhibition of HIF-1α.
Collapse
Affiliation(s)
- Francesco Bellanti
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Lucia Mirabella
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Domenica Mitarotonda
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maria Blonda
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Tamborra
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gilda Cinnella
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Alberto Fersini
- Department of General Surgery, University of Foggia, Foggia, Italy
| | - Antonio Ambrosi
- Department of General Surgery, University of Foggia, Foggia, Italy
| | - Michele Dambrosio
- Department of Anesthesia and Intensive Care, University of Foggia, Foggia, Italy
| | - Gianluigi Vendemiale
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gaetano Serviddio
- C.U.R.E. University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|