Skip to main content
Erschienen in: Journal of Anesthesia 1/2020

Open Access 31.10.2019 | Original Article

Differential effects of sevoflurane on the growth and apoptosis of human cancer cell lines

verfasst von: Takahiro Hirai, Yuko Konishi, Shoko Mizuno, Zhou Rui, Yao Sun, Kimitoshi Nishiwaki

Erschienen in: Journal of Anesthesia | Ausgabe 1/2020

Abstract

Purpose

There have been contradictory findings regarding the effects of sevoflurane on the oncogenic properties of cancer cells. This study was conducted to gain insights into the fundamental rules governing the differential effects of sevoflurane exposure on various cancer cells derived from multiple origins.

Methods

A series of cancer cell lines were exposed to 1% (v/v) sevoflurane for 2–8 h and then assessed for their proliferation, Matrigel invasion, and apoptotic cell death, in comparison with their untreated counterparts. Cell proliferation and Matrigel invasion assays were performed using Coulter counter and Boyden chamber techniques, respectively. Apoptosis was evaluated by staining cells with Annexin V and 7-AAD followed by fluorescence flow cytometry. In addition, the expression of cleaved caspase-3 protein, another marker of apoptosis, was assessed using immunoblotting.

Results

Proliferation was significantly enhanced after sevoflurane exposure in six of eight cancer cell lines (NCI-H1299, MDA-MB-231, HCT116, DLD-1, HT29, and RKO). In contrast, sevoflurane attenuated proliferation in the last two cancer cell lines, A549 and MCF-7, as well as in the non-cancerous MCF10A cell line. Cell biological assays using four cancer cell lines demonstrated that accelerated but not attenuated cancer cell proliferation after sevoflurane exposure is associated with enhanced Matrigel invasion and suppressed apoptosis.

Conclusion

Sevoflurane augmented or hampered cell proliferation and Matrigel invasion depending on the cancer cell line examined. Loss of sevoflurane-induced apoptosis occurring in cancer cell lines is likely to be correlated with their enhanced proliferation after sevoflurane exposure.
Hinweise
The original version of this article was revised due to retrospective open access cancellation order.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s00540-019-02719-0.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Sevoflurane, a volatile anesthetic, has commonly been used during surgery, including tumor resection, as it allows for rapid and predictable recovery relative to intravenously administered anesthetics [13]. However, growing in vitro evidence suggests that sevoflurane potentially plays a role in the progression of tumors [4, 5]. Sevoflurane-exposed HepG2, a human hepatocellular carcinoma cell line, exhibits accelerated proliferation under conditions of high glucose and insulin [6]. Sevoflurane also elicits accelerated growth of cultured glioma stem cells through upregulation of hypoxia-inducible factors (HIF; Ref [7]). Another volatile anesthetic, isoflurane, reportedly activates the HIF signaling pathway and promotes proliferation and migration of the renal cancer cell line RCC4 [8]. Isoflurane also suppresses apoptosis in colon cancer cell lines via a caveolin-1-dependent mechanism [9].
In contrast, other studies have suggested anti-tumorigenic effects of sevoflurane [1012]. It has been demonstrated that sevoflurane attenuates the invasion of colon and lung cancer cells by suppressing the expression of metalloproteinase-9 (MMP-9) [13, 14], and induces apoptosis in Caco-2 and HEp-2 cells [12]. The growth of leukemia stem/progenitor cells is hampered by sevoflurane exposure via the suppression of Wnt/β-catenin activity [15]. As such, sevoflurane has been shown to exert diverse effects on the oncogenic properties of cancer cells, and a number of molecules and biological processes are reportedly involved in altered cell functions induced by sevoflurane. However, it remains to be fully elucidated whether or not differential proliferation of cancer cells after sevoflurane exposure is governed by a fundamental key determinant that is globally applicable to various types of cancer.
In this study, we exposed eight human cancer cell lines of multiple origins as well as a non-cancerous breast epithelial cell line to 1–3% (v/v) sevoflurane, and analyzed its impact on cell proliferation. In addition, we investigated Matrigel invasion, early apoptosis, and cell death in a series of cancer cell lines exposed to sevoflurane, and determined whether these biological processes are correlated with the proliferation of cancer cell lines.

Materials and methods

Cell lines, reagents, and antibodies

The human breast cancer cell line MDA-MB-231; colon cancer cell lines HCT116, HT-29, and RKO; lung cancer cell line NCI-H1299; and a spontaneously immortalized human breast epithelial cell line, MCF10A, were obtained from the American Tissue Culture Collection (ATCC; Manassas, VA, USA). The human breast cancer cell line MCF7, colon cancer cell line DLD-1, and lung cancer cell line A549 were obtained from the National Institutes of Biomedical Innovation (NIBIO; Osaka, Japan). MCF10A cells were cultured in Dulbecco's modified Eagle’s medium/nutrient mixture F-12 (D-MEM/F-12; NacalaiTesque, Kyoto, Japan) supplemented with 5% horse serum (Biowest, Nuaille, France), 20 ng/mL epidermal growth factor, 10 μg/mL insulin, 0.5 μg/mL hydrocortisone, and 0.1 μg/mL cholera toxin (Sigma-Aldrich, St. Louis, MO, USA). Cell lines other than MCF10A were cultured in D-MEM (NacalaiTesque) supplemented with 5% fetal bovine serum (Atlas Biologicals, Ft. Collins, CO, USA) and 1% penicillin–streptomycin (NacalaiTesque). Cell lines were maintained at 37 °C with 5% CO2 in a humidified incubator (Thermo Fisher Scientific, Waltham, MA, USA), except when the cells were exposed to sevoflurane. Cells were reseeded every 3–4 days prior to reaching confluence.

Sevoflurane exposure

Cell lines were plated in tissue culture plates (TPP, Trasadingen, Switzerland) with their respective media and placed in a CO2 incubator (Thermo Fisher Scientific). At 24 h post seeding, adherence of the cells to the wells and the confluency between 40 and 80% were confirmed by phase-contrast microscopy. Cells were then subjected to 2–8 h exposure to 1–3% (v/v) sevoflurane (Maruishi Pharmaceutical, Osaka, Japan) with 5% CO2 in atmospheric air at a flow rate of 0.5 L/min, at 37 °C in a humidified CO2 incubator (ASTEC, Fukuoka, Japan). The specific time courses of sevoflurane exposure in respective experiments are schematically shown in the corresponding figures. The concentrations of sevoflurane and CO2 in the incubator were monitored using a Vamos Plus Anaesthetic Gas analyzer (Dräger, Lubeck, Germany). After exposure to sevoflurane, cells were transferred to a CO2 incubator (Thermo Fisher Scientific) pre-set to a standard culture condition, and further incubated until they were examined in assays. Untreated controls were prepared by seeding the cells to tissue culture plates together with the other samples, left unexposed to sevoflurane, incubated, and examined in parallel with the other samples.

Proliferation assay

Cell lines were seeded in 6-well tissue culture plates at a density of 4 × 104 cells/cm2 in 1.0 mL media, exposed to 1–3% (v/v) sevoflurane for predetermined periods of time, and then incubated for 48 h. In the experiments in which 1% sevoflurane was applied, cell lines were dissociated with trypsin (Nacalai Tesque) and enumerated using a Coulter counter (Beckman Coulter, Brea, CA, USA). When cell lines were treated with 1.5–3.0% sevoflurane, confluency of the cells was assessed using the Incucyte Zoom live-cell microscopy incubator system (Essen BioScience, Ann Arbor, MI, USA) as a surrogate for cell number. During assays, the exponential growth of untreated control cells, as well as the proliferation of cells without reaching confluence in any condition, was confirmed by phase-contrast microscopy.

Cell invasion assay

Cell invasion to Matrigel was assessed using Transwell cell culture chambers with 8.0 μm pores (Corning, Oneonta, NY, USA). The Transwell inserts were coated with 200 μL/well of 300 μg/mL Matrigel (Corning) and air dried for 2 h at 37 °C in a regular CO2 incubator. Filters in the Transwell inserts were reconstituted with serum-free D-MEM immediately before use. Cells were incubated with or without 1% sevoflurane in a CO2 incubator for 4 h, dissociated with trypsin, and seeded in triplicate into the Transwell inserts (5 × 104 cells/well). The lower compartment was filled with D-MEM supplemented with 5% FBS. After 24 h of incubation at 37 °C with 5% CO2, the filters were fixed using 0.5% paraformaldehyde and stained with 0.1% crystal violet in PBS. The migration through Matrigel was scored under microscopy at a magnification of 40× (AXIO, Carl Zeiss Microscopy, Oberkochen, Germany).

Detection of early apoptosis and cell death

Cells seeded in 6-well tissue culture plates at a density of 8 × 104 cells/well were exposed to 1–3% sevoflurane and dissociated and collected after 24 h of incubation. Collected cells were washed with PBS once, stained with fluorescein isothiocyanate (FITC)-conjugated Annexin V (100-fold dilution; Medical & Biological Laboratories, Aichi, Japan) for 20 min, again washed once with PBS, and then stained with 7-aminoactinomycin D (7-AAD; 500-fold dilution; AAT Bioquest, Sunnyvale, CA, USA) for 15 min. The ratios of FITC-positive and 7-AAD-positive cells were determined using flow cytometry in a Gallios Flow Cytometer (Beckman Coulter). Data were analyzed using Kaluza software (Beckman Coulter).

Immunoblotting

Cells seeded in 6-well tissue culture plates at 6 × 104 cells/well were exposed to 1% sevoflurane in air for the predetermined duration, and incubated for 24 h with or without the pan-caspase inhibitor Z-VAD-FMK (20 µM; AdooQBioScience, Irvine, CA, USA). Whole-cell extracts were lysed by sonication in RIPA buffer (NacalaiTesque), and protein concentration in each extract was determined using a BCA protein assay (Thermo Scientific, Rockford, IL, USA). Equal amounts of protein (50 µg) were resolved via SDS-PAGE using Real Gel plate (Bio Craft, Tokyo, Japan), electro-transferred to an Immobilon-P polyvinylidene fluoride membrane (Merck KGaA, Darmstadt, Germany), and probed with primary and horseradish-peroxidase-conjugated secondary antibodies. The primary antibodies used in this study are anti-caspase-3 rabbit polyclonal antibody (1:1000 dilution; Cell Signaling Technology, Danvers, MA, USA) and anti-β-actin mouse monoclonal antibody (1:2000 dilution; Medical & Biological Laboratories). Chemiluminescent signals were captured and imaged using Amersham Imager 600 (GE Healthcare Life Sciences, Pittsburgh, PA, USA).

Statistical analysis

Data are reported as mean ± SD values; n represents the number of times an independent assay was performed. One-way-ANOVA and Scheffe’s post-hoc test were performed for cell proliferation, Annexin-V, and 7-AAD assays, whereas two-sided Student’s t-test was applied for cell invasion assays. All statistical analyses were performed using IBM SPSS statistics 24 (SPSS Inc., Chicago, IL, USA). A P value less than 0.05 indicated statistical significance.

Results

1% Sevoflurane exerts differential effects on cell growth depending on the specific cell line

To comprehensively understand the impact of sevoflurane exposure on cancer cell growth, we performed cell proliferation assays using eight cancer cell lines. The cell lines examined included two non-small cell lung cancer cell lines (NCI-H1299 and A549), two breast cancer cell lines (MDA-MB-231 and MCF-7), and four colon cancer cell lines (HCT116, DLD-1, HT29, and RKO). A spontaneously immortalized human breast epithelial cell line MCF10A was also included in this assay as a non-cancerous cell control. These cell lines were exposed to 1% sevoflurane in a CO2 incubator for 2–8 h. After exposure, the cells were allowed to grow for 48 h, and the numbers of cells was determined (Fig. 1a). The results revealed that 4–8 h exposure to sevoflurane augmented proliferation in NCI-H1299 by 1.3-fold (P = 0.01), MDA-MB-231 by 1.4-fold (P = 0.01), DLD-1 by 1.3-fold (P = 0.03), HCT116 by 1.6-fold (P = 0.001), RKO by 1.3-fold (P = 0.0001), and HT29 by 1.2-fold (P = 0.0001), compared with their respective untreated controls. In NCI-H1299 and MDA-MB-231, 8 h exposure to sevoflurane resulted in a suppression of cell growth compared with 4 h exposure, suggesting that a long-term exposure to sevoflurane may cause modest cellular toxicity and cancel the growth promotion conferred by sevoflurane itself in these cell lines. In the cell lines A549, MCF-7, and MCF10A, sevoflurane reduced cell proliferation by 24% (P = 0.0001), 13% (P = 0.002), and 21% (P = 0.04), respectively, largely in an exposure time-dependent fashion (Fig. 1b).
Collectively, these results demonstrate that sevoflurane differentially perturbs cell proliferation among human cancer cell lines regardless of their organ origins, and it attenuates the proliferation of non-cancerous cells as described previously [16].

1% sevoflurane enhances matrigel invasion in parallel with proliferation in cancer cell lines

Invasion to extracellular matrix is a notable property of cancer cells, which in fact was found to be suppressed after sevoflurane exposure in a few types of cancer cells [13, 14]. To address how sevoflurane exposure in our experimental condition affects cancer cell invasion, we performed Boyden chamber-based Matrigel invasion assays using four cancer cell lines originating from lung (NCI-H1299 and A549) and breast (MDA-MB-231 and MCF-7). These cell lines were employed because sevoflurane triggered contradictory changes in proliferation capacity in these pairs of cell lines derived from the same organs.
As shown in Fig. 2, sevoflurane significantly suppressed Matrigel invasion by A549 (23% reduction, P = 0.02) and MCF7 (14% reduction, P = 0.007), two cell lines exhibiting attenuated growth capacities after sevoflurane exposure. In contrast, invasion into Matrigel was upregulated (MDA-MB-231; 2.0-fold, P = 0.007) or not altered significantly (NCI-H1299) upon sevoflurane exposure in the cell lines whose proliferation was enhanced after sevoflurane exposure. These data suggest that alteration in Matrigel invasion occurs in parallel with that in proliferation capacity in cancer cell lines. Of note, changes in the number of Matrigel-invading cells after sevoflurane exposure may result from altered cellular capacity to invade Matrigel, altered proliferation rate, or both.

1% sevoflurane induces early apoptosis along with growth retardation in some cancer cell lines

We subsequently addressed apoptosis, one of the biological processes that may be associated with attenuated cell proliferation, in cells exposed to sevoflurane. We used the same four cell lines derived from lung (NCI-H1299 and A549) and breast (MDA-MB-231 and MCF-7) cancers in this assay. Incidence of early apoptosis was evaluated by staining cells with fluorescence-labeled Annexin V, a cellular protein that specifically binds to phosphatidylserine exposed to the outer surface of the plasma membrane in an early phase of apoptosis.
The cells were exposed to sevoflurane for 2–8 h and, after a 24-h incubation period, incubated with FITC-conjugated Annexin V (Fig. 3a). Fluorescence flow cytometric analyses demonstrated 4.1-fold (P = 0.0001) and 1.5-fold (P = 0.01) increases in the ratio of apoptotic cells relative to the untreated controls in A549 and MCF-7, respectively. Conversely, the apoptotic ratio of NCI-H1299 was reduced by 40% (P = 0.0001), and no significant change in apoptosis was observed in MDA-MB-231, after sevoflurane exposure (Fig. 3b, c).
Induction of early apoptosis in the four cell lines was also assessed by a distinct assay quantifying cleaved (i.e., activated) caspase-3, a protein playing a critical role in the signaling pathway initiating apoptotic processes. Cells were exposed to 1% sevoflurane for 4–8 h and processed for immunoblotting against caspase-3 after a 24 h incubation period (Fig. 4a). This assay demonstrated that cleaved caspase-3 accumulated in A549 and MCF-7 cells but decreased in NCI-H1299 and MDA-MB-231 cells after sevoflurane exposure (Fig. 4b). These data collectively suggest that sevoflurane elicits early apoptosis only in cell lines wherein proliferation is hampered by sevoflurane exposure.

1% sevoflurane induces cell death along with growth retardation in some cancer cell lines

We subsequently investigated the effects of sevoflurane on cell death using 7-AAD. 7-AAD is a fluorescent dye that exclusively marks nonviable cells by moving into the nuclei and intercalating into genomic DNA only when the cell membrane is disintegrated during the late phase of apoptosis. The four cell lines (NCI-H1299, MDA-MB-231, A549, and MCF-7) were first exposed to 1% sevoflurane for 4–8 h, maintained for 24 h after cessation of exposure, stained with 7-AAD, and then analyzed using flow cytometry (Fig. 5a). As shown in Fig. 5b, c, the proportion of 7-AAD-stained cells among A549 and MCF-7 cells significantly increased by 1.7-fold (P = 0.0001) and 1.2-fold (P = 0.006), respectively, in an exposure-time-dependent manner. In contrast, NCI-H1299 and MDA-MB-231 showed no statistically significant difference in the proportion of 7-AAD-stained cells relative to the non-exposed controls. Altogether, these results suggest that the suppression, but not the acceleration, of cell proliferation after sevoflurane exposure is accompanied by an increase in early apoptosis and cell death.

High-dose sevoflurane induces differential cell proliferation and apoptotic cell death in cancer cell lines, similar to 1% sevoflurane

Lastly, we exposed the same set of cell lines (NCI-H1299, MDA-MB-231, A549, and MCF-7) to higher concentrations of sevoflurane and evaluated its effects on cell proliferation, early apoptosis, and cell death. In the assessment of cell proliferation, the cell lines were treated with 1.5%, 2.0%, and 3.0% sevoflurane for 4 h, incubated for 48 h, and analyzed by whole well imaging using the Incucyte Zoom system to determine cell confluency (Fig. 6a). We found that exposure to sevoflurane at any tested concentration augmented cell growth in NCI-H1299 and MDA-MB-231 cells, while attenuating cell growth in A549 and MCF-7 cells (Fig. 6b), when compared to unexposed controls in the same cell lines. Increasing the sevoflurane concentration from 1% (v/v) to a higher concentration resulted in slight reduction of the cell numbers/confluency in NCI-H1299, MDA-MB-231, and A549 cells, suggesting potential cellular toxicity caused by high-dose sevoflurane (Figs. 1b and 6b).
To assess early apoptosis and cell death after sevoflurane exposure, cell lines were exposed to 2% and 3% sevoflurane for 4 h, maintained for 24 h, fluoro-stained for Annexin V and 7-AAD, and analyzed by flow cytometry (Fig. 7a). The results demonstrated that both early apoptosis and cell death were reduced in NCI-H1299 and MDA-MB-231 cells, but increased in A549 and MCF-7 cells when cells were exposed to sevoflurane (Fig. 7b, c). Collectively, these data indicate that high-dose sevoflurane differentially elicited cell proliferation, early apoptosis, and cell death in individual cell lines, similar to the results obtained with 1% sevoflurane.

Discussion

In this study, we found an acceleration of cell growth upon exposure to clinically relevant 1% (v/v) sevoflurane in six out of eight human cancer cell lines. In contrast, the last two cancer cell lines, A549 and MCF-7, and a non-cancerous cell line MCF10A, exhibited suppressed cell growth under the same experimental condition. The growth-suppressed cell lines but not growth-accelerated cell lines demonstrated reduced Matrigel invasion, enhanced early apoptotic changes, and increased cell death, upon exposure to 1% sevoflurane. To the best of our knowledge, this is the first report indicating a solid correlation between apoptosis and hampered cell growth post sevoflurane exposure, in multiple cancer cell lines derived from various organ origins. This study suggests that the differential induction of apoptosis is, at least in part, responsible for the distinct growth properties among individual cell lines post sevoflurane exposure. However, the possibility remains that sevoflurane not only perturbs apoptosis but also increases or reduces the proliferation capacity per se of individual cell lines, both of which can explain the observed up/downregulation of cell number by sevoflurane.
Previous studies have demonstrated that in vitro exposure of cells to sevoflurane results in differential induction of apoptosis depending on the cellular context. Sevoflurane was shown to trigger apoptotic changes in several types of noncancerous and cancerous cells, including fetal neural stem cells and the Caco-2, HEp-2, A549, and H4 cell lines [11, 14, 17]. Halothane, another volatile anesthesia, was also shown to elicit apoptosis in several cancer cell lines, including Caco-2, HEp-2 and A549 [11, 18]. In contrast, Jaura et al. demonstrated that sera derived from patients who received sevoflurane and opioids reduce apoptosis when added to MDA-MB-231 cell culture [5]. In addition, apoptotic cells were increased in HT29 but reduced in HCT116 after isoflurane exposure [9], despite the fact that these cancer cell lines originate from the same tissue. These previous findings as well as our data in this study collectively demonstrate that individual cancer cell lines exhibit differential apoptotic changes after sevoflurane exposure, whereas noncancerous cells may generally exhibit increased apoptosis [16, 19]. It is speculated that the intrinsic susceptibility to sevoflurane has been lost from a subset of cancer cell lines as a consequence of their gaining the ability to evade apoptosis, which had been acquired during oncogenesis through the accumulation of genetic and epigenetic changes within the genome [20].
The experimental results obtained in this study were largely consistent with previous reports studying cell proliferation and apoptosis after sevoflurane exposure; this includes the phenotypic changes observed in MDA-MB-231 [4] and A549 [14, 18] in response to sevoflurane exposure. A previous study showed enhancement of cell proliferation, migration, and invasion in MCF7 cells upon exposure to sevoflurane [4], which is somewhat inconsistent with our observation. However, as the previous study did not assess the induction of apoptosis after sevoflurane exposure, its findings provide no argument regarding the probable correlation between proliferation and apoptosis in sevoflurane-exposed cancer cells as suggested by our study. The disparities in growth properties of sevoflurane-exposed MCF7 cells between that study and ours are likely to be a result of differences in experimental design, including the approach and duration of sevoflurane exposure and cell culture condition during and post exposure.
The molecular mechanisms underlying sevoflurane-induced apoptosis remain largely unclear. It has been suggested that the activation of c-JUN N-terminal kinase (JNK), a well-characterized mediator of apoptotic signaling [21], plays an important role in the induction of apoptosis after sevoflurane exposure [19]. Apoptosis induced by isoflurane was shown to be suppressed via a mechanism depending on caveolin-1, a protein composing caveola membrane, in colon cancer cell lines [9]. Collectively, volatile anesthetics seem to induce apoptosis primarily by activating cellular apoptotic signaling, rather than by causing DNA damage in the genome which triggers p53-mediated apoptosis. However, it remains unclear how the apoptotic signaling is initiated by volatile anesthetics, and further studies are warranted to fully understand the apoptotic process elicited by volatile anesthetics as well as the genetic alterations of cancer cells permitting the evasion of apoptosis. The evaluation of sevoflurane-induced apoptosis in animal models and patient samples will provide additional insight into this topic in preclinical and clinical settings. In the future, these studies could enable us to predict whether individual tumors are responsive to sevoflurane-induced apoptosis by examining preoperative biopsy specimens. Such efforts will eventually lead to safe, personalized use of volatile anesthetics for cancer treatment without concern for tumor progression.

Acknowledgements

We wish to thank Mr. Minoru Tanaka in the Division of Medical Research Engineering, Nagoya University Graduate School of Medicine, for assistance with flow cytometry. This study was supported by Grants-in-Aid for Scientific Research (KAKENHI) from the Japan Society for the Promotion of Science (JSPS) [15K15569 to TH, 16K10932 to YK, 15K20042 to SM], as well as the Program to supporting research activities of female researchers from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan, 2016–2018 (to YK). This study constitutes the doctoral thesis of T.H.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no competing interests.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Anästhesiologie

Kombi-Abonnement

Mit e.Med Anästhesiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes AINS, den Premium-Inhalten der AINS-Fachzeitschriften, inklusive einer gedruckten AINS-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Robinson BJ, Uhrich TD, Ebert TJ. A review of recovery from sevoflurane anaesthesia: comparisons with isoflurane and propofol including meta-analysis. Acta Anaesthesiol Scand. 1999;43(2):185–90.CrossRef Robinson BJ, Uhrich TD, Ebert TJ. A review of recovery from sevoflurane anaesthesia: comparisons with isoflurane and propofol including meta-analysis. Acta Anaesthesiol Scand. 1999;43(2):185–90.CrossRef
2.
Zurück zum Zitat Tavare AN, Perry NJS, Benzonana LL, Takata M, Ma D. Cancer recurrence after surgery: Direct and indirect effects of anesthetic agents. Int J Cancer. 2012;130(6):1237–50.CrossRef Tavare AN, Perry NJS, Benzonana LL, Takata M, Ma D. Cancer recurrence after surgery: Direct and indirect effects of anesthetic agents. Int J Cancer. 2012;130(6):1237–50.CrossRef
3.
Zurück zum Zitat Brioni JD, Varughese S, Ahmed R, Bein B. A clinical review of inhalation anesthesia with sevoflurane: from early research to emerging topics. J Anesth. 2017;31(5):764–78.CrossRef Brioni JD, Varughese S, Ahmed R, Bein B. A clinical review of inhalation anesthesia with sevoflurane: from early research to emerging topics. J Anesth. 2017;31(5):764–78.CrossRef
4.
Zurück zum Zitat Ecimovic P, McHugh B, Murray D, Doran P, Buggy DJ. Effects of sevoflurane on breast cancer cell function in vitro. Anticancer Res. 2013;33(10):4255–60.PubMed Ecimovic P, McHugh B, Murray D, Doran P, Buggy DJ. Effects of sevoflurane on breast cancer cell function in vitro. Anticancer Res. 2013;33(10):4255–60.PubMed
5.
Zurück zum Zitat Jaura AI, Flood G, Gallagher HC, Buggy DJ. Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study. Br J Anaesth. 2014;113(Suppl 1):i63–i6767.CrossRef Jaura AI, Flood G, Gallagher HC, Buggy DJ. Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study. Br J Anaesth. 2014;113(Suppl 1):i63–i6767.CrossRef
6.
Zurück zum Zitat Nishiwada T, Kawaraguchi Y, Uemura K, Sugimoto H, Kawaguchi M. Effect of sevoflurane on human hepatocellular carcinoma HepG2 cells under conditions of high glucose and insulin. J Anesth. 2015;29(5):805–8.CrossRef Nishiwada T, Kawaraguchi Y, Uemura K, Sugimoto H, Kawaguchi M. Effect of sevoflurane on human hepatocellular carcinoma HepG2 cells under conditions of high glucose and insulin. J Anesth. 2015;29(5):805–8.CrossRef
7.
Zurück zum Zitat Shi QY, Zhang SJ, Liu L, Chen QS, Yu LN, Zhang FJ, Yan M. Sevoflurane promotes the expansion of glioma stem cells through activation of hypoxia-inducible factors in vitro. Br J Anaesth. 2015;114(5):825–30.CrossRef Shi QY, Zhang SJ, Liu L, Chen QS, Yu LN, Zhang FJ, Yan M. Sevoflurane promotes the expansion of glioma stem cells through activation of hypoxia-inducible factors in vitro. Br J Anaesth. 2015;114(5):825–30.CrossRef
8.
Zurück zum Zitat Benzonana LL, Perry NJ, Watts HR, Yang B, Perry IA, Coombes C, Takata M, Ma D. Isoflurane, a commonly used volatile anesthetic, enhances renal cancer growth and malignant potential via the hypoxia-inducible factor cellular signaling pathway in vitro. Anesthesiology. 2013;119(3):593–605.CrossRef Benzonana LL, Perry NJ, Watts HR, Yang B, Perry IA, Coombes C, Takata M, Ma D. Isoflurane, a commonly used volatile anesthetic, enhances renal cancer growth and malignant potential via the hypoxia-inducible factor cellular signaling pathway in vitro. Anesthesiology. 2013;119(3):593–605.CrossRef
9.
Zurück zum Zitat Kawaraguchi Y, Horikawa YT, Murphy AN, Murray F, Miyanohara A, Ali SS, Head BP, Patel PM, Roth DM, Patel HH. Volatile anesthetics protect cancer cells against tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via caveolins. Anesthesiology. 2011;115(3):499–508.CrossRef Kawaraguchi Y, Horikawa YT, Murphy AN, Murray F, Miyanohara A, Ali SS, Head BP, Patel PM, Roth DM, Patel HH. Volatile anesthetics protect cancer cells against tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via caveolins. Anesthesiology. 2011;115(3):499–508.CrossRef
10.
Zurück zum Zitat Niwa H, Rowbotham DJ, Lambert DG, Buggy DJ. Can anesthetic techniques or drugs affect cancer recurrence in patients undergoing cancer surgery? J Anesth. 2013;27(5):731–41.CrossRef Niwa H, Rowbotham DJ, Lambert DG, Buggy DJ. Can anesthetic techniques or drugs affect cancer recurrence in patients undergoing cancer surgery? J Anesth. 2013;27(5):731–41.CrossRef
11.
Zurück zum Zitat Kvolik S, Glavas-Obrovac L, Bares V, Karner I. Effects of inhalation anesthetics halothane, sevoflurane, and isoflurane on human cell lines. Life Sci. 2005;77(19):2369–83.CrossRef Kvolik S, Glavas-Obrovac L, Bares V, Karner I. Effects of inhalation anesthetics halothane, sevoflurane, and isoflurane on human cell lines. Life Sci. 2005;77(19):2369–83.CrossRef
12.
Zurück zum Zitat Kvolik S, Dobrosevic B, Marczi S, Prlic L, Glavas-Obrovac L. Different apoptosis ratios and gene expressions in two human cell lines after sevoflurane anaesthesia. Acta Anaesthesiol Scand. 2009;53(9):1192–9.CrossRef Kvolik S, Dobrosevic B, Marczi S, Prlic L, Glavas-Obrovac L. Different apoptosis ratios and gene expressions in two human cell lines after sevoflurane anaesthesia. Acta Anaesthesiol Scand. 2009;53(9):1192–9.CrossRef
13.
Zurück zum Zitat Muller-Edenborn B, Roth-Z'graggen B, Bartnicka K, Borgeat A, Hoos A, Borsig L, Beck-Schimmer B. Volatile anesthetics reduce invasion of colorectal cancer cells through down-regulation of matrix metalloproteinase-9. Anesthesiology. 2012;117(2):293–301.CrossRef Muller-Edenborn B, Roth-Z'graggen B, Bartnicka K, Borgeat A, Hoos A, Borsig L, Beck-Schimmer B. Volatile anesthetics reduce invasion of colorectal cancer cells through down-regulation of matrix metalloproteinase-9. Anesthesiology. 2012;117(2):293–301.CrossRef
14.
Zurück zum Zitat Liang H, Gu M, Yang C, Wang H, Wen X, Zhou Q. Sevoflurane inhibits invasion and migration of lung cancer cells by inactivating the p38 MAPK signaling pathway. J Anesth. 2012;26(3):381–92.CrossRef Liang H, Gu M, Yang C, Wang H, Wen X, Zhou Q. Sevoflurane inhibits invasion and migration of lung cancer cells by inactivating the p38 MAPK signaling pathway. J Anesth. 2012;26(3):381–92.CrossRef
15.
Zurück zum Zitat Liu S, Fang F, Song R, Gao X, Jiang M, Cang J. Sevoflurane affects neurogenesis through cell cycle arrest via inhibiting wnt/beta-catenin signaling pathway in mouse neural stem cells. Life Sci. 2018;209:34–42.CrossRef Liu S, Fang F, Song R, Gao X, Jiang M, Cang J. Sevoflurane affects neurogenesis through cell cycle arrest via inhibiting wnt/beta-catenin signaling pathway in mouse neural stem cells. Life Sci. 2018;209:34–42.CrossRef
16.
Zurück zum Zitat Loop T, Dovi-Akue D, Frick M, Roesslein M, Egger L, Humar M, Hoetzel A, Schmidt R, Borner C, Pahl HL, Geiger KK, Pannen BH. Volatile anesthetics induce caspase-dependent, mitochondria-mediated apoptosis in human T lymphocytes in vitro. Anesthesiology. 2005;102(6):1147–57.CrossRef Loop T, Dovi-Akue D, Frick M, Roesslein M, Egger L, Humar M, Hoetzel A, Schmidt R, Borner C, Pahl HL, Geiger KK, Pannen BH. Volatile anesthetics induce caspase-dependent, mitochondria-mediated apoptosis in human T lymphocytes in vitro. Anesthesiology. 2005;102(6):1147–57.CrossRef
17.
Zurück zum Zitat Dong Y, Zhang G, Zhang B, Moir RD, Xia W, Marcantonio ER, Culley DJ, Crosby G, Tanzi RE, Xie Z. The common inhalational anesthetic sevoflurane induces apoptosis and increases beta-amyloid protein levels. Arch Neurol. 2009;66(5):620–31.CrossRef Dong Y, Zhang G, Zhang B, Moir RD, Xia W, Marcantonio ER, Culley DJ, Crosby G, Tanzi RE, Xie Z. The common inhalational anesthetic sevoflurane induces apoptosis and increases beta-amyloid protein levels. Arch Neurol. 2009;66(5):620–31.CrossRef
18.
Zurück zum Zitat Topouzova-Hristova T, Daza P, Garcia-Herdugo G, Stephanova E. Volatile anaesthetic halothane causes DNA damage in A549 lung cells. Toxicol In Vitro. 2006;20(5):585–93.CrossRef Topouzova-Hristova T, Daza P, Garcia-Herdugo G, Stephanova E. Volatile anaesthetic halothane causes DNA damage in A549 lung cells. Toxicol In Vitro. 2006;20(5):585–93.CrossRef
19.
Zurück zum Zitat Yang Z, Lv J, Li X, Meng Q, Yang Q, Ma W, Li Y, Ke ZJ. Sevoflurane decreases self-renewal capacity and causes c-Jun N-terminal kinase–mediated damage of rat fetal neural stem cells. Sci Rep. 2017;7:46304.CrossRef Yang Z, Lv J, Li X, Meng Q, Yang Q, Ma W, Li Y, Ke ZJ. Sevoflurane decreases self-renewal capacity and causes c-Jun N-terminal kinase–mediated damage of rat fetal neural stem cells. Sci Rep. 2017;7:46304.CrossRef
20.
Zurück zum Zitat Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133(6):1019–31.CrossRef Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133(6):1019–31.CrossRef
21.
Zurück zum Zitat Dhanasekaran DN, Reddy EP. JNK signaling in apoptosis. Oncogene. 2008;27(48):6245–51.CrossRef Dhanasekaran DN, Reddy EP. JNK signaling in apoptosis. Oncogene. 2008;27(48):6245–51.CrossRef
Metadaten
Titel
Differential effects of sevoflurane on the growth and apoptosis of human cancer cell lines
verfasst von
Takahiro Hirai
Yuko Konishi
Shoko Mizuno
Zhou Rui
Yao Sun
Kimitoshi Nishiwaki
Publikationsdatum
31.10.2019
Verlag
Springer Singapore
Erschienen in
Journal of Anesthesia / Ausgabe 1/2020
Print ISSN: 0913-8668
Elektronische ISSN: 1438-8359
DOI
https://doi.org/10.1007/s00540-019-02701-w

Weitere Artikel der Ausgabe 1/2020

Journal of Anesthesia 1/2020 Zur Ausgabe

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Hinter dieser Appendizitis steckte ein Erreger

23.04.2024 Appendizitis Nachrichten

Schmerzen im Unterbauch, aber sonst nicht viel, was auf eine Appendizitis hindeutete: Ein junger Mann hatte Glück, dass trotzdem eine Laparoskopie mit Appendektomie durchgeführt und der Wurmfortsatz histologisch untersucht wurde.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.