Open Access

Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma

  • Authors:
    • Min Zhou
    • Junchao Dai
    • Yu Zhou
    • Jian Wu
    • Tao Xu
    • Denglian Zhou
    • Xiaobin Wang
  • View Affiliations

  • Published online on: May 8, 2018     https://doi.org/10.3892/etm.2018.6140
  • Pages: 83-92
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Postoperative immunosuppression is associated with the recurrence and metastasis of esophageal squamous cell carcinoma (ESCC). Propofol is a commonly used intravenous anesthetic and has been reported to be associated with immunosuppression; however, little is known about its effect on innate immune cells during the postoperative period in patients with ESCC. The aim of the present study was to investigate the effects of propofol on the phenotype and cytotoxicity of natural killer (NK) cells derived from the peripheral blood of patients with ESCC. The percentage, phenotype and function of NK cells were compared between patients with ESCC and healthy volunteers using flow cytometry. NK cells were negatively sorted using magnetic beads and cocultured with propofol to assess changes in phenotype and function. The results revealed that the percentage of NK cells was significantly increased in the peripheral blood of patients with ESCC, while their activity and cytotoxicity were impaired. NK cells were successfully separated from peripheral blood in vitro and it was demonstrated that propofol enhanced their activity by influencing the expression of activating or inhibitory receptors. Furthermore, propofol was able to increase the cytotoxicity of NK cells from the peripheral blood of patients with ESCC. These results suggest that propofol is able to improve the function of NK cells in patients with ESCC and may therefore be an appropriate anesthetic for ESCC surgery.

Introduction

Esophageal carcinoma is one of the most common digestive tract-derived malignancies worldwide (1,2). Esophageal carcinoma is comprised of squamous carcinoma and adenocarcinoma (3). A high incidence of esophageal squamous carcinoma (ESCC) has been reported in China (4); in 2015, 429,200 cases of cancer were reported in China, with 281,400 mortalities (5). Of these cases, 259,000 were ESCC, making it the fifth most prevalent malignant tumor (6). Tumor recurrence and metastasis are leading causes of mortality in patients with ESCC (7). It has previously been reported that immunosuppression is associated with tumor recurrence and metastasis (8,9), however the underlying mechanism remains to be elucidated.

It is well established that immunosuppression is associated with the recurrence and metastasis of multiple tumors (10). The immune response of infiltrating lymphocytes in tumor tissues is impaired by certain components in the tumor microenvironment, which leads to tumor recurrence and metastasis (11,12). Furthermore, it has been reported that restoration of the immune system may improve the survival of patients with ESCC (13). It has been demonstrated that various anesthetic agents exert different but important effects on immune cells in patients with tumors (14). It may therefore be beneficial to select an anesthetic that improves immunosuppression in patients with tumors.

Innate immunity is the body's first line of defense against infection, tumors and virus invasion (15). Compromised innate immunity has been reported in a number of tumor types and is associated with the survival of patients with tumors, suggesting a crucial role of innate immunity in the inhibition of tumors (16). Natural killer (NK) cells are an important component of the innate immune system, they are typically cluster of differentiation (CD)3CD56+cells, which and are primarily distributed in the peripheral blood, with 10–15% in lymphocytes (17,18). NK cells are responsible for immune surveillance without antigen presentation and major histocompatibility complex (MHC) restriction (19). Once activated, NK cells are able to recognize target cells rapidly and release cytotoxic effector molecules to trigger the immune response (20). The aim of the present study was to evaluate the effects of propofol on NK cells derived from the peripheral blood of Patients with ESCC. Propofol is one of the most commonly used anesthetics and has been reported to exert analgesic and anti-inflammatory effects (21).

Materials and methods

Patients and tissue sample

A total of 5 ml fresh peripheral blood was collected from 15 patients with ESCC (age, 39–43 years; 10 male, 5 female) prior to surgery and 15 healthy volunteers (age, 26–35 years; 11 male, 4 female) at the Affiliated Hospital of Southwest Medical University (Luzhou, China) between January and May 2017. All patients were diagnosed with primary ESCC and had not undergone therapy. The exclusion criteria for patients with ESCC were as follows: Previous chemotherapy or radiotherapy, chronic disease, infectious disease or multi-primary cancer. Exclusion criteria for the healthy patients included chronic, autoimmune or genetic diseases. Patient clinical data is presented in Table I. The present study was approved by the Ethics Committee of the Southwest Medical University and informed consent was obtained from each patient.

Table I.

Clinical data of the patients with esophageal squamous cell carcinoma.

Table I.

Clinical data of the patients with esophageal squamous cell carcinoma.

IndexNumber of patients
Sex
  Male3
  Female12
Age
  ≤605
  ≥6010
Tumor size
  ≤5 cm9
  ≥5 cm6
Location
  Middle11
  Lower4
Differentiation
  Well8
  Moderate7
  Poor0
Invasion depth
  Outer layer2
  Inner layer13
Lymph node metastases
  With7
  Without8
Cell isolation

Fresh peripheral blood of patients with ESCC was stored in anticoagulant tubes (BD Biosciences, Franklin Lakes, NJ, USA) and used to isolate mononuclear lymphocytes and NK cells as previously described (22). In brief, mononuclear lymphocytes were isolated from 5 ml peripheral blood using Ficoll separation medium (Tianjin Yanyang Biochemical Co., Ltd., Tianjin, China). The cells were centrifuged at 2,000 × g for 20 min at room temperature and the middle layer was mononuclear lymphocytes used for the NK separation. NK cells were negatively sorted using a human NK cell separation medium kit purchased from BD Biosciences according to the manufacturer's protocol. NK cells were negatively sorted using the MagniSort™ Human NK cell Enrichment kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. Isolated NK cell suspensions were then identified using flow cytometry analysis characterized by CD3-CD56+.

Flow cytometry analysis

NK cells were incubated with the appropriate surface antibodies for 15 min at room temperature. Cells were fixed with 4% formaldehyde at room temperature for 10 min and subsequently permeabilized for 40 min using Cytofix/Cytoperm (BD Biosciences) according to the manufacturer's protocol. Permeabilized cells were stained with antibodies against Ki67, granzyme B and perforin for 15 min at room temperature. For intracellular staining of interferon (IFN)γ and tumor necrosis factor (TNF)α, NK cells were stimulated for 4 h at 37°C in 5% CO2 with 50 ng/ml phorbol myristate acetate and 1 mg/ml ionomycin in the presence of Golgistop (BD Biosciences) prior to staining with fluorescence labeling antibodies (Table II) for 15 min at room temperature. A flow cytometer was used for analysis with FAC Suite version 1.0.3.2942 (BD Biosciences).

Table II.

Antibodies used for flow cytometry.

Table II.

Antibodies used for flow cytometry.

AntibodyCompanyCat. no.ConjugateDilution
NKG2DBD Biosciences552364Percp-cy5.51:40
NKp30BD Biosciences558407PE1:40
NKp44BD Biosciences558563PE1:40
NKG2AR&D Systems, Inc.FAB1059CPercp1:40
CD3BD Biosciences555916Fluorescein isothiocyanate1:40
CD56BD Biosciences555518 Allophycocyanin1:40
CD16BD Biosciences557744PE-cy71:40
CD226BD Biosciences559789PE1:40
CD158bBD Biosciences559785PE1:40
NKp46BD Biosciences557991PE1:40
Ki67BD Biosciences561283PE-cy71:40
CD107aBD Biosciences555801PE1:40
Interferon-γBD Biosciences559326PE1:40
Granzyme BBD Biosciences561142PE1:40
PerforinBD Biosciences563762Percp-cy5.51:40
Tumor necrosis factor αBD Biosciences560679Percp-cy5.51:40

[i] BD Biosciences is located in Franklin Lakes, NJ, USA. R&D Systems, Inc. is located in Minneapolis, MN, USA. NK, natural killer; PE, phycoerythrin; Percp, peridinin-chlorophyll-protein; CD, cluster of differentiation.

In vitro coculture of NK cells with propofol

NK cells were separated from the peripheral blood and cultured at 37°C in RPMI 1640 medium with 10% fetal bovine serum (both BD Biosciences) in the presence of 100 U/ml interleukin (IL)2, 100 U/ml IL12 and 100 U/ml IL15 for 48 h. Cells were subsequently incubated in RPMI 1,640 medium with 50 µmol/l propofol (Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) for 24 h at 37°C. The purity of NK cells was >90% as confirmed by flow cytometry. The markers of NK cells (NKG2D, NKp30, NKp44, NKG2A, CD3, CD56, CD16, CD226, CD158b, NKp46, Ki67, CD107a, IFNγ, granzyme B, perforin and TNFα) were detected using a flow cytometer.

NK cells cytotoxicity assay

Following coculture with propofol, the number of NK cells was calculated. NK cells were subsequently mixed with K562 and Eca109 cells (Type Culture Collection of the Chinese Academy of Sciences, Shanghai, China), each at a ratio of 1:5. Apoptosis was evaluated using flow cytometry. NK cells were treated using the fluorescein isothiocyanate Annexin V Apoptosis Detection kit I (BD Biosciences) according to the manufacturer's protocol. Cells positive for Annexin V alone were in early apoptosis, cells positive for propidium iodide (PI) alone were necrotic and cells positive for Annexin V and PI were in late apoptosis. Untreated NK cells from patients with ESCC were used as the control for this experiment.

Statistical analysis

Data are presented as the mean ± standard deviation. Student's t-test was used to compare between groups. P<0.05 was considered to indicate a statistically significant difference.

Results

The phenotype and function of NK cells from the peripheral blood of patients with ESCC

NK cells have been reported to be important for suppressing the recurrence and metastasis of tumor cells. Furthermore, it has been reported that immunosuppression occurs in the tumor microenvironment as well as the peripheral blood (23). The results of flow cytometry revealed that the percentage of NK cells gated by CD3-CD56+ in the peripheral blood of patients with ESCC (21.6±0.89%) was significantly increased compared with the control (11.8±0.54%; Fig. 1). Furthermore, the percentage of NKT cells gated by CD3+CD56+, another type of innate immune cell (24), was significantly increased in the peripheral blood of patients with ESCC compared with the control group (3.99±0.43 vs. 1.15±0.10); Fig. 1), indicating that the innate immune response activity was increased.

NK cell activation is known to be controlled by activating and inhibitory receptors (25). In the present study, the expression of activating and inhibitory receptors was assessed to evaluate the activity and function of NK cells. The results revealed that the expression of activating receptors of CD16, NKG2D, CD226 and p30 were significantly increased in compared with the control group (Fig. 2). However, no significant difference in p44, p46, CD158b, G2A, granzyme B, perforin, IFNγ and TNFα expression was observed (Fig. 3). The cytotoxicity of NK cells to K562 was assessed by detecting the expression of CD107a in NK cells. It was demonstrated that the number of CD107a+ NK cells was significantly increased in patients with ESCC compared with the healthy controls, suggesting that the cytotoxicity of NK cells is reduced in patients with ESCC (Fig. 2). Ki67+ cells were also counted to evaluate the proliferation potential of NK cells. No significant difference in proliferation was observed between groups (Fig. 3). These data suggest that the function of NK cells in the peripheral blood of patients with ESCC is impaired compared with those from healthy subjects.

Purification and identification of NK cells

To further investigate the potential influence of propofol on NK cells in ESCC, NK cells were negatively isolated using magnetic beads sorting (26). CD3-CD56+ cells associated with the lymphocyte gate by forward scatter (FSC) and side scatter (SSC) on a dot-plot were defined as NK cells. The results demonstrated that the number of sorted NK cells from the peripheral blood of patients with ESCC or healthy volunteers was 3.5±0.25×105 or 2.7±0.11×105, respectively, while the purity of NK cells was >90% and the percentage of CD3+ T cells was <1%, suggesting that the sorting of NK cells was successful and suitable for subsequent experiments (Fig. 4).

Surface markers of NK cells as detected by flow cytometry

To investigate the impact of propofol on NK cells, the phenotype of isolated NK cells cocultured with propofol was assessed. A concentration of 50 µmol/l propofol was selected as this has been reported to be an appropriate concentration for studying its function (27). The expression of activating and inhibitory receptors on the surface of NK cells was determined using flow cytometry. The results revealed that the percentage of several activating receptors was significantly increased in NK cells cocultured with propofol compared with the control group, including CD16, NKp30, NKp44 and NKG2D (Fig. 5). Conversely, the expression of inhibitory receptors CD158b and NKG2A was significantly decreased in NK cells treated with propofol compared with the control group (Fig. 5). No significant differences in NKp44, NKp46 and CD226 expression was observed (data not shown). These data suggest that propofol may enhance the activity of NK cells in ESCC by regulating the equilibrium between activating and inhibitory receptors.

Propofol enhances the expression of granzyme B and IFN-γin NK cells

It is well established that activated NK cells destroy tumor cells mainly via cytotoxicity effector molecules, including granzyme B, perforin, IFN-γ and TNF-α (28). The results of flow cytometry revealed that the expression of granzyme B and IFN-γ was significantly increased following incubation with propofol (Fig. 6), suggesting that NK cell cytotoxicity was increased. These data suggest that propofol may promote the cytotoxicity of NK cells from patients with ESCC.

Propofol promotes the proliferation potential of isolated NK cells

The influence of propofol on the proliferation potential of isolated NK cells was assessed by detecting expression of Ki67 through flow cytometry. The results demonstrated that the number of Ki67+ NK cells was significantly higher following propofol stimulation compared with control cells (Fig. 7). These data suggest that propofol may promote the proliferative potential of NK cells in patients with ESCC.

Propofol enhances the function of NK cells in vitro

Based on previous findings, the cytotoxicity of NK cells cocultured with propofol to tumor cells was assessed in vitro using apoptosis analysis. K562 was selected as the target cell line as it does not express MHCI molecules (29). The apoptosis rate of K562 cells cocultured with NK cells stimulated by propofol was significantly higher compared with the control group (Fig. 8). To further investigate the cytotoxicity of NK cells to ESCC cells, the apoptosis rate of Eca109 cells incubated with NK cells was assessed. Consistent with K562, NK cells cultured with propofol exerted a greater cytotoxic effect on Eca109 cells compared with the control (Fig. 8). These data suggest that propofol may enhance the cytotoxicity of NK cells from the peripheral blood of patients with ESCC.

Discussion

Elucidating the effect of anesthesia on immune inhibition during the postoperative period is essential for preventing tumor metastasis and improving the prognosis of patients with ESCC (30). Although anesthetic agents have been demonstrated to affect tumor recurrence and metastasis (31), the impact of anesthetics on anti-tumor immune cells is not well understood. In the present study, NK cells were successfully isolated from the peripheral blood of patients with ESCC and it was confirmed that propofol is able to increase the activity of NK cells by regulating the expression of receptors and cytotoxicity effect molecules. Furthermore, propofol enhances the cytotoxicity of NK cells to ESCC cells in vitro, indicating that it serves as a promotion effector for the postoperative immune system in ESCC.

NK cells are a group of innate lymphoid cells, which serve a crucial role in the inhibition of multiple tumors (32). A novel tumor therapy technique, adoptive immunotherapy, based on NK cells has been investigated in human trials (33). The activation or inhibition of NK cells has been reported to depend on the recognition of altered tumor cells by the activation or inhibition of receptors on the NK cell surface. Therefore, screening factors that influence the expression of NK cell surface receptors is a critical area of immunotherapy for tumors (34). Anesthetics have been reported to be associated with innate immune system regulation, providing a novel perspective that anesthesia was implicated with tumor recurrence and metastasis by regulation of immune response (35). The cytotoxicity of NK cells from patients with breast cancer receiving propofol-remifentanil anesthesia with postoperative ketorolac analgesia was clearly lower compared with those receiving sevoflurane-remifentanil anesthesia with postoperative fentanyl analgesia (36); Inada et al (37) reported that propofol promotes the expression of IFNγ in NK cells by suppressing prostaglandin E2 (37). This suggests that propofol is associated with the regulation of NK cytotoxicity; however, its impact on the expression of activation and inhibitory receptors remains unclear. Consistent with previous studies (38), the percentage of NK cells from patients with ESCC was increased compared with the control, which may be a response to tumorigenesis. The phenotype and cytotoxicity of NK cells was investigated and the results demonstrated that NK cells from patients with ESCC had a higher expression of activating receptors (p30, NKG2D, CD226 and CD16) compared with the control, suggesting that NK cells from the peripheral blood of patients with ESCC were activated. Conversely, it has previously been reported that NK cells patients with tumors had impaired function (39,40). These contradictory results may be because some crucial signaling pathway downstream of activating receptors also serves a role in the regulation of NK cells,.

To further evaluate the effect of propofol on NK cells, isolated NK cells from patients with ESCC were incubated with propofol followed by analysis via flow cytometry. The results revealed that propofol increased the expression of activating receptors (p30, NKG2D, p44, CD16) expression and suppressed inhibitory receptors (CD158b, NKG2A). The cytotoxicity of NK cells from patients with ESCC was also enhanced, as indicated by the increased expression of IFNγ and granzyme B. Ki67 was also upregulated in NK cells stimulated with propofol, indicating that propofol improves the proliferation potential of NK cells. Although data within the present study indicated that propofol probably promoted the activation of NK cells from patients with ESCC, the cytotoxicity of NK cells still needs to be confirmed. Blocking the activating interaction between activating receptors and matched ligands led to impaired NK cell function in the presence of high expression of activating receptors. The cytotoxic effects of NK cells on K562 and Eca109 were investigated and it was revealed that propofol-stimulated NK cells increased apoptosis in K562 and Eca109 cells. Furthermore, a significant increase in CD107a + NK cells, which are characteristic of degranulation, was observed following propofol stimulation. These data suggest that propofol is able to enhance the cytotoxic effects of NK cells from the peripheral blood of patients with ESCC in vitro. In the present study, the number of patients recruited was insufficient to analyze the association between NK cell markers and clinicopathological characteristics, including lymph node metastasis and TNM stage. Furthermore, the function of propofol on NK cells should be verified using an in vivo mouse model in the future.

In conclusion, the results of the present study demonstrated that propofol is able to enhance the function of NK cells from the peripheral blood of patients with ESCC. Based on these results, propofol may have the potential to improve postoperative immunosuppression in patients with ESCC.

Acknowledgements

The authors would like to thank Miss XueMei He and Dr Yang Long (The Affiliated Hospital of South West Medical University, Luzhou, China) for providing valuable technical supports and writing assistance.

Funding

This study was supported by National Natural Science Foundation of China (grant no. 81271478).

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

XW designed the study and wrote the paper. MZ and JCD performed the experiments. YZ, JW, TX and DZ collected the samples and performed flow cytometry. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Written informed consent was obtained from all participants in the present study. The study was approved by the Ethics Review Board at Southwest Medical University (Luzhou, China).

Consent for publication

All patients provided written informed consent for the publication of their data.

Competing interests

The authors declare that there were no competing interests.

References

1 

Lin C, Zhang N, Wang Y, Wang Y, Nice E, Guo C, Zhang E, Yu L, Li M, Liu C, et al: Functional role of a novel long noncoding RNA TTN-AS1 in esophageal squamous cell carcinoma progression and metastasis. Clin Cancer Res. 24:486–498. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Zhou X, Qiu GQ, Bao WA and Zhang DH: The prognostic role of nutrition risk score (NRS) in patients with metastatic or recurrent esophageal squamous cell carcinoma (ESCC). Oncotarget. 8:77465–77473. 2017.PubMed/NCBI

3 

Kurimoto K, Hayashi M, Guerrero-Preston R, Koike M, Kanda M, Hirabayashi S, Tanabe H, Takano N, Iwata N, Niwa Y, et al: PAX5 gene as a novel methylation marker that predicts both clinical outcome and cisplatin sensitivity in esophageal squamous cell carcinoma. Epigenetics. 12:865–874. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Wu X, Dinglin X, Wang X, Luo W, Shen Q, Li Y, Gu L, Zhou Q, Zhu H, Li Y, et al: Long noncoding RNA XIST promotes malignancies of esophageal squamous cell carcinoma via regulation of miR-101/EZH2. Oncotarget. 8:76015–76028. 2017.PubMed/NCBI

5 

Cai W, Lu JJ, Xu R, Xin P, Xin J, Chen Y, Gao B, Chen J and Yang X: Survival based radiographic-grouping for esophageal squamous cell carcinoma may impact clinical T stage. Oncotarget. 9:9512–9530. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Zhang Y, Feng Z, Wang W, Dong J, Gong X, Pu H and Chen X: Expression of heat shock protein-27 (Hsp27) and P38MAPK in esophageal squamous cell carcinoma. Med Sci Monit. 23:5246–5253. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Kunizaki M, Hamasaki K, Wakata K, Tobinaga S, Sumida Y, Hidaka S, Yasutake T, Miyazaki T, Matsumoto K, Yamasaki T, et al: Clinical value of serum p53 antibody in the diagnosis and prognosis of esophageal squamous cell carcinoma. Anticancer Res. 38:1807–1813. 2018.PubMed/NCBI

8 

Chen K, Zhu Z, Zhang N, Cheng G, Zhang F, Jin J, Wu J, Ying L, Mao W and Su D: Tumor-infiltrating CD4+ lymphocytes predict a favorable survival in patients with operable esophageal squamous cell carcinoma. Med Sci Monit. 23:4619–4632. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Jesinghaus M, Steiger K, Slotta-Huspenina J, Drecoll E, Pfarr N, Meyer P, Konukiewitz B, Bettstetter M, Wieczorek K, Ott K, et al: Increased intraepithelial CD3+ T-lymphocytes and high PD-L1 expression on tumor cells are associated with a favorable prognosis in esophageal squamous cell carcinoma and allow prognostic immunogenic subgrouping. Oncotarget. 8:46756–46768. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Yankuzo HM, Baraya YS, Mustapha Z, Wong KK and Yaacob NS: Immunomodulatory effects of a bioactive fraction of Strobilanthes crispus in NMU-induced rat mammary tumor model. J Ethnopharmacol. 213:31–37. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Zhuang PY, Zhang KW, Wang JD, Zhou XP, Liu YB, Quan ZW and Shen J: Effect of TALEN-mediated IL-6 knockout on cell proliferation, apoptosis, invasion and anti-cancer therapy in hepatocellular carcinoma (HCC-LM3) cells. Oncotarget. 8:77915–77927. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Maj T, Wang W, Crespo J, Zhang H, Wang W, Wei S, Zhao L, Vatan L, Shao I, Szeliga W, et al: Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol. 18:1332–1341. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Wang L, Wang C, Wang J, Huang X and Cheng Y: A novel systemic immune-inflammation index predicts survival and quality of life of patients after curative resection for esophageal squamous cell carcinoma. J Cancer Res Clin Oncol. 143:2077–2086. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Pérez-González O, Cuéllar-Guzmán LF, Soliz J and Cata JP: Impact of regional anesthesia on recurrence, metastasis, and immune response in breast cancer surgery: A Systematic review of the literature. Reg Anesth Pain Med. 42:751–756. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Eroglu T, Bozkurt M, Kapi E, Selcuk CT, Kuvat SV, Tufek A, Isik FB, Bozarslan BH, Firat U and Satici O: A study on the effects of the use of propofol in experimental model inferior epigastric island flap on ischemia-reperfusion injury. J Craniofac Surg. 28:2193–2198. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Kim R, Kawai A, Wakisaka M, Funaoka Y, Ohtani S, Ito M, Kadoya T and Okada M: Differences in immune response to anesthetics used for day surgery versus hospitalization surgery for breast cancer patients. Clin Transl Med. 6:342017. View Article : Google Scholar : PubMed/NCBI

17 

Javaheri A, Wang AR, Prak Luning E, Lal P, Goldberg LR and Kamoun M: Fatal accelerated rejection with a prominent natural killer cell infiltrate in a heart transplant recipient with peripartum cardiomyopathy. Transpl Immunol. 47:49–54. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Pollmann J, Götz JJ, Rupp D, Strauss O, Granzin M, Grünvogel O, Mutz P, Kramer C, Lasitschka F, Lohmann V, et al: Hepatitis C virus-induced natural killer cell proliferation involves monocyte-derived cells and the OX40/OX40L axis. J Hepatol. 68:421–430. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Baltner K, Kübler A, Pal M, Balvociute M, Mezger M, Handgretinger R and André MC: Expression of KIR2DS1 does not significantly contribute to NK cell cytotoxicity in HLA-C1/C2 heterozygous haplotype B donors. Int Immunol. 29:423–429. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Dansako H, Imai H, Ueda Y, Satoh S, Wakita T and Kato N: ULBP1 is induced by hepatitis C virus infection and is the target of the NK cell-mediated innate immune response in human hepatocytes. FEBS Open Bio. 8:361–371. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Yang Z, Cheng F, Yan G, Xiong L and Liu H: Propofol protects against endotoxin-induced myocardial injury by inhibiting NF-κB-mediated inflammation. Exp Ther Med. 15:2032–2036. 2018.PubMed/NCBI

22 

Klöß S, Oberschmidt O, Morgan M, Dahlke J, Arseniev L, Huppert V, Granzin M, Gardlowski T, Matthies N, Soltenborn S, et al: Optimization of human NK cell manufacturing: Fully automated separation, improved ex vivo expansion using IL-21 with autologous feeder cells, and generation of anti-CD123-CAR-expressing effector cells. Hum Gene Ther. 28:897–913. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Voutsadakis IA: Expression and function of immune ligand-receptor pairs in NK cells and cancer stem cells: Therapeutic implications. Cell Oncol (Dordr). 41:107–121. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Cameron G and Godfrey DI: Differential surface phenotype and context-dependent reactivity of functionally diverse NKT cells. Immunol Cell Biol. Mar 5–2018.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

25 

Nham T, Poznanski SM, Fan IY, Shenouda MM, Chew MV, Lee AJ, Vahedi F, Karimi Y, Butcher M, Lee DA, et al: Ex vivo-expanded NK cells from blood and ascites of ovarian cancer patients are cytotoxic against autologous primary ovarian cancer cells. Cancer Immunol Immunother. 67:575–587. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Nakashima Y, Deie M, Yanada S, Sharman P and Ochi M: Magnetically labeled human natural killer cells, accumulated in vitro by an external magnetic force, are effective against HOS osteosarcoma cells. Int J Oncol. 27:965–971. 2005.PubMed/NCBI

27 

Liu S, Gu X, Zhu L, Wu G, Zhou H, Song Y and Wu C: Effects of propofol and sevoflurane on perioperative immune response in patients undergoing laparoscopic radical hysterectomy for cervical cancer. Medicine (Baltimore). 95:e54792016. View Article : Google Scholar : PubMed/NCBI

28 

Glasner A, Isaacson B, Viukov S, Neuman T, Friedman N, Mandelboim M, Sexl V, Hanna JH and Mandelboim O: Increased NK cell immunity in a transgenic mouse model of NKp46 overexpression. Sci Rep. 7:130902017. View Article : Google Scholar : PubMed/NCBI

29 

Erokhina SA, Streltsova MA, Kanevskiy LM, Telford WG, Sapozhnikov AM and Kovalenko EI: HLA-DR+ NK cells are mostly characterized by less mature phenotype and high functional activity. Immunol Cell Biol. 96:212–228. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Matsota P, Kostopanagiotou G, Kalimeris K, Pandazi A, Kotsaki A, Kontogiannopoulou S and Giamarellos-Bourboulis EJ: Transient effects of anesthesia on leukocyte apoptosis and monocyte cytokine stimulation: A clinical study. Immunol Invest. 1–8. 2018.

31 

Rossaint J and Zarbock A: Perioperative inflammation and its modulation by anesthetics. Anesth Analg. 126:1058–1067. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Acuff NV, Li X, Latha K, Nagy T and Watford WT: Tpl2 promotes innate cell recruitment and effector T cell differentiation to limit citrobacter rodentium burden and dissemination. Infect Immun. 85:pii: e00193. –17. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Li L, Li W, Wang C, Yan X, Wang Y, Niu C, Zhang X, Li M, Tian H, Yao C, et al: Adoptive transfer of natural killer cells in combination with chemotherapy improves outcomes of patients with locally advanced colon carcinoma. Cytotherapy. 20:134–148. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Gumbleton M, Sudan R, Fernandes S, Engelman RW, Russo CM, Chisholm JD and Kerr WG: Dual enhancement of T and NK cell function by pulsatile inhibition of SHIP1 improves antitumor immunity and survival. Sci Signal. 10:pii: eaam5353. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Sedghi S, Kutscher HL, Davidson BA and Knight PR: Volatile anesthetics and immunity. Immunol Invest. 46:793–804. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Cho JS, Lee MH, Kim SI, Park S, Park HS, Oh E, Lee JH and Koo BN: The effects of perioperative anesthesia and analgesia on immune function in patients undergoing breast cancer resection: A prospective randomized stud. Int J Med Sci. 14:970–976. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Inada T, Hirota K and Shingu K: Intravenous anesthetic propofol suppresses prostaglandin E2 and cysteinyl leukotriene production and reduces edema formation in arachidonic acid-induced ear inflammation. J Immunotoxicol. 12:261–265. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Dai K, Huang Y, Chen Z, Sun X, Yang L and Jiang Y: Kbtbd2 inhibits the cytotoxic activity of immortalized NK cells through down-regulating mTOR signaling in a mouse hepatocellular carcinoma model. Eur J Immunol. Jan 13–2018.(Epub ahead of print). View Article : Google Scholar

39 

Hoshikawa M, Aoki T, Matsushita H, Karasaki T, Hosoi A, Odaira K, Fujieda N, Kobayashi Y, Kambara K, Ohara O, et al: NK cell and IFN signatures are positive prognostic biomarkers for resectable pancreatic cancer. Biochem Biophys Res Commun. 495:2058–2065. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Stangl S, Tontcheva N, Sievert W, Shevtsov M, Niu M, Schmid TE, Pigorsch S, Combs SE, Haller B, Balermpas P, et al: Heat shock protein 70 and tumor-infiltrating NK cells as prognostic indicators for patients with squamous cell carcinoma of the head and neck after radiochemotherapy: A multicentre retrospective study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer. 142:1911–1925. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 16 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou M, Dai J, Zhou Y, Wu J, Xu T, Zhou D and Wang X: Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma. Exp Ther Med 16: 83-92, 2018
APA
Zhou, M., Dai, J., Zhou, Y., Wu, J., Xu, T., Zhou, D., & Wang, X. (2018). Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma. Experimental and Therapeutic Medicine, 16, 83-92. https://doi.org/10.3892/etm.2018.6140
MLA
Zhou, M., Dai, J., Zhou, Y., Wu, J., Xu, T., Zhou, D., Wang, X."Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma". Experimental and Therapeutic Medicine 16.1 (2018): 83-92.
Chicago
Zhou, M., Dai, J., Zhou, Y., Wu, J., Xu, T., Zhou, D., Wang, X."Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma". Experimental and Therapeutic Medicine 16, no. 1 (2018): 83-92. https://doi.org/10.3892/etm.2018.6140