Differential role of intravenous anesthetics in colorectal cancer progression: implications for clinical application

Fengliu Deng, Mingwen Ouyang, Xiaofei Wang, Xueqing Yao, Yeming Chen, Tao Tao, Xuegang Sun, Lijun Xu, Jing Tang, Liang Zhao, Fengliu Deng, Mingwen Ouyang, Xiaofei Wang, Xueqing Yao, Yeming Chen, Tao Tao, Xuegang Sun, Lijun Xu, Jing Tang, Liang Zhao

Abstract

Anesthetics are unavoidable to colorectal cancer (CRC) patients who underwent surgical treatment. Thus, the molecular mechanisms underlying the role of the intravenous anesthetics in CRC metastasis are still unclear. In this study, the effects of intravenous anesthetics, such as propofol, etomidate and dexmedetomidine, on cell migration were determined. The migration of CRC cells was inhibited by propofol in vitro, but not in vivo. Etomidate, however, promoted the migration of CRC cells both in vitro and in vivo. Epithelial-mesenchymal transition (EMT) mediated the promotive effect of propofol and etomidate on the migration of CRC cells through PI3K/AKT signaling pathway. Dexmedetomidine alone or in combination with propofol or etomidate had minor effect on the migration of CRC cells. These findings indicate that propofol inhibites CRC cell migration in vitro. Etomidate playes a role for prompting CRC metastasis progression by activating (PI3K)/AKT signaling and inducing EMT. It provides an important hint for the clinical application of these anesthetics.

Keywords: colorectal cancer; dexmedetomidine; epithelial-mesenchymal transition; etomidate; propofol.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors have declared that no conflicts of interest exists.

Figures

Figure 1. Chemical structure of propofol (A),…
Figure 1. Chemical structure of propofol (A), etomidate (B) and dexmedetomidine (C)
Figure 2. The effects of propofol, etomidate…
Figure 2. The effects of propofol, etomidate and dexmedetomidine on the migration ability of CRC cells
(A) Representative figures and data of transwell assay for SW480 and HCT116 cells treated with 2 and 4 μg/ml of propofol for 24 h. (B) Representative figures and data of transwell assay for SW480 and HCT116 cells treated with 2 and 4 μg/ml of etomidate for 24 h. (C) Representative figures and data of transwell assay for SW480 and HCT116 cells treated with 100 and 200 μg/ml of dexmedetomidine for 24 h. Each bar represented the mean ± SD. The results were reproduced in three independent experiments. The asterisk (*) indicates P < 0.05. The asterisk (**) indicates P < 0.01.
Figure 3. Propofol decreases and etomidate increases…
Figure 3. Propofol decreases and etomidate increases the migration ability of CRC cells
(A) Representative figures and data of transwell assay for SW480 and HCT116 cells treated with 100 μg/ml of dexmedetomidine in combination with 2 or 4 μg/ml of propofol for 24 h. (B) Representative figures and data of transwell assay for SW480 and HCT116 cells treated with 100 μg/ml of dexmedetomidine in combined with 2 or 4 μg/ml of etomidate for 24 h. Each bar represented the mean ± SD. The results were reproduced in three independent experiments. The asterisk (*) indicates P < 0.05. The asterisk (**) indicates P < 0.01.
Figure 4. Propofol decreases and etomidate increases…
Figure 4. Propofol decreases and etomidate increases AKT activation and epithelial-mesenchymal transition (EMT)
(A) Western blotting analysis of EMT markers and phosphorylated AKT in SW480 and HCT116 cells treated with 100 μg/ml of dexmedetomidine in combined with 2 or 4 μg/ml of propofol for 24 h. (B) Western blotting analysis of EMT markers and phosphorylated AKT in SW480 and HCT116 cells treated with 100 μg/ml of dexmedetomidine in combined with 2 or 4 μg/ml of etomidate for 24 h. Representative figures were shown. The results were reproduced in 3 independent experiments.
Figure 5. AKT activation is responsible for…
Figure 5. AKT activation is responsible for the promotive effect of etomidate on cell migration
(A) Representative figures and data of transwell assay for indicated cells. Each bar represented the mean ± SD. The results were reproduced in three independent experiments. The asterisk (*) indicates P < 0.05. The asterisk (**) indicates P < 0.01. (B) Western blotting analysis of EMT markers and phosphorylated AKT in indicated cells. Representative figures were shown. The results were reproduced in 3 independent experiments.
Figure 6. Etomidate, but not propofol, promotes…
Figure 6. Etomidate, but not propofol, promotes CRC progression in vivo
Mice were anesthesia with an intraperitoneal injection of propofol 20 mg/kg or etomidate 30 mg/kg. Tumor cells were injected into nude mice through the tail vein to evaluate the lung homing potential of cells. The number of metastatic lung nodules in individual mice was counted under the microscope. The magnification areas indicated metastatic nodes in the lung.

References

    1. Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–117.
    1. Scheer A, Auer RA. Surveillance after curative resection of colorectal cancer. Clin Colon Rectal Surg. 2009;22:242–250.
    1. Young PE, Womeldorph CM, Johnson EK, Maykel JA, Brucher B, Stojadinovic A, Avital I, Nissan A, Steele SR. Early detection of colorectal cancer recurrence in patients undergoing surgery with curative intent: current status and challenges. J Cancer. 2014;5:262–271.
    1. Tang J, Sun Y, Wu WK, Zhong T, Liu Y, Xiao J, Tao T, Zhao Z, Gu M. Propofol lowers serum PF4 level and partially corrects hypercoagulopathy in endotoxemic rats. Biochim Biophys Acta. 2010;1804:1895–1901.
    1. Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology. 2011;114:695–707.
    1. Tang J, Chen X, Tu W, Guo Y, Zhao Z, Xue Q, Lin C, Xiao J, Sun X, Tao T, Gu M, Liu Y. Propofol inhibits the activation of p38 through up-regulating the expression of annexin A1 to exert its anti-inflammation effect. PLoS One. 2011;6:e27890.
    1. McDermott BJ, McWilliams S, Smyth K, Kelso EJ, Spiers JP, Zhao Y, Bell D, Mirakhur RK. Protection of cardiomyocyte function by propofol during simulated ischemia is associated with a direct action to reduce pro-oxidant activity. J Mol Cell Cardiol. 2007;42:600–608.
    1. Tang J, Hu JJ, Lu CH, Liang JN, Xiao JF, Liu YT, Lin CS, Qin ZS. Propofol inhibits lipopolysaccharide-induced tumor necrosis factor-alpha expression and myocardial depression through decreasing the generation of superoxide anion in cardiomyocytes. Oxid Med Cell Longev. 2014;2014:157376.
    1. Lin FY, Tsai YT, Lee CY, Lin CY, Lin YW, Li CY, Shih CM, Huang CY, Chang NC, Tsai JC, Chen TL, Tsai CS. TNF-alpha-decreased thrombomodulin expression in monocytes is inhibited by propofol through regulation of tristetraprolin and human antigen R activities. Shock. 2011;36:279–288.
    1. Hsing CH, Lin MC, Choi PC, Huang WC, Kai JI, Tsai CC, Cheng YL, Hsieh CY, Wang CY, Chang YP, Chen YH, Chen CL, Lin CF. Anesthetic propofol reduces endotoxic inflammation by inhibiting reactive oxygen species-regulated Akt/IKKbeta/NF-kappaB signaling. PLoS One. 2011;6:e17598.
    1. Mammoto T, Mukai M, Mammoto A, Yamanaka Y, Hayashi Y, Mashimo T, Kishi Y, Nakamura H. Intravenous anesthetic, propofol inhibits invasion of cancer cells. Cancer Lett. 2002;184:165–170.
    1. Huang H, Benzonana LL, Zhao H, Watts HR, Perry NJ, Bevan C, Brown R, Ma D. Prostate cancer cell malignancy via modulation of HIF-1alpha pathway with isoflurane and propofol alone and in combination. Br J Cancer. 2014;111:1338–1349.
    1. Zhang D, Zhou XH, Zhang J, Zhou YX, Ying J, Wu GQ, Qian JH. Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer. Biochem Biophys Res Commun. 2015;468:561–567.
    1. Zhang L, Wang N, Zhou S, Ye W, Jing G, Zhang M. Propofol induces proliferation and invasion of gallbladder cancer cells through activation of Nrf2. J Exp Clin Cancer Res. 2012;31:66.
    1. Paris A, Philipp M, Tonner PH, Steinfath M, Lohse M, Scholz J, Hein L. Activation of alpha 2B-adrenoceptors mediates the cardiovascular effects of etomidate. Anesthesiology. 2003;99:889–895.
    1. Devlin JW, Al-Qadheeb NS, Chi A, Roberts RJ, Qawi I, Garpestad E, Hill NS. Efficacy and safety of early dexmedetomidine during noninvasive ventilation for patients with acute respiratory failure: a randomized, double-blind, placebo-controlled pilot study. Chest. 2014;145:1204–1212.
    1. Candiotti KA, Bergese SD, Bokesch PM, Feldman MA, Wisemandle W, Bekker AY. Monitored anesthesia care with dexmedetomidine: a prospective, randomized, double-blind, multicenter trial. Anesth Analg. 2010;110:47–56.
    1. Snyder GL, Greenberg S. Effect of anaesthetic technique and other perioperative factors on cancer recurrence. Br J Anaesth. 2010;105:106–115.
    1. Chen X, Lu P, Chen L, Yang SJ, Shen HY, Yu DD, Zhang XH, Zhong SL, Zhao JH, Tang JH. Perioperative propofol-paravertebral anesthesia decreases the metastasis and progression of breast cancer. Tumour Biol. 2015;36:8259–8266.
    1. Buckley A, McQuaid S, Johnson P, Buggy DJ. Effect of anaesthetic technique on the natural killer cell anti-tumour activity of serum from women undergoing breast cancer surgery: a pilot study. Br J Anaesth. 2014;113:i56–62.
    1. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112:1776–1784.
    1. Iwatsuki M, Mimori K, Yokobori T, Ishi H, Beppu T, Nakamori S, Baba H, Mori M. Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 2010;101:293–299.
    1. Shen L, Qu X, Ma Y, Zheng J, Chu D, Liu B, Li X, Wang M, Xu C, Liu N, Yao L, Zhang J. Tumor suppressor NDRG2 tips the balance of oncogenic TGF-beta via EMT inhibition in colorectal cancer. Oncogenesis. 2014;3:e86.
    1. Takahashi Y, Sawada G, Kurashige J, Uchi R, Matsumura T, Ueo H, Takano Y, Akiyoshi S, Eguchi H, Sudo T, Sugimachi K, Doki Y, Mori M, et al. Paired related homoeobox 1, a new EMT inducer, is involved in metastasis and poor prognosis in colorectal cancer. Br J Cancer. 2013;109:307–311.
    1. Liu S, Zhang XP, Han NN, Lv S, Xiong JY. Pretreatment with low dose etomidate prevents etomidate-induced rat adrenal insufficiency by regulating oxidative stress-related MAPKs and apoptosis. Environmental toxicology and pharmacology. 2015;39:1212–1220.
    1. Campagna-Slater V, Weaver DF. Anaesthetic binding sites for etomidate and propofol on a GABAA receptor model. Neurosci Lett. 2007;418:28–33.
    1. Maldifassi MC, Baur R, Sigel E. Functional sites involved in modulation of the GABAA receptor channel by the intravenous anesthetics propofol, etomidate and pentobarbital. Neuropharmacology. 2016;105:207–214.
    1. Rafael D, Doktorovova S, Florindo HF, Gener P, Abasolo I, Schwartz S, Jr, Videira MA. EMT blockage strategies: Targeting Akt dependent mechanisms for breast cancer metastatic behaviour modulation. Curr Gene Ther. 2015;15:300–312.
    1. Bacigalupo ML, Manzi M, Espelt MV, Gentilini LD, Compagno D, Laderach DJ, Wolfenstein-Todel C, Rabinovich GA, Troncoso MF. Galectin-1 triggers epithelial-mesenchymal transition in human hepatocellular carcinoma cells. J Cell Physiol. 2015;230:1298–1309.
    1. Xu SH, Huang JZ, Xu ML, Yu G, Yin XF, Chen D, Yan GR. ACK1 promotes gastric cancer epithelial-mesenchymal transition and metastasis through AKT-POU2F1-ECD signalling. J Pathol. 2015;236:175–185.
    1. Jiang H, Gao M, Shen Z, Luo B, Li R, Jiang X, Ding R, Ha Y, Wang Z, Jie W. Blocking PI3K/Akt signaling attenuates metastasis of nasopharyngeal carcinoma cells through induction of mesenchymal-epithelial reverting transition. Oncol Rep. 2014;32:559–566.
    1. Goubran HA, Kotb RR, Stakiw J, Emara ME, Burnouf T. Regulation of tumor growth and metastasis: the role of tumor microenvironment. Cancer Growth Metastasis. 2014;7:9–18.
    1. Croci DO, Salatino M. Tumor immune escape mechanisms that operate during metastasis. Curr Pharm Biotechnol. 2011;12:1923–1936.
    1. Smyth MJ, Cretney E, Kershaw MH, Hayakawa Y. Cytokines in cancer immunity and immunotherapy. Immunol Rev. 2004;202:275–293.

Source: PubMed

3
Prenumerera