Prostate cancer cell malignancy via modulation of HIF-1α pathway with isoflurane and propofol alone and in combination

H Huang, L L Benzonana, H Zhao, H R Watts, N J S Perry, C Bevan, R Brown, D Ma, H Huang, L L Benzonana, H Zhao, H R Watts, N J S Perry, C Bevan, R Brown, D Ma

Abstract

Background: Surgery is considered to be the first line treatment for solid tumours. Recently, retrospective studies reported that general anaesthesia was associated with worse long-term cancer-free survival when compared with regional anaesthesia. This has important clinical implications; however, the mechanisms underlying those observations remain unclear. We aim to investigate the effect of anaesthetics isoflurane and propofol on prostate cancer malignancy.

Methods: Prostate cancer (PC3) cell line was exposed to commonly used anaesthetic isoflurane and propofol. Malignant potential was assessed through evaluation of expression level of hypoxia-inducible factor-1α (HIF-1α) and its downstream effectors, cell proliferation and migration as well as development of chemoresistance.

Results: We demonstrated that isoflurane, at a clinically relevant concentration induced upregulation of HIF-1α and its downstream effectors in PC3 cell line. Consequently, cancer cell characteristics associated with malignancy were enhanced, with an increase of proliferation and migration, as well as development of chemoresistance. Inhibition of HIF-1α neosynthesis through upper pathway blocking by a PI-3K-Akt inhibitor or HIF-1α siRNA abolished isoflurane-induced effects. In contrast, the intravenous anaesthetic propofol inhibited HIF-1α activation induced by hypoxia or CoCl2. Propofol also prevented isoflurane-induced HIF-1α activation, and partially reduced cancer cell malignant activities.

Conclusions: Our findings suggest that modulation of HIF-1α activity by anaesthetics may affect cancer recurrence following surgery. If our data were to be extrapolated to the clinical setting, isoflurane but not propofol should be avoided for use in cancer surgery. Further work involving in vivo models and clinical trials is urgently needed to determine the optimal anaesthetic regimen for cancer patients.

Figures

Figure 1
Figure 1
Effect of isoflurane exposure on expression and translocation of HIF-1α in cultured human prostate cancer cells (PC3 cells). Cultured cells were exposed to 0.5–2% for 2 h and were harvested immediately or 2–24 h after exposure for western blotting. (A, B) A time- and concentration-dependent increase in HIF-1α expression after isoflurane exposure; (C) immunostaining further revealed isoflurane exposure promoted the translocation of HIF-1α from cytoplasm to nucleus where its downstream effects are initiated; (D, E) HIF-1β expression was unchanged after exposure to isoflurane. Results are expressed as mean±s.d.; *P<0.05; **P<0.001 vs naïve control. Bar=25 μm.
Figure 2
Figure 2
Propofol abolished increased HIF-1α expression induced by various treatments in human prostate cancer cells. (A) Propofol abolished increased HIF-1α production triggered by hypoxia (less than 1% oxygen for 18 h); (B) propofol antagonised CoCl2-induced upregulation of HIF-1α (CoCl2 at 100 μM for 6 h); propofol in intralipid (C) or DMSO (D) at clinically relevant concentrations (2–4 μg ml−1) effectively prevented isoflurane-induced activated expression of HIF-1α. Results are mean±s.d. (n=4); *P<0.05; **P<0.01 vs naïve control. #P<0.05; ##P<0.01 vs isoflurane or CoCl2 alone.
Figure 3
Figure 3
Isoflurane exposure increases human prostate cancer cell growth-promoting signalling and proliferation markers. Exposure to isoflurane induced activation of markers for angiogenesis (VEGF, (A)) and proliferation (Ki67, (B)), while propofol at 4 μg ml−1 or HIF-1α-specific siRNA (20 nM) abolished these changes. Fluorescent intensity of VEGF (C) and percentage of Ki-67+(D) cells were shown. *P<0.01 vs naïve control; **P=0.001 vs naïve control; ###P<0.001 naive control vs isoflurane alone. Abbreviations: HSi=HIF-1α siRNA; Iso=isoflurane; Pro=propofol; SSi=scrambled siRNA. Bar=50 μm.
Figure 4
Figure 4
Isoflurane exposure enhanced human prostate cancer cell cycle progression and cell proliferation. (A and B) Dual labelling of cyclin D (green) or cyclin E (green) with HIF-1 (red). Exposure to isoflurane induced activation of markers for cell cycle (cyclin D, (A) and cyclin E, (B)), while propofol at 4 μg ml−1 or HIF-1α-specific siRNA (20 nM) abolished these changes. Percentage of cyclin D+ (C) and cyclin E+ cells (D) after treatment. (E) MTT assay (n=6) further indicated that propofol at 4 μg ml−1 effectively blocked isoflurane-induced cell proliferation. Results are mean±s.d. (n=6); *P<0.01 vs naïve control; **P=0.001 vs naïve control; ###P<0.001 navie control vs isoflurane alone. Abbreviations: HSi=HIF-1α siRNA; Iso=isoflurane; Pro=propofol; SSi=scrambled siRNA. Bar=50 μm. The full colour version of this figure is available at British Journal of Cancer online.
Figure 5
Figure 5
Propofol attenuated chemotherapy-resistance development induced by isoflurane exposure via inhibition of HIF-1α in human prostate cancer cells. (A) Exposure to 2% isoflurane induced resistance against docetaxel (an anti-mitotic chemotherapy medication), shown as positive staining for Ki67, a marker for active mitosis (mean±s.d., n=8; **P<0.001 vs docetaxel; ##P<0.001 vs isoflurane+docetaxel); (B) MTT assay also demonstrated that with isoflurane pre-treatment, cells had a higher proliferation rate, which was prevented with propofol (mean±s.d., n=6; **P<0.01 vs docetaxel; #P<0.05 vs isoflurane+docetaxel); (C) cytochrome-c release, as a marker of cellular injury caused by decetaxel treatment, was reduced in isoflurane-exposed cells, while treatment with propofol at 4 μg ml−1 or HIF-1α-specific siRNA (20 nM) restored the sensitivity to Dectaxel treatment; (D) anti-apopotic protein, Bcl-2 was also overexpressed following isoflurane exposure before chemotherapy challenge. All these changes in favour of cell survival were blocked by co-administration of propofol at 4 μg ml−1 or HIF-1α-specific siRNA (20 nM) with isoflurane (n=4; **P<0.001 vs naïve control; ##P<0.001 vs isoflurane alone). Abbreviations: Doc=docetaxel; Iso=isoflurane; Pro=propofol. Bar=50 μm.
Figure 6
Figure 6
Molecular mechanisms of isoflurane (Iso)-induced hypoxia-inducible factor (HIF)-1α expression in human prostate cancer cells. Cultured cells were treated with a PI-3K inhibitor, LY294002 (LY) (50 μM), or an ERK inhibitor, U0126 (U) (50 μM), dissolved in DMSO (D), for 16 h prior to 2% isoflurane, or 2% isoflurane combined with propofol (Pro) (4 μg ml−1) for 2 h and then cell extracts were used for western blotting 24 h after treatment. Other cohort cultured cells were exposed to 2% isoflurane or propofol (Pro) (4 μg ml−1) for 2 h and were then harvested immediately or 2–24 h after exposure. (A) HIF-1α; (B) p-Akt; (C) p-ERK; (D, E) p-Akt vs time course. Results are mean±s.d. (n=5); *P<0.05; **P<0.01 vs naïve control. #P<0.05; ##P<0.01 vs Iso.
Figure 7
Figure 7
Isoflurane exposure enhanced human prostate cancer cell migration and invasion. (A and B) ELISA assessment of MMP-2 and MMP-9 in the cell culture medium. (C and D) Scratch assay of PC3 cells before and 24 h after exposure to isoflurane demonstrated accelerated gap closure, while propofol at 4 μg ml−1 or HIF-1α-specific siRNA (20 nM) abolished these changes. Results are mean±s.d. (n=4); *P<0.01 and ***P=0.001; #P<0.001, naive control vs isoflurane alone. Abbreviations: HSi=HIF-1α siRNA; Iso=isoflurane; NC=naive control; Pro=propofol; SSi=scrambled siRNA.

References

    1. Ban HS, Uno M, Nakamura H. Suppression of hypoxia-induced HIF-1alpha accumulation by VEGFR inhibitors: different profiles of AAL993 versus SU5416 and KRN633. Cancer Lett. 2010;296 (1:17–26.
    1. 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;119:593–605.
    1. Bickler PE, Fahlman CS. The inhaled anesthetic, isoflurane, enhances Ca2+-dependent survival signaling in cortical neurons and modulates MAP kinases, apoptosis proteins and transcription factors during hypoxia. Anesth Analg. 2006;103 (2:419–429.
    1. Bickler PE, Fahlman CS. Enhanced hypoxic preconditioning by isoflurane: signaling gene expression and requirement of intracellular Ca2+ and inositol triphosphate receptors. Brain Res. 2010;1340:86–95.
    1. Biki B, Mascha E, Moriarty D, Fitzpatrick J, Sessler D, Buggy D. Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: a retrospective analysis. Anesthesiology. 2008;109 (2:180–187.
    1. Biki B, Mascha E, Moriarty DC, Fitzpatrick JM, Sessler DI, Buggy DJ. Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: a retrospective analysis. Anesthesiology. 2008;109 (2:180–187.
    1. Bray F, Jemal A, Grey N, Ferlay J, Forman D. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol. 2012;13 (8:790–801.
    1. Burrows N, Babur M, Resch J, Ridsdale S, Mejin M, Rowling EJ, Brabant G, Williams KJ. GDC-0941 inhibits metastatic characteristics of thyroid carcinomas by targeting both the phosphoinositide-3 kinase (PI3K) and hypoxia-inducible factor-1alpha (HIF-1alpha) pathways. J Clin Endocrinol Metab. 2011;96 (12:E1934–E1943.
    1. Christopherson R, James KE, Tableman M, Marshall P, Johnson FE. Long-term survival after colon cancer surgery: a variation associated with choice of anesthesia. Anesth Analg. 2008;107 (1:325–332.
    1. Daijo H, Kai S, Tanaka T, Wakamatsu T, Kishimoto S, Suzuki K, Harada H, Takabuchi S, Adachi T, Fukuda K, Hirota K. Fentanyl activates hypoxia-inducible factor 1 in neuronal SH-SY5Y cells and mice under non-hypoxic conditions in a mu-opioid receptor-dependent manner. Eur J Pharmacol. 2011;667 (1-3:144–152.
    1. de Oliveira GS, Jr, Ahmad S, Schink JC, Singh DK, Fitzgerald PC, McCarthy RJ. Intraoperative neuraxial anesthesia but not postoperative neuraxial analgesia is associated with increased relapse-free survival in ovarian cancer patients after primary cytoreductive surgery. Reg Anesth Pain Med. 2011;36 (3:271–277.
    1. Deegan CA, Murray D, Doran P, Ecimovic P, Moriarty DC, Buggy DJ. Effect of anaesthetic technique on oestrogen receptor-negative breast cancer cell function in vitro. Br J Anaesth. 2009;103 (5:685–690.
    1. Eisinger-Mathason TS, Simon MC. HIF-1alpha partners with FoxA2, a neuroendocrine-specific transcription factor, to promote tumorigenesis. Cancer Cell. 2010;18 (1:3–4.
    1. Exadaktylos AK, Buggy DJ, Moriarty DC, Mascha E, Sessler DI. Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis. Anesthesiology. 2006;105 (4:660–664.
    1. Fujioka N, Nguyen J, Chen C, Li Y, Pasrija T, Niehans G, Johnson KN, Gupta V, Kratzke RA, Gupta K. Morphine-induced epidermal growth factor pathway activation in non-small cell lung cancer. Anesth Analg. 2011;113 (6:1353–1364.
    1. Garcia PS, Kolesky SE, Jenkins A. General anesthetic actions on GABA(A) receptors. Curr Neuropharmacol. 2010;8 (1:2–9.
    1. Gopinathan L, Tan SL, Padmakumar VC, Coppola V, Tessarollo L, Kaldis P. Loss of Cdk2 and cyclin A2 impairs cell proliferation and tumorigenesis. Cancer Res. 2014;74 (14:3870–3879.
    1. Gottschalk A, Brodner G, Van Aken HK, Ellger B, Althaus S, Schulze HJ. Can regional anaesthesia for lymph-node dissection improve the prognosis in malignant melanoma. Br J Anaesth. 2012;109 (2:253–259.
    1. Gottschalk A, Ford JG, Regelin CC, You J, Mascha EJ, Sessler DI, Durieux ME, Nemergut EC. Association between epidural analgesia and cancer recurrence after colorectal cancer surgery. Anesthesiology. 2010;113 (1:27–34.
    1. Greijer AE, de Jong MC, Scheffer GL, Shvarts A, van Diest PJ, van der Wall E. Hypoxia-induced acidification causes mitoxantrone resistance not mediated by drug transporters in human breast cancer cells. Cell Oncol. 2005;27 (1:43–49.
    1. Gupta K, Kshirsagar S, Chang L, Schwartz R, Law PY, Yee D, Hebbel RP. Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth. Cancer Res. 2002;62 (15:4491–4498.
    1. Heaney A, Buggy DJ. Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis. Br J Anaesth. 2012;109 (Suppl 1:i17–i28.
    1. Hu J, Verkman AS. Increased migration and metastatic potential of tumor cells expressing aquaporin water channels. FASEB J. 2006;20 (11:1892–1894.
    1. Jiang BH, Jiang G, Zheng JZ, Lu Z, Hunter T, Vogt PK. Phosphatidylinositol 3-kinase signaling controls levels of hypoxia-inducible factor 1. Cell Growth Differ. 2001;12 (7:363–369.
    1. Kushida A, Inada T, Shingu K. Enhancement of antitumor immunity after propofol treatment in mice. Immunopharmacol Immunotoxicol. 2007;29 (3-4:477–486.
    1. Li QF, Wang XR, Yang YW, Su DS. Up-regulation of hypoxia inducible factor 1alpha by isoflurane in Hep3B cells. Anesthesiology. 2006;105 (6:1211–1219.
    1. Liang CC, Park AY, Guan JL. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2007;2 (2:329–333.
    1. Liao D, Johnson RS. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007;26 (2:281–290.
    1. Lin L, Liu C, Tan H, Ouyang H, Zhang Y, Zeng W. Anaesthetic technique may affect prognosis for ovarian serous adenocarcinoma: a retrospective analysis. Br J Anaesth. 2011;106 (6:814–822.
    1. Ma D, Lim T, Xu J, Tang H, Wan Y, Zhao H, Hossain M, Maxwell PH, Maze M. Xenon preconditioning protects against renal ischemic-reperfusion injury via HIF-1alpha activation. J Am Soc Nephrol. 2009;20 (4:713–720.
    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 (2:165–170.
    1. Monti E, Gariboldi MB. HIF-1 as a target for cancer chemotherapy, chemosensitization and chemoprevention. Curr Mol Pharmacol. 2011;4 (1:62–77.
    1. Ren XF, Li WZ, Meng FY, Lin CF. Differential effects of propofol and isoflurane on the activation of T-helper cells in lung cancer patients. Anaesthesia. 2010;65 (5:478–482.
    1. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3 (10:721–732.
    1. Semenza G. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene. 2010;29 (5:625–634.
    1. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3 (10:721–732.
    1. Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 2012;33 (4:207–214.
    1. Shukla S, Maclennan GT, Hartman DJ, Fu P, Resnick MI, Gupta S. Activation of PI3K-Akt signaling pathway promotes prostate cancer cell invasion. Int J Cancer. 2007;121 (7:1424–1432.
    1. Smul TM, Stumpner J, Blomeyer C, Lotz C, Redel A, Lange M, Roewer N, Kehl F. Propofol inhibits desflurane-induced preconditioning in rabbits. J Cardiothorac Vasc Anesth. 2010;25 (2:276–281.
    1. Takabuchi S, Hirota K, Nishi K, Oda S, Oda T, Shingu K, Takabayashi A, Adachi T, Semenza G, Fukuda K. The intravenous anesthetic propofol inhibits hypoxia-inducible factor 1 activity in an oxygen tension-dependent manner. FEBS Lett. 2004;577 (3:434–438.
    1. Takabuchi S, Hirota K, Nishi K, Oda S, Oda T, Shingu K, Takabayashi A, Adachi T, Semenza GL, Fukuda K. The intravenous anesthetic propofol inhibits hypoxia-inducible factor 1 activity in an oxygen tension-dependent manner. FEBS Lett. 2004;577 (3:434–438.
    1. Talks K, Turley H, Gatter K, Maxwell P, Pugh C, Ratcliffe P, Harris A. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol. 2000;157 (2:411–421.
    1. Tanaka T, Takabuchi S, Nishi K, Oda S, Wakamatsu T, Daijo H, Fukuda K, Hirota K. The intravenous anesthetic propofol inhibits lipopolysaccharide-induced hypoxia-inducible factor 1 activation and suppresses the glucose metabolism in macrophages. J Anesth. 2010;24 (1:54–60.
    1. Tavare AN, Perry NJ, Benzonana LL, Takata M, Ma D. Cancer recurrence after surgery: direct and indirect effects of anesthetic agents. Int J Cancer. 2012;130 (6:1237–1250.
    1. Taylor JT, Zeng XB, Pottle JE, Lee K, Wang AR, Yi SG, Scruggs JA, Sikka SS, Li M. Calcium signaling and T-type calcium channels in cancer cell cycling. World J Gastroenterol. 2008;14 (32:4984–4991.
    1. Terraneo L, Bianciardi P, Caretti A, Ronchi R, Samaja M. Chronic systemic hypoxia promotes LNCaP prostate cancer growth in vivo. Prostate. 2010;70 (11:1243–1254.
    1. Tsuda K. Neuroprotective effect of isoflurane and N-methyl-D-aspartate receptors in ischemic brain injury. Stroke. 2010;41 (10:e578.
    1. Wang H, Bian S, Yang CS. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1alpha. Carcinogenesis. 2011;32 (12:1881–1889.
    1. Wei H. The role of calcium dysregulation in anesthetic-mediated neurotoxicity. Anesth Analg. 2011;113 (5:972–974.
    1. Wei H, Inan S. Dual effects of neuroprotection and neurotoxicity by general anesthetics: role of intracellular calcium homeostasis. Prog Neuropsychopharmacol Biol Psychiatry. 2013;47:156–161.
    1. Welden B, Gates G, Mallari R, Garrett N. Effects of anesthetics and analgesics on natural killer cell activity. AANA J. 2009;77 (4:287–292.
    1. Xin WK, Kwan CL, Zhao XH, Xu J, Ellen RP, McCulloch CA, Yu XM. A functional interaction of sodium and calcium in the regulation of NMDA receptor activity by remote NMDA receptors. J Neurosci. 2005;25 (1:139–148.
    1. Yeh CH, Cho W, So EC, Chu CC, Lin MC, Wang JJ, Hsing CH. Propofol inhibits lipopolysaccharide-induced lung epithelial cell injury by reducing hypoxia-inducible factor-1alpha expression. Br J Anaesth. 2011;106 (4:590–599.
    1. Zhang Y, Zhen Y, Dong Y, Xu Z, Yue Y, Golde TE, Tanzi RE, Moir RD, Xie Z. Anesthetic propofol attenuates the isoflurane-induced caspase-3 activation and Abeta oligomerization. PLoS One. 2011;6 (11:e27019.
    1. Zuo Z. Are volatile anesthetics neuroprotective or neurotoxic. Med Gas Res. 2012;2 (1:10.

Source: PubMed

3
Sottoscrivi