The angiotensin II type 1 receptor antagonist telmisartan inhibits cell proliferation and tumor growth of esophageal adenocarcinoma via the AMPKα/mTOR pathway in vitro and in vivo

Shintaro Fujihara, Asahiro Morishita, Kana Ogawa, Tomoko Tadokoro, Taiga Chiyo, Kiyohito Kato, Hideki Kobara, Hirohito Mori, Hisakazu Iwama, Tsutomu Masaki, Shintaro Fujihara, Asahiro Morishita, Kana Ogawa, Tomoko Tadokoro, Taiga Chiyo, Kiyohito Kato, Hideki Kobara, Hirohito Mori, Hisakazu Iwama, Tsutomu Masaki

Abstract

Telmisartan, a widely used antihypertensive drug, is an angiotensin II type 1 (AT1) receptor blocker (ARB). This drug inhibits cancer cell proliferation, but the underlying mechanisms in various cancers, including esophageal cancer, remain unknown. The aim of the present study was to evaluate the effects of telmisartan on human esophageal cancer cell proliferation in vitro and in vivo. We assessed the effects of telmisartan on human esophageal adenocarcinoma (EAC) cells using the cell lines OE19, OE33, and SKGT-4. Telmisartan inhibited the proliferation of these three cell lines via blockade of the G0 to G1 cell cycle transition. This blockade was accompanied by a strong decrease in cyclin D1, cyclin E, and other cell cycle-related proteins. Notably, the AMP-activated protein kinase (AMPK) pathway, a fuel sensor signaling pathway, was enhanced by telmisartan. Compound C, which inhibits the two catalytic subunits of AMPK, enhanced the expression of cyclin E, leading to G0/G1 arrest in human EAC cells. In addition, telmisartan reduced the phosphorylation of epidermal growth factor receptor (p-EGFR) and ERBB2 in vitro. In our in vivo study, intraperitoneal injection of telmisartan led to a 73.2% reduction in tumor growth in mice bearing xenografts derived from OE19 cells. Furthermore, miRNA expression was significantly altered by telmisartan in vitro and in vivo. In conclusion, telmisartan suppressed human EAC cell proliferation and tumor growth by inducing cell cycle arrest via the AMPK/mTOR pathway.

Keywords: AMPKα; angiotensin II type 1 receptor blocker; cell cycle arrest; esophageal adenocarcinoma; telmisartan.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors disclose no potential conflicts of interest.

Figures

Figure 1. The effects of the ARBs…
Figure 1. The effects of the ARBs telmisartan, irbesartan, losartan, and valsartan on the proliferation of EAC cell lines in vitro
Telmisartan suppresses the proliferation of EAC cells. OE19, OE33, and SKGT-4 cells were seeded in 96-well plates. After 24 h, ARB (telmisartan, irbesartan, losartan, and valsartan; 1, 10, and 100 μM) or vehicle was added to the culture medium; 24 h later, the cells were evaluated with CCK-8 assays. Cell viability was assayed daily from 0 to 48 h. The viability of the ARB-treated cells was significantly different from that of the control cells (**, P

Figure 2. The antiproliferative effects of telmisartan…

Figure 2. The antiproliferative effects of telmisartan in EAC cells are mediated via cell cycle…

Figure 2. The antiproliferative effects of telmisartan in EAC cells are mediated via cell cycle arrest
A. Cell cycle analysis of OE19, SKGT-4, and OE33 cells treated with 100 μM telmisartan at 48 h (*, P < 0.05). B. Western blot analysis of PPAR-γ in EAC cells. C. Western blot analysis of cyclin D1, Cdk4, Cdk6, Cdk2, cyclin E, and E2F2 in OE19 and SKGT-4 cells treated with 100 μM telmisartan. D. Western blot analysis of p21Cip1 and p27Kip in OE19 and SKGT-4 cells at 24 and 48 h after the addition of 100 μM telmisartan.

Figure 3. Telmisartan induces cell cycle arrest…

Figure 3. Telmisartan induces cell cycle arrest via activation of the AMPK pathway and suppression…

Figure 3. Telmisartan induces cell cycle arrest via activation of the AMPK pathway and suppression of mTOR signaling
A. Three EAC cell lines were treated with 100 μM telmisartan for the indicated times, and the activation status of AMPKα was assessed by western blotting. B. OE19 cells were treated with 100 μM telmisartan, and the activation status of the LKB1/AMPK/mTOR pathway was determined. C. The antiproliferative effects of telmisartan or the control in combination with various concentrations of compound C were assessed in OE19 cells for 48 h. D. Western blot analysis of cyclin D1 and cyclin E in OE19 cells treated with control, telmisartan alone, compound C alone, or telmisartan combined with compound C for 48 h. n.s., not significant; *, P<0.05.

Figure 4

A. The template indicates the…

Figure 4

A. The template indicates the locations of tyrosine kinase antibodies spotted onto a…
Figure 4
A. The template indicates the locations of tyrosine kinase antibodies spotted onto a human phospho-RTK array. B. Representative expression of various phosphorylated tyrosine kinase receptors in OE19 cells treated with or without 100 μM telmisartan at 24 h. C. Densitometry indicated that the ratios of p-EGFR and ERBB2 spots of telmisartan-treated to untreated cells were 11.6% and 17.5%, respectively. D. Western blot analysis of Akt and p-Akt (Ser473), which are downstream of EGFR signaling, in EAC cells treated with 100 μM telmisartan. E. The antiproliferative effects of telmisartan or the control in combination with various concentrations of MK-2206 were assessed in OE19 cells for 48 h. (D) Western blot analysis of cyclin D1 and cyclin E in OE19 cells treated with the control, telmisartan alone, MK-2206 alone, or telmisartan combined with MK-2206 for 48 h. *,P<0.05.

Figure 5. The growth of OE19-derived tumors…

Figure 5. The growth of OE19-derived tumors in mice treated with telmisartan is inhibited

A.…
Figure 5. The growth of OE19-derived tumors in mice treated with telmisartan is inhibited
A. Representative images of gross OE19 tumors from nude mice treated with vehicle (i) or 50 μg of telmisartan. (ii). B. Histological examination using H and E staining. C. Fibrotic tissue as determined by Azan staining of subcutaneous xenograft tumors 22 days after telmisartan injection. D. Tumor growth curves of the control and telmisartan groups. Tumor volume (mm3) was calculated as (tumor length (mm) × tumor width (mm)2)/2. The tumors were significantly smaller in the telmisartan-treated mice than those in the vehicle-treated mice. Each point represents the mean ± standard deviation of 7 animals. P = 0.0007 by two-way ANOVA.

Figure 6. Schematic model for telmisartan inhibition…

Figure 6. Schematic model for telmisartan inhibition of cell proliferation and G1 cell cycle progression…

Figure 6. Schematic model for telmisartan inhibition of cell proliferation and G1 cell cycle progression in EAC cells
The activation of AMPKα in human EAC cells inhibits mTOR and p70S6K, inducing G1 cell cycle arrest. Telmisartan may affect the cell cycle via the phosphorylation of EGFR and Akt/mTOR.
Figure 2. The antiproliferative effects of telmisartan…
Figure 2. The antiproliferative effects of telmisartan in EAC cells are mediated via cell cycle arrest
A. Cell cycle analysis of OE19, SKGT-4, and OE33 cells treated with 100 μM telmisartan at 48 h (*, P < 0.05). B. Western blot analysis of PPAR-γ in EAC cells. C. Western blot analysis of cyclin D1, Cdk4, Cdk6, Cdk2, cyclin E, and E2F2 in OE19 and SKGT-4 cells treated with 100 μM telmisartan. D. Western blot analysis of p21Cip1 and p27Kip in OE19 and SKGT-4 cells at 24 and 48 h after the addition of 100 μM telmisartan.
Figure 3. Telmisartan induces cell cycle arrest…
Figure 3. Telmisartan induces cell cycle arrest via activation of the AMPK pathway and suppression of mTOR signaling
A. Three EAC cell lines were treated with 100 μM telmisartan for the indicated times, and the activation status of AMPKα was assessed by western blotting. B. OE19 cells were treated with 100 μM telmisartan, and the activation status of the LKB1/AMPK/mTOR pathway was determined. C. The antiproliferative effects of telmisartan or the control in combination with various concentrations of compound C were assessed in OE19 cells for 48 h. D. Western blot analysis of cyclin D1 and cyclin E in OE19 cells treated with control, telmisartan alone, compound C alone, or telmisartan combined with compound C for 48 h. n.s., not significant; *, P<0.05.
Figure 4
Figure 4
A. The template indicates the locations of tyrosine kinase antibodies spotted onto a human phospho-RTK array. B. Representative expression of various phosphorylated tyrosine kinase receptors in OE19 cells treated with or without 100 μM telmisartan at 24 h. C. Densitometry indicated that the ratios of p-EGFR and ERBB2 spots of telmisartan-treated to untreated cells were 11.6% and 17.5%, respectively. D. Western blot analysis of Akt and p-Akt (Ser473), which are downstream of EGFR signaling, in EAC cells treated with 100 μM telmisartan. E. The antiproliferative effects of telmisartan or the control in combination with various concentrations of MK-2206 were assessed in OE19 cells for 48 h. (D) Western blot analysis of cyclin D1 and cyclin E in OE19 cells treated with the control, telmisartan alone, MK-2206 alone, or telmisartan combined with MK-2206 for 48 h. *,P<0.05.
Figure 5. The growth of OE19-derived tumors…
Figure 5. The growth of OE19-derived tumors in mice treated with telmisartan is inhibited
A. Representative images of gross OE19 tumors from nude mice treated with vehicle (i) or 50 μg of telmisartan. (ii). B. Histological examination using H and E staining. C. Fibrotic tissue as determined by Azan staining of subcutaneous xenograft tumors 22 days after telmisartan injection. D. Tumor growth curves of the control and telmisartan groups. Tumor volume (mm3) was calculated as (tumor length (mm) × tumor width (mm)2)/2. The tumors were significantly smaller in the telmisartan-treated mice than those in the vehicle-treated mice. Each point represents the mean ± standard deviation of 7 animals. P = 0.0007 by two-way ANOVA.
Figure 6. Schematic model for telmisartan inhibition…
Figure 6. Schematic model for telmisartan inhibition of cell proliferation and G1 cell cycle progression in EAC cells
The activation of AMPKα in human EAC cells inhibits mTOR and p70S6K, inducing G1 cell cycle arrest. Telmisartan may affect the cell cycle via the phosphorylation of EGFR and Akt/mTOR.

References

    1. Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet. 2013;381:400–12.
    1. Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003;349:2241–2252.
    1. Pennathur A, Farkas A, Krasinskas AM, Ferson PF, Gooding WE, Gibson MK, Schuchert MJ, Landreneau RJ, Luketich JD. Esophagectomy for T1 esophageal cancer: outcomes in 100 patients and implications for endoscopic therapy. Ann Thorac Surg. 2009;87:1048–1054. discussion 54-55.
    1. Edgren G, Adami H-O, Weiderpass E, Nyrén O. A global assessment of the oesophageal adenocarcinoma epidemic. Gut. 2013;62:1406–1414.
    1. Rubenstein JH, Shaheen NJ. Epidemiology, diagnosis, and management of esophageal adenocarcinoma. Gastroenterology. 2015;149:302–317.e1.
    1. Kinoshita J, Fushida S, Harada S, Yagi Y, Fujita H, Kinami S, Ninomiya I, Fujimura T, Kayahara M, Yashiro M, Hirakawa K, Ohta T. Local angiotensin II-generation in human gastric cancer: correlation with tumor progression through the activation of ERK1/2, NF-κB and survivin. Int J Oncol. 2009;34:1573–1582.
    1. Okamoto K, Tajima H, Ohta T, Nakanuma S, Hayashi H, Nakagawara H, Onishi I, Takamura H, Ninomiya I, Kitagawa H, Fushida S, Tani T, Fujimura T, et al. Angiotensin II induces tumor progression and fibrosis in intrahepatic cholangiocarcinoma through an interaction with hepatic stellate cells. Int J Oncol. 2010;37:1251–1259.
    1. Du N, Feng J, Hu L-J, Sun X, Sun H-B, Zhao Y, Yang Y-P, Ren H. Angiotensin II receptor type 1 blockers suppress the cell proliferation effects of angiotensin II in breast cancer cells by inhibiting AT1R signaling. Oncology Rep. 2012;27:1893.
    1. Chen X, Meng Q, Zhao Y, Liu M, Li D, Yang Y, Sun L, Sui G, Cai L, Dong X. Angiotensin II type 1 receptor antagonists inhibit cell proliferation and angiogenesis in breast cancer. Cancer Lett. 2013;328:318–324.
    1. Koyama N, Nishida Y, Ishii T, Yoshida T, Furukawa Y, Narahara H. Telmisartan induces growth inhibition, DNA double-strand breaks and apoptosis in human endometrial cancer cells. PLoS One. 2014;9:e93050.
    1. Okazaki M, Fushida S, Harada S, Tsukada T, Kinoshita J, Oyama K, Tajima H, Ninomiya I, Fujimura T, Ohta T. The angiotensin II type 1 receptor blocker candesartan suppresses proliferation and fibrosis in gastric cancer. Cancer Lett. 2014;355:46–53.
    1. Bhaskaran K, Douglas I, Evans S, van Staa T, Smeeth L. Angiotensin receptor blockers and risk of cancer: cohort study among people receiving antihypertensive drugs in UK general practice research database. BMJ. 2012;344:e2697.
    1. Makar GA, Holmes JH, Yang YX. Angiotensin-converting enzyme inhibitor therapy and colorectal cancer risk. J Natl Cancer Inst. 2014;106:djt374.
    1. Benson SC, Pershadsingh HA, Ho CI, Chittiboyina A, Desai P, Pravenec M, Qi N, Wang J, Avery MA, Kurtz TW. Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPAR -modulating activity. Hypertension. 2004;43:993–1002.
    1. Ishiguro H, Ishiguro Y, Kubota Y, Uemura H. Regulation of prostate cancer cell growth and PSA expression by angiotensin II receptor blocker with peroxisome proliferator-activated receptor gamma ligand like action. Prostate. 2007;67:924–932.
    1. Komatsu Y, Ito I, Wayama M, Fujimura A, Akaogi K, Machida H, Nakajima Y, Kuroda T, Ohmori K, Murayama A, Kimura K, Yanagisawa J. PPARgamma ligands suppress the feedback loop between E2F2 and cyclin-E1. Biochem Biophys Res Commun. 2008;370:145–148.
    1. Sawayama H, Ishimoto T, Watanabe M, Yoshida N, Sugihara H, Kurashige J, Hirashima K, Iwatsuki M, Baba Y, Oki E, Morita M, Shiose Y, Baba H. Small molecule agonists of PPAR- exert therapeutic effects in esophageal cancer. Cancer Res. 2014;74:575–585.
    1. Shimazaki N, Togashi N, Hanai M, Isoyama T, Wada K, Fujita T, Fujiwara K, Kurakata S. Anti-tumour activity of CS-7017, a selective peroxisome proliferator-activated receptor gamma agonist of thiazolidinedione class, in human tumour xenografts and a syngeneic tumour implant model. Eur J Cancer. 2008;44:1734–1743.
    1. Funao K, Matsuyama M, Kawahito Y, Sano H, Chargui J, Touraine J-L, Nakatani T, Yoshimura R. Telmisartan is a potent target for prevention and treatment in human prostate cancer. Oncology Rep. 2008;20:295–300.
    1. Funao K, Matsuyama M, Kawahito Y, Sano H, Chargui J, Touraine J-L, Nakatani T, Yoshimura R. Telmisartan as a peroxisome proliferator-activated receptor-gamma ligand is a new target in the treatment of human renal cell carcinoma. Mol Med Rep. 2009;2:193–198.
    1. Lee L, Mafura B, Lauscher J, Seeliger H, Kreis M, Grone J. Antiproliferative and apoptotic effects of telmisartan in human colon cancer cells. Oncology Lett. 2014;8:2681–2866.
    1. Rehman G, Shehzad A, Khan AL, Hamayun M. Role of AMP-activated protein kinase in cancer therapy. Archiv der Pharmazie. 2014;347:457–468.
    1. Dann SG, Thomas G. The amino acid sensitive TOR pathway from yeast to mammals. FEBS Lett. 2006;580:2821–2829.
    1. Fingar DC, Blenis J. Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene. 2004;23:3151–3171.
    1. Kurokawa H, Sugiyama S, Nozaki T, Sugamura K, Toyama K, Matsubara J, Fujisue K, Ohba K, Maeda H, Konishi M, Akiyama E, Sumida H, Izumiya Y, et al. Telmisartan enhances mitochondrial activity and alters cellular functions in human coronary artery endothelial cells via AMP-activated protein kinase pathway. Atherosclerosis. 2015;239:375–385.
    1. Myojo M, Nagata D, Fujita D, Kiyosue A, Takahashi M, Satonaka H, Morishita Y, Akimoto T, Nagai R, Komuro I, Hirata Y. Telmisartan activates endothelial nitric oxide synthase via Ser1177 phosphorylation in vascular endothelial cells. PLoS One. 2014;9:e96948.
    1. He L-R, Qiao W, Liao Z-X, Komaki R, Ho L, Hofstetter WL, Lin SH. Impact of comorbidities and use of common medications on cancer and non-cancer specific survival in esophageal carcinoma. BMC Cancer. 2015;15:1095.
    1. Masaki T, Shiratori Y, Rengifo W, Igarashi K, Yamagata M, Kurokohchi K, Uchida N, Miyauchi Y, Yoshiji H, Watanabe S, Omata M, Kuriyama S. Cyclins and cyclin-dependent kinases: comparative study of hepatocellular carcinoma versus cirrhosis. Hepatology. 2003;37:534–543.
    1. Itami A, Shimada Y, Watanabe G, Imamura M. Prognostic value of p27Kip1 and cyclinD1 expression in esophageal cancer. Oncology. 1999;57:311–317.
    1. Fukuchi M, Fukai Y, Kimura H, Sohda M, Miyazaki T, Nakajima M, Masuda N, Tsukada K, Kato H, Kuwano H. Prolyl isomerase Pin1 expression predicts prognosis in patients with esophageal squamous cell carcinoma and correlates with cyclinD1 expression. Int J Oncol. 2006;29:329–334.
    1. Kato K, Gong J, Iwama H, Kitanaka A, Tani J, Miyoshi H, Nomura K, Mimura S, Kobayashi M, Aritomo Y, Kobara H, Mori H, Himoto T, et al. The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol Cancer Ther. 2012;11:549–560.
    1. Zakikhani M, Dowling R, Fantus IG, Sonenberg N, Pollak M. Metformin Is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res. 2006;66:10269–10273.
    1. Sahra IB, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, Tanti JF, le Marchand-Brustel Y, Bost F. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene. 2008;27:3576–3586.
    1. Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LGD, Neumann D, Schlattner U, Wallimann T, Carlson M, Carling D. LKB1 Is the upstream kinase in the AMP-activated protein kinase cascade. Current Biol. 2003;13:2004–2008.
    1. Xiang X, Saha AK, Wen R, Ruderman NB, Luo Z. AMP-activated protein kinase activators can inhibit the growth of prostate cancer cells by multiple mechanisms. Biochem Biophys Res Commun. 2004;321:161–167.
    1. Deshayes F, Nahmias C. Angiotensin receptors: a new role in cancer? Trends Endocrinol Metab. 2005;16:293–299.
    1. Uemura H, Ishiguro H, Nakaigawa N, Nagashima Y, Miyoshi Y, Fujinami K, Sakaguchi A, Kubota Y. Angiotensin II receptor blocker shows antiproliferative activity in prostate cancer cells: a possibility of tyrosine kinase inhibitor of growth factor. Mol Cancer Ther. 2003;2:1139–1147.
    1. Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, Carr D, Deng Y, Jin W, Black S, Long B, Liu J, Dinunzio E, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3:742–750.
    1. Gao N, Flynn DC, Zhang Z, Zhong XS, Walker V, Liu KJ, Shi X, Jiang BH. G1 cell cycle progression and the expression of G1 cyclins are regulated by PI3K/AKT/mTOR/p70S6K1 signaling in human ovarian cancer cells. Am J Physiol Cell Physiol. 2004;287:C281–C291.
    1. Stangier J, Su CA, Roth W. Pharmacokinetics of orally and intravenously administered telmisartan in healthy young and elderly volunteers and in hypertensive patients. J Int Med Res. 2000;28:149–167.
    1. Scalera F, Martens-Lobenhoffer J, Bukowska A, Lendeckel U, Tager M, Bode-Boger SM. Effect of telmisartan on nitric oxide-asymmetrical dimethylarginine system: role of angiotensin II type 1 receptor and peroxisome proliferator activated receptor signaling during endothelial aging. Hypertension. 2008;51:696–703.
    1. Morishita A, Masaki T. miRNA in hepatocellular carcinoma. Hepatology Res. 2015;45:128–141.
    1. Tsouko E, Wang J, Frigo DE, Aydoğdu E, Williams C. miR-200a inhibits migration of triple-negative breast cancer cells through direct repression of the EPHA2 oncogene. Carcinogenesis. 2015;36:1051–1060.
    1. Zhang Y, Duan G, Feng S. MicroRNA-301a modulates doxorubicin resistance in osteosarcoma cells by targeting AMP-activated protein kinase alpha 1. Biochem Biophys Res Commun. 2015;459:367–373.
    1. Masaki T, Tokuda M, Yoshida S, Nakai S, Morishita A, Uchida N, Funaki T, Kita Y, Funakoshi F, Nonomura T, Himoto T, Deguchi A, Kimura Y, et al. Comparison study of the expressions of myristoylated alanine-rich C kinase substrate in hepatocellular carcinoma, liver cirrhosis, chronic hepatitis, and normal liver. Int J Oncol. 2005;26:661–671.
    1. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–254.
    1. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970;227:680–685.
    1. Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci U S A. 1979;76:4350–4354.
    1. D’Incalci M, Colombo T, Ubezio P, Nicoletti I, Giavazzi R, Erba E, Ferrarese L, Meco D, Riccardi R, Sessa C, Cavallini E, Jimeno J, Faircloth GT. The combination of yondelis and cisplatin is synergistic against human tumor xenografts. Eur J Cancer. 2003;39:1920–1926.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc. 1995;57:289–300.

Source: PubMed

3
Se inscrever