Telmisartan anti-cancer activities mechanism through targeting N-cadherin by mimicking ADH-1 function

Marjan Khorsand, Sahar Khajeh, Mahboobeh Eslami, Navid Nezafat, Younes Ghasemi, Vahid Razban, Zohreh Mostafavi-Pour, Marjan Khorsand, Sahar Khajeh, Mahboobeh Eslami, Navid Nezafat, Younes Ghasemi, Vahid Razban, Zohreh Mostafavi-Pour

Abstract

This study aimed to investigate if Telmisartan as a novel N-cadherin antagonist, can overcome cell migration of cancer cells. We investigated the mechanism and influence of Docetaxel and Telmisartan (as an analogous to ADH-1, which is a well-known N-cadherin antagonist) on cancer cells. The effect of ADH-1 and Telmisartan on cell attachment in PC3, DU145, MDA-MB-468 cell lines using recombinant human N-cadherin was studied. Cell viability assay was performed to examine the anti-proliferative effects of Telmisartan, ADH-1 and Docetaxel. Migration was examined via wound healing assay, and apoptosis was determined by flow cytometry. The expression of AKT-1 as a downstream gene of N-cadherin signalling pathway was assayed by real-time PCR. Treatment of PC3, MDA-MB-468 and DU145 cells with Telmisartan (0.1 µM) and ADH-1 (40 µM) resulted in 50%, 58% and approximately 20% reduction in cell attachment to N-cadherin coated plate respectively. It shows reduction of cell attachment in PC3 and MDA-MB-468 cell lines appeared to be more sensitive than that of DU145 cells to the Telmisartan and ADH-1 treatments. Telmisartan (0.1 µM) and Docetaxel (0.01 nM) significantly reduced cell migration in PC3 and MDA-MB-468 cell lines compared with the control group. Using Real-time PCR, we found that Telmisartan, Docetaxel and ADH-1 had significant influence on the AKT-1 mRNA level. The results of the current study for the first time suggest that, Telmisartan, exerts anti-proliferation and anti-migration effects by targeting antagonistically N-cadherin. Also, these data suggest that Telmisartan as a less expensive alternative to ADH-1 could potentiate Docetaxel anticancer effects.

Keywords: ADH-1; N-cadherin; cancer; cell attachment; docetaxel; telmisartan.

Conflict of interest statement

The authors confirm that there are no conflicts of interest.

© 2022 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Effect of DTX, Tel and ADH‐1 on viability of prostate (PC3, DU145) and breast cancer (MDA‐MB‐468) cell lines. The cells were treated with different doses of DTX (0.001–100 nM), Tel and ADH‐1 (0.001–500 µM) for 48 hours. The control cells were treated only with the cell culture medium. Data are expressed as the mean ±SD of three independent experiments (n = 12). DTX; Docetaxel, Tel; Telmisartan
FIGURE 2
FIGURE 2
Effect of DTX and Tel on migration of prostate and breast cancer cell lines. The scratch wound was created using 10 µl sterile pipette in 70% confluency of PC3, DU‐145 and MDA‐MB‐468 cells. Media containing certain concentration of each drug was added to defined wells, and the images were taken at 0 and 24 hours by an inverted microscope (×40 magnification). The lines show the area where the scratch wound was created. The scratch wound assay was performed in triplicate wells for each concentration. Data are expressed as the mean ±SD of three independent experiments (n = 9). DTX; Docetaxel, Tel; Telmisartan. *p < 0.05, ** p < 0.01, ***p < 0.001
FIGURE 3
FIGURE 3
Effect of Tel 0.1 μM and ADH‐1 40 μM on PC3, DU145 and MDA‐MB‐468 cell attachment to recombinant human N‐cadherin (5 μg/mL). The level of nonspecific binding, determined from these cells attachment to wells coated with BSA alone, was subtracted. Values are mean ±SD of three independent experiments. Results show representative of repeated experiments and normalized to 100% binding for untreated cells as control. Tel, Telmisartan. *p < 0.05, ** p < 0.01, ***p < 0.001
FIGURE 4
FIGURE 4
Effect of DTX, Tel and ADH‐1 on AKT‐1 mRNA expression following 48 hours of treatment of PC3, DU145 and MDA‐MB‐468 cell lines that performed by RT‐qPCR. Level of AKT‐1 mRNA expression in various cell lines following treatment with DTX (0.01 nM), Tel (0.1 µM) and ADH‐1 (40 µM) relative to the controls. Each experiment was performed in three independent times. GUSB (β‐Glucuronidase) was used as the loading control. RT‐qPCR, reverse transcription‐quantitative polymerase chain reaction; mRNA, messenger RNA; DTX, Docetaxel; Tel, Telmisartan. *p < 0.05, ** p < 0.01, ***p < 0.001
FIGURE 5
FIGURE 5
Effect of DTX, Tel and ADH‐1 on cell apoptosis following 48 hours treatment of PC3, DU145 and MDA‐MB‐468cell lines that performed by flow cytometry. Percentage of early apoptosis in various cell lines following treatment with DTX (0.01 nM), Tel (0.1 µM) and ADH‐1 (40 µM). Each experiment was performed in three independent times. DTX, Docetaxel; Tel, Telmisartan. *p < 0.05, **p < 0.01, ***p < 0.001

References

    1. Welti J, Sharp A, Brooks N, et al. Targeting the p300/CBP axis in lethal prostate cancer. Cancer Discov. 2021;11(5):1118‐1137.
    1. Bai T, Liu Y, Li B. LncRNA LOXL1‐AS1/miR‐let‐7a‐5p/EGFR‐related pathway regulates the doxorubicin resistance of prostate cancer DU‐145 cells. IUBMB Life. 2019;71(10):1537‐1551.
    1. Thambiraj S, Vijayalakshmi R, Shankaran DR. An effective strategy for development of docetaxel encapsulated gold nanoformulations for treatment of prostate cancer. Sci Rep. 2021;11(1):1‐17.
    1. Lu X, Yang F, Chen D, et al. Quercetin reverses docetaxel resistance in prostate cancer via androgen receptor and PI3K/Akt signaling pathways. Int J Biol Sci. 2020;16(7):1121.
    1. Wang SW, Tai HC, Tang CH, et al. Melatonin impedes prostate cancer metastasis by suppressing MMP‐13 expression. J Cell Physiol. 2021;236(5):3979‐3990.
    1. Voss G, Haflidadóttir BS, Järemo H, et al. Regulation of cell–cell adhesion in prostate cancer cells by microRNA‐96 through upregulation of E‐Cadherin and EpCAM. Carcinogenesis. 2020;41(7):865‐874.
    1. Parol M, Gzil A, Bodnar M, Grzanka D. Systematic review and meta‐analysis of the prognostic significance of microRNAs related to metastatic and EMT process among prostate cancer patients. J Transl Med. 2021;19(1):1‐24.
    1. Feng YL, Chen DQ, Vaziri ND, Guo Y, Zhao YY. Small molecule inhibitors of epithelial‐mesenchymal transition for the treatment of cancer and fibrosis. Med Res Rev. 2020;40(1):54‐78.
    1. Hu S, Duan YX, Zhou Q, Wang Y, Lu Q. Nimotuzumab inhibits epithelial–mesenchymal transition in prostate cancer by targeting the Akt/YB‐1/AR axis. IUBMB Life. 2019;71(7):928‐941.
    1. Williams E, Williams G, Gour BJ, Blaschuk OW, Doherty P. A novel family of cyclic peptide antagonists suggests that N‐cadherin specificity is determined by amino acids that flank the HAV motif. J Biol Chem. 2000;275(6):4007‐4012.
    1. Yarom N, Stewart D, Malik R, et al. Phase I clinical trial of Exherin (ADH‐1) in patients with advanced solid tumors. Curr Clin Pharmacol. 2013;8(1):81‐88.
    1. Shintani Y, Fukumoto Y, Chaika N, et al. ADH‐1 suppresses N‐cadherin‐dependent pancreatic cancer progression. Int J Cancer. 2008;122(1):71‐77.
    1. Long L‐Y, Zhang J, Yang Z, et al. Transdermal delivery of peptide and protein drugs: strategies, advantages and disadvantages. J Drug Deliv Sci Technol. 2020;60:102007.
    1. Sun L. Peptide‐based drug development. Mod Chem Appl. 2013;1(1):1‐2.
    1. Khajeh S, Eslami M, Nezafat N, et al. Surveying FDA‐approved drugs as new potential inhibitors of N‐cadherin protein: a virtual screening approach. Struct Chem. 2020;31(6):2355‐2369.
    1. Eslami M, Nezafat N, Khajeh S, et al. Deep analysis of N‐cadherin/ADH‐1 interaction: a computational survey. J Biomol Struct Dyn. 2019;37(1):210‐228.
    1. Mielczarek‐Puta M, Otto‐Ślusarczyk D, Chrzanowska A, Filipek A, Graboń W. Telmisartan influences the antiproliferative activity of linoleic acid in human colon cancer cells. Nutr Cancer. 2020;72(1):98–109.
    1. Rasheduzzaman M, Moon J‐H, Lee J‐H, Nazim UM, Park S‐Y. Telmisartan generates ROS‐dependent upregulation of death receptor 5 to sensitize TRAIL in lung cancer via inhibition of autophagy flux. Int J Biochem Cell Biol. 2018;102:20‐30.
    1. Matsuyama M, Funao K, Kuratsukuri K, Tanaka T, Kawahito Y, Sano H, Chargui J, Touraine J‐L, Yoshimura N, Yoshimura R. Telmisartan inhibits human urological cancer cell growth through early apoptosis. Exp Therap Med. 2010;1(2):301–306.
    1. Samukawa E, Fujihara S, Oura K, et al. Angiotensin receptor blocker telmisartan inhibits cell proliferation and tumor growth of cholangiocarcinoma through cell cycle arrest. Int J Oncol. 2017;51(6):1674‐1684.
    1. Koyama N, Nishida Y, Ishii T, et al. Telmisartan induces growth inhibition, DNA double‐strand breaks and apoptosis in human endometrial cancer cells. PLoS One. 2014;9(3):e93050.
    1. Nalla AK, Estes N, Patel J, Rao JS. N‐cadherin mediates angiogenesis by regulating monocyte chemoattractant protein‐1 expression via PI3K/Akt signaling in prostate cancer cells. Exp Cell Res. 2011;317(17):2512‐2521.
    1. Tran NL, Adams DG, Vaillancourt RR, Heimark RL. Signal transduction from N‐cadherin increases Bcl‐2: regulation of the phosphatidylinositol 3‐kinase/Akt pathway by homophilic adhesion and actin cytoskeletal organization. J Biol Chem. 2002;277(36):32905‐32914.
    1. Wang W, Wang L, Mizokami A, et al. Down‐regulation of E‐cadherin enhances prostate cancer chemoresistance via Notch signaling. Chin J Cancer. 2017;36(1):1‐13.
    1. Ramezani F, Samadi N, Mostafavi‐Pour Z. Sequential therapy of breast cancer cell lines with vitamin C and quercetin improves the efficacy of chemotherapeutic drugs. Nutr Cancer. 2017;69(6):881‐891.
    1. Hoffman A, Sasaki H, Roberto D, et al. Effect of combination therapy of desmopressin and docetaxel on prostate cancer cell (DU145) proliferation, migration and tumor growth. J Cancer Biol Therap. 2016;1:129‐136.
    1. Dehghani M, Kianpour S, Zangeneh A, Mostafavi‐Pour Z. CXCL12 modulates prostate cancer cell adhesion by altering the levels or activities of β1‐containing integrins. Int. J Cell Biol. 2014.
    1. Pfaffl MW. A new mathematical model for relative quantification in real‐time RT–PCR. Nucleic Acids Res. 2001;29(9):45e‐45.
    1. Lammens T, Swerts K, Derycke L, et al. N‐cadherin in neuroblastoma disease: expression and clinical significance. PLoS One. 2012;7(2):e31206.
    1. Mrozik KM, Cheong CM, Hewett DR, et al. LCRF‐0006, a small molecule mimetic of the N‐cadherin antagonist peptide ADH‐1, synergistically increases multiple myeloma response to bortezomib. FASEB BioAdv. 2020;2(6):339‐353.
    1. Esma F, Salvini M, Troia R, et al. Melphalan hydrochloride for the treatment of multiple myeloma. Expert Opin Pharmacother. 2017;18(11):1127‐1136.
    1. Fujihara S, Morishita A, Ogawa K, et al. The angiotensin II type 1 receptor antagonist telmisartan inhibits cell proliferation and tumor growth of esophageal adenocarcinoma via the AMPKa/mTOR pathway in vitro and in vivo. Oncotarget. 2017;8(5):8536.
    1. Matsui T, Chiyo T, Kobara H, et al. Telmisartan inhibits cell proliferation and tumor growth of esophageal squamous cell carcinoma by inducing S‐phase arrest in vitro and in vivo. Int J Mol Sci. 2019;20(13):3197.
    1. Wu TT‐L, Niu H‐S, Chen L‐J, Cheng J‐T, Tong Y‐C. Increase of human prostate cancer cell (DU145) apoptosis by telmisartan through PPAR‐delta pathway. Eur J Pharmacol. 2016;775:35‐42.
    1. Cao Z‐Q, Wang Z, Leng P. Aberrant N‐cadherin expression in cancer. Biomed Pharmacother. 2019;118:109320.
    1. Hu T, Yang C, Fu M, et al. Cytotoxic effects of docetaxel as a candidate drug of drug‐eluting stent on human umbilical vein endothelial cells and the signaling pathway of cell migration inhibition, adhesion delay and shape change. Regen Biomat. 2017;4(3):167‐178.
    1. Fontana F, Raimondi M, Marzagalli M, et al. Epithelial‐to‐mesenchymal transition markers and CD44 isoforms are differently expressed in 2D and 3D cell cultures of prostate cancer cells. Cells. 2019;8(2):143.
    1. Rasha F, Ramalingam L, Menikdiwela K, et al. Renin angiotensin system inhibition attenuates adipocyte‐breast cancer cell interactions. Exp Cell Res. 2020;394(1):112114.
    1. Zhang S, Wang Y. Telmisartan inhibits NSCLC A549 cell proliferation and migration by regulating the PI3K/AKT signaling pathway. Oncol Letters. 2018;15(4):5859‐5864.
    1. de Araújo Júnior RF, Leitão Oliveira ALC, de Melo Silveira RF, et al. Telmisartan induces apoptosis and regulates Bcl‐2 in human renal cancer cells. Exp Biol Med. 2015;240(1):34‐44.
    1. Singh SK, Apata T, Gordetsky JB, Singh R. Docetaxel combined with thymoquinone induces apoptosis in prostate cancer cells via inhibition of the PI3K/AKT signaling pathway. Cancers (Basel). 2019;11(9):1390.
    1. Mahmoud YK, Abdelrazek HM. Cancer: thymoquinone antioxidant/pro‐oxidant effect as potential anticancer remedy. Biomed Pharmacother. 2019;115:108783.
    1. To KK, Tomlinson B. Targeting the ABCG 2‐overexpressing multidrug resistant (MDR) cancer cells by PPAR γ agonists. Br J Pharmacol. 2013;170(5):1137‐1151.
    1. To KK, Wu WK, Loong HH. PPARgamma agonists sensitize PTEN‐deficient resistant lung cancer cells to EGFR tyrosine kinase inhibitors by inducing autophagy. Eur J Pharmacol. 2018;823:19‐26.
    1. Toren P, Zoubeidi A. Targeting the PI3K/Akt pathway in prostate cancer: challenges and opportunities. Int J Oncol. 2014;45(5):1793‐1801.
    1. Kolinsky M, Rescigno P, Bianchini D, et al. A phase I dose‐escalation study of enzalutamide in combination with the AKT inhibitor AZD5363 (capivasertib) in patients with metastatic castration‐resistant prostate cancer. Ann Oncol. 2020;31(5):619‐625.
    1. Garg M, Manik G, Singhal A, et al. Efficacy and safety of azilsartan medoxomil and telmisartan in hypertensive patients: a randomized, assessor‐blinded study. Saudi J Med Medical Sci. 2020;8(2):87.
    1. Banerjee S, Singh SK, Chowdhury I, Lillard JW Jr, Singh R. Combinatorial effect of curcumin with docetaxel modulates apoptotic and cell survival molecules in prostate cancer. Front Biosci (Elite edition). 2017;9:235.

Source: PubMed

3
Se inscrever