Anti-tumor effect of memantine, an N-methyl-D-aspartate receptor antagonist, against DMH-induced colon cancer in rats

Anti-tumor effect of memantine, an N-methyl-D-aspartate receptor antagonist, against DMH-induced colon cancer in rats

Glutamate levels are significantly higher in colon cancer cells than in normal cells. Increased expression of N-methyl-D-aspartate (NMDA) receptors has been observed in tumor cell lines that cause angiogenesis. Vascular endothelial growth factor (VEGF) promotes proliferation and endothelial migration through the calcium influx. As a result, NMDA receptors may be a therapeutic target of cancer, and inhibition of these receptors may reduce tumor growth. In this study, the effects of memantine, an NMDA receptor antagonist, on histology, tumor size, and number, as well as VEGF level in 1,2 dimethylhydrazine (DMH)-induced colon cancer in rats were investigated. Thirty male Wistar rats were divided into three groups: the control group, the colon cancer group (30 mg/kg of DMH solution was injected subcutaneously twice a week for 24 weeks), and the memantine group (20 mg/kg). The results showed that the injection of DMH induced colon polyps (P

___

  • ]1[ Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol. 2019;14(2):89-103. [CrossRef]
  • ]2[ Kekelidze M, D’Errico L, Pansini M, Tyndall A, Hohmann J. Colorectal cancer: current imaging methods and future perspectives for the diagnosis, staging and therapeutic response evaluation. World J Gastroenterol. 2013;19(46):8502- 8514. [CrossRef]
  • ]3[ Okada A, Takehara H, Yoshida K, Nishi M, Miyake H, Kita Y, Komi N. Increased aspartate and glutamate levels in both gastric and colon cancer tissues. Tokushima J Exp Med. 1993;40(1-2):19-25.
  • ]4[ Stepulak A, Rola R, Polberg K, Ikonomidou C. Glutamate and its receptors in cancer. J Neural Transm. 2014;121(8):933-944. [CrossRef]
  • ]5[ Peng S, Zhang Y, Zhang J, Wang H, Ren B. Glutamate receptors and signal transduction in learning and memory. Mol Biol Rep. 2011;38(1):453-460. [CrossRef]
  • ]6[ Komuro H, Rakic P. Modulation of neuronal migration by NMDA receptors. Science. 1993;260(5104):95-97. [CrossRef]
  • ]7[ Ikonomidou C, Bosch F, Miksa M, Bittigau P, Vöckler J, Dikranian K, Stefovska V. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science. 1999;283(5398):70-74. [CrossRef]
  • ]8[ Mehrotra A, Koiri RK. N-Methyl-D-aspartate (NMDA) receptors: therapeutic target against cancer. Int. J. Immunother. Cancer Res. 2015;1(1):013-017.
  • ]9[ Liu JW, Kim MS, Nagpal J, Yamashita K, Poeta L, Chang X, Lee J, Lui Park H, Jeronimo C, Westra W, Mori M, Moon C, Trink B, Sidransky D. Quantitative hypermethylation of NMDAR2B in human gastric cancer. Int J Cancer. 2007;121(9):1994-2000. [CrossRef]
  • ]10[ Watanabe K, Kanno T, Oshima T, Miwa H, Tashiro C, Nishizaki T. The NMDA receptor NR2A subunit regulates proliferation of MKN45 human gastric cancer cells. Biochem Biophys Res Commun. 2008;367(2):487-490. [CrossRef]
  • ]11[ Kim MS, Yamashita K, Baek JH, Park HL, Carvalho AL, Osada M, Hoque MO, Upadhyay S, Mori M, Moon C, Sidransky D. N-methyl-D-aspartate receptor type 2B is epigenetically inactivated and exhibits tumor-suppressive activity in human esophageal cancer. Cancer Res. 2006;66(7):3409-3418. [CrossRef]
  • ]12[ Abdul M, Hoosein N. N-methyl-D-aspartate receptor in human prostate cancer. J Membr Biol. 2005;205(3):125-128. [CrossRef]
  • ]13[ Stepulak A, Luksch H, Gebhardt C, Uckermann O, Marzahn J, Sifringer M, Rzeski W, Staufner C, Brocke KS, Turski L, Ikonomidou C. Expression of glutamate receptor subunits in human cancers. Histochem Cell Biol. 2009;132(4):435- 445. [CrossRef]
  • ]14[ North WG, Gao G, Memoli VA, Pang RH, Lynch L. Breast cancer expresses functional NMDA receptors. Breast Cancer Res Treat. 2010;122(2):307-314. [CrossRef]
  • ]15[ Stepulak A, Luksch H, Uckermann O, Sifringer M, Rzeski W, Polberg K, Kupisz K, Klatka J, Kiełbus M, Grabarska A, Marzahn J, Turski L, Ikonomidou C. Glutamate receptors in laryngeal cancer cells. Anticancer Res. 2011;31(2):565- 573.
  • ]16[ North WG, Gao G, Jensen A, Memoli VA, Du J. NMDA receptors are expressed by small-cell lung cancer and are potential targets for effective treatment. Clin Pharmacol. 2010;2:31-40. [CrossRef]
  • ]17[ Stepulak A, Sifringer M, Rzeski W, Endesfelder S, Gratopp A, Pohl EE, Bittigau P, Felderhoff-Mueser U, Kaindl AM, Bührer C, Hansen HH, Stryjecka-Zimmer M, Turski L, Ikonomidou C. NMDA antagonist inhibits the extracellular signal-regulated kinase pathway and suppresses cancer growth. Proc Natl Acad Sci U S A. 2005;102(43):15605-15610. [CrossRef]
  • ]18[ Stepulak A, Sifringer M, Rzeski W, Brocke K, Gratopp A, Pohl EE, Turski L, Ikonomidou C. AMPA antagonists inhibit the extracellular signal regulated kinase pathway and suppress lung cancer growth. Cancer Biol Ther. 2007;6(12):1908-1915. [CrossRef]
  • ]19[ Yamaguchi F, Hirata Y, Akram H, Kamitori K, Dong Y, Sui L, Tokuda M. FOXO/TXNIP pathway is involved in the suppression of hepatocellular carcinoma growth by glutamate antagonist MK-801. BMC cancer. 2013;13(1):1-11. [CrossRef]
  • [20] Gynther M, Proietti Silvestri I, Hansen JC, Hansen KB, Malm T, Ishchenko Y, Larsen Y, Han L, Kayser S, Auriola S, Petsalo A, Nielsen B, Pickering DS, Orcid, Bunch L. Augmentation of anticancer drug efficacy in murine hepatocellular carcinoma cells by a peripherally acting competitive N-methyl-d-aspartate (NMDA) receptor antagonist. J Med Chem. 2017; 60(23): 9885-9904. [crossRef]
  • ]21[ Li L, Hanahan D. Hijacking the neuronal NMDAR signaling circuit to promote tumor growth and invasion. Cell. 2013;153(1):86-100. [CrossRef]
  • ]22[ Zadeh MH, Glass CA, Magnussen A, Hancox JC, Bates DO. VEGF‐mediated elevated intracellular calcium and angiogenesis in human microvascular endothelial cells in vitro are inhibited by dominant negative TRPC6. Microcirculation. 2008;15(7):605-614. [CrossRef]
  • ]23[. Munaron L, Scianna M. Multilevel complexity of calcium signaling: Modeling angiogenesis. World J Biol Chem. 2012;3(6):121-126. [CrossRef]
  • [24] Ferguson HJ, Wragg JW, Ward S, Heath VL, Ismail T, Bicknell R. Glutamate dependent NMDA receptor 2D is a novel angiogenic tumor endothelial marker in colorectal cancer. Oncotarget. 2016;7(15):20440-20454. [CrossRef] [25] Duan W, Hu J, Liu Y. Ketamine inhibits colorectal cancer cells malignant potential via blockage of NMDA receptor. Exp Mol Pathol. 2019;107:171-178. [CrossRef]
  • [26] Chen X, Wu Q, You L, Chen S, Zhu M, Miao C. Propofol attenuates pancreatic cancer malignant potential via inhibition of NMDA receptor. Eur J Pharmacol. 2017;795:150-159. [CrossRef]
  • [27] North WG, Liu F, Lin LZ, Tian R, Akerman B. NMDA receptors are important regulators of pancreatic cancer and are potential targets for treatment. Clin Pharmacol. 2017;9:79-86. [CrossRef]
  • [28] Albayrak G, Korkmaz FD. Memantine shifts cancer cell metabolism via AMPK1/2 mediated energetic switch in A549 lung cancer cells. EXCLI J. 2021;20:223-231. [CrossRef]
  • [29] Albayrak G, Konac E, Dikmen A, Bilen C. Memantine induces apoptosis and inhibits cell cycle progression in LNCaP prostate cancer cells. Hum Exp Toxicol. 2018;37(9):953-958. [CrossRef]
  • [30] Seifabadi S, Vaseghi G, Javanmard SH, Omidi E, Tajadini M, Zarrin B. The cytotoxic effect of memantine and its effect on cytoskeletal proteins expression in metastatic breast cancer cell line. Iran J Basic Med Sci. 2017;20(1):41-45. [CrossRef]
  • [31] MacDonald JF, Jackson MF, Beazely MA. Hippocampal long-term synaptic plasticity and signal amplification of NMDA receptors. Crit Rev Neurobiol. 2006;18(1-2):71-84. [CrossRef]
  • [32] Takano T, Lin JH-C, Arcuino G, Gao Q, Yang J, Nedergaard M. Glutamate release promotes growth of malignant gliomas. Nat Med. 2001;7(9):1010-1015. [CrossRef]
  • [33] North WG, Fay MJ, Du J, Cleary M, Gallagher JD, McCann FV. Presence of functional NMDA receptors in a human neuroblastoma cell line. Mol Chem Neuropathol. 1997;30(1):77-94. [CrossRef]
  • [34] Ribeiro MP, Custódio JB, Santos AE. Ionotropic glutamate receptor antagonists and cancer therapy: time to think out of the box? Cancer Chemother Pharmacol. 2017;79(2):219-225. [CrossRef]
  • [35] Hinoi E, Takarada T, Ueshima T, Tsuchihashi Y, Yoneda Y. Glutamate signaling in peripheral tissues. Eur J Biochem. 2004;271(1):1-13. [CrossRef]
  • [36] Tajbakhsh A, Pasdar A, Rezaee M, Fazeli M, Soleimanpour S, Hassanian SM, FarshchiyanYazdi Z, Younesi Rad T, Ferns GA, Avan A. The current status and perspectives regarding the clinical implication of intracellular calcium in breast cancer. J Cell Physiol. 2018;233(8):5623-5641. [CrossRef]
  • [37] Deutsch SI, Tang AH, Burket JA, Benson AD. NMDA receptors on the surface of cancer cells: target for chemotherapy? Biomed Pharmacother. 2014;68(4):493-496. [CrossRef]
  • [38] Rzeski W, Turski L, Ikonomidou C. Glutamate antagonists limit tumor growth. PNAS. 2001;98(11):6372-6377. [CrossRef]
  • [39] Malsy M, Gebhardt K, Gruber M, Wiese C, Graf B, Bundscherer A. Effects of ketamine, s-ketamine, and MK 801 on proliferation, apoptosis, and necrosis in pancreatic cancer cells. BMC Anesthesiol. 2015;15(1):111-118. [CrossRef]
  • [40] Frezzetti D, Gallo M, Maiello MR, D’Alessio A, Esposito C, Chicchinelli N, Normanno N. VEGF as a potential target in lung cancer. Expert Opin Ther Targets. 2017;21(10):959-966. [CrossRef]
  • [41] Atzori MG, Tentori L, Ruffini F, Ceci C, Lisi L, Bonanno E, Scimeca M, Eskilsson E, Daubon T, Miletic H, Vitiani LR, Pallini R, Navarra P, Bjerkvig R, D'Atri S, Lacal PM, Graziani G. The anti-vascular endothelial growth factor receptor-1 monoclonal antibody D16F7 inhibits invasiveness of human glioblastoma and glioblastoma stem cells. J Exp Clin Cancer Res. 2017;36(1):106-121. [CrossRef]
  • [42] Long Y, Karachi A, Grippin A, Tao H, Zhang W, Sayour E, Deleyrolle L, Mitchell D, Lin Z, Huang J. DRES 13. VEGF Blockade Enhances T Regulatory Cell Function By Dysregulating Glutamate Transport in Gbm. Neuro Oncol. 2018;20(Suppl 6):vi78.
  • [43] Deyama S, Bang E, Wohleb ES, Li X-Y, Kato T, Gerhard DM, Dutheil S, Dwyer JM, Taylor SR, Picciotto MR, Duman RS. Role of neuronal VEGF signaling in the prefrontal cortex in the rapid antidepressant effects of ketamine. Am J Psychiatry. 2019;176(5):388-400. [CrossRef]
  • [44] Hoosein NM, Abdul M. Antiproliferative effect of memantine on human prostate, breast and colon cancer cell lines. AACR. 2004;64(7):1010.
  • [45] El-Khadragy MF, Nabil HM, Hassan BN, Tohamy AA, Waaer HF, Yehia HM, Alharbi AM, Moneim AEA. Bone Marrow Cell Therapy on 1,2-Dimethylhydrazine (DMH)-Induced Colon Cancer in Rats. Cell Physiol Biochem. 2018;45(3):1072-1083. [CrossRef]
  • [46] Brunicardi FC, Andersen DK, Billiar TR, Dunn DL, Hunter JG, Matthews JB, Pollock RE, Schwartz's Principles of Surgery, 10th edition., McGraw-Hill Education 2014.
Journal of research in pharmacy (online)-Cover
  • Yayın Aralığı: Yılda 6 Sayı
  • Yayıncı: Marmara Üniversitesi
Sayıdaki Diğer Makaleler

Investigation of total phenolic and flavonoid content of Salvia willeana (Holmboe) Hedge, an endemic plant of Cyprus, and screening of its antioxidant and cholinesterase inhibitory properties

Tugba ERCETIN, Maryam SAFAEI, Hayrettin Ozan GULCAN

In vitro acetylcholinesterase inhibitory activities of fractions and iso-agelasine C isolated from the marine sponge Agelas nakamurai

Suciati SUCIATI, Kornkanok INGKANINAN, Andhika Dwi ARISTYAWAN, Valentika Fitria SETYANINGTYAS, Tutik Sri WAHYUNI, Aty WIDYAWARUYANTİ

Synthesis and study of the effect of 3-substituted chromone derivatives on changes in the activity of mitochondrial complex III under experimental cerebral ischemia

Viktoriya RUKOVITSINA, Eduard OGANESYAN, Dmitry POZDNYAKOV

Chemical profile by LC-Q-TOF-MS of Nigella sativa seed extracts and in vitro antimicrobial activity on bacteria which are determined resistance gene and isolated from nosocomial infection

Hüseyin SERVİ, Dilek ŞATANA, Özgül KISA, Ayhan Ibrahim AYSAL

Downregulation of aromatase by siRNA decreases acetylcholinesterase mRNA and specific activity in SHSY5Y cells

Tuba TÜYLÜ KÜÇÜKKILINÇ

RP-HPLC method development and validation for quantification of letrozole solid lipid nanoparticle

Swaroop Rameshwarji LAHOTI, Sowmya PRIYADARSINI

Anti-tumor effect of memantine, an N-methyl-D-aspartate receptor antagonist, against DMH-induced colon cancer in rats

Kosar JANNESAR, Parisa EFTEKHARİ, Masoumeh POURJABALİ, Naser MASOUDİ, Hamid SORAYA

Nuclear receptor agonist activity studies on some Plantago species and Scutellaria salviifolia Benth.: A particular focus on liver x receptor alpha and retinoid x receptor alpha connected with the inflammation process

Vahap Murat KUTLUAY, Makoto INOUE, Yasin GENC, Zeynep DOGAN, Iclal SARACOGLU

Biological activities of three Phlomis species

Süleyman DOĞU, Nuraniye ERUYGUR, Yavuz BAĞCI, Fatma AYAZ, Damla KIRCI

Preparation and optimization of berberine phospholipid complexes using QbD approach and in vivo evaluation for anti-inflammatory, analgesic and antipyretic activity

Buket AKSU, Ayşegül KARATAŞ, Ayça GÜNGÖR AK, Esra KÜPELİ AKKOL