The SOD Mimic MnTnHex-2-PyP5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells
Abstract
:1. Introduction
2. Materials and Methods
2.1. Chemicals
2.2. Cell Culture
2.3. Crystal Violet (CV) Staining Assay
2.4. MTS Reduction Assay
2.5. Cell DNA Content Analysis
2.6. Apoptosis Assay
2.7. Intracellular ROS Evaluation
2.8. Cytokinesis-block Micronucleus (CBMN) Assay
2.9. In Vitro Wound-Healing Assay
2.10. Chemotaxis
2.11. Statistical Analyses
3. Results
3.1. MnP Decreases 786-O Cell Viability
3.2. MnP Increases 786-O Cell Death
3.3. MnP Increases Intracellular Levels of ROS in 786-O Cells
3.4. MnP Does Not Induce Genotoxicity in 786-O Cells
3.5. MnP Reduces the Chemotactic 786-O Cells’ Migration
4. Discussion
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Koul, H.; Huh, J.-S.; Rove, K.O.; Crompton, L.; Koul, S.; Meacham, R.B.; Kim, F.J. Molecular aspects of renal cell carcinoma: A review. Am. J. Cancer Res. 2011, 1, 240–254. [Google Scholar] [PubMed]
- Escudier, B.; Porta, C.; Schmidinger, M.; Rioux-Leclercq, N.; Bex, A.; Khoo, V.; Gruenvald, V.; Horwich, A. On behalf of the ESMO Guidelines Committee Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, v58–v68. [Google Scholar] [CrossRef] [PubMed]
- Block, K. Oxidative Stress and Redox-Signaling in Renal Cell Cancer. In Emerging Research and Treatments in Renal Cell Carcinoma; Amato, R.J., Ed.; InTech: Rijeka, Croatia, 2012; ISBN 978-953-51-0022-5. [Google Scholar] [Green Version]
- Linehan, W.M.; Ricketts, C.J. Decade in review-kidney cancer: Discoveries, therapies and opportunities. Nat. Rev. Urol. 2014, 11, 614–616. [Google Scholar] [CrossRef] [PubMed]
- Shuch, B.; Amin, A.; Armstrong, A.J.; Eble, J.N.; Ficarra, V.; Lopez-Beltran, A.; Martignoni, G.; Rini, B.I.; Kutikov, A. Understanding pathologic variants of renal cell carcinoma: Distilling therapeutic opportunities from biologic complexity. Eur. Urol. 2015, 67, 85–97. [Google Scholar] [CrossRef]
- Linehan, W.M.; Ricketts, C.J. Kidney cancer in 2016: RCC—advances in targeted therapeutics and genomics. Nat. Rev. Urol. 2016, 14, 76–78. [Google Scholar] [CrossRef]
- Keefe, S.M.; Rathmell, W.K.; Nathanson, K.L. Genomic Assessment of Renal Cancer. In Renal Cell Carcinoma; Springer: New York, NY, USA, 2015; pp. 39–56. [Google Scholar]
- Waris, G.; Ahsan, H. Reactive oxygen species: Role in the development of cancer and various chronic conditions. J. Carcinog. 2006, 5, 14. [Google Scholar] [CrossRef]
- Valko, M.; Izakovic, M.; Mazur, M.; Rhodes, C.J.C.J.; Telser, J. Role of oxygen radicals in DNA damage and cancer incidence. Mol. Cell. Biochem. 2004, 266, 37–56. [Google Scholar] [CrossRef]
- Gius, D.; Spitz, D.R. Redox Signaling in Cancer Biology. Antioxid. Redox Signal. 2006, 8, 1249–1252. [Google Scholar] [CrossRef]
- Šverko, A.; Sobočanec, S.; Kušić, B.; Mačak-Šafranko, Ž.; Šarić, A.; Leniček, T.; Kraus, O.; Andrišić, L.; Korolija, M.; Balog, T.; et al. Superoxide dismutase and cytochrome P450 isoenzymes might be associated with higher risk of renal cell carcinoma in male patients. Int. Immunopharmacol. 2011, 11, 639–645. [Google Scholar] [CrossRef]
- Pljesa-Ercegovac, M.; Mimic-Oka, J.; Dragicevic, D.; Savic-Radojevic, A.; Opacic, M.; Pljesa, S.; Radosavljevic, R.; Simic, T. Altered antioxidant capacity in human renal cell carcinoma: Role of glutathione associated enzymes. Urol. Oncol. 2008, 26, 175–181. [Google Scholar] [CrossRef]
- Atilgan, D.; Parlaktas, B.S.; Uluocak, N.; Kolukcu, E.; Erdemir, F.; Ozyurt, H.; Erkorkmaz, U. The relationship between ALA16VAL single gene polymorphism and renal cell carcinoma. Adv. Urol. 2014, 2014, 932481. [Google Scholar] [CrossRef] [PubMed]
- Holley, A.K.; Miao, L.; St Clair, D.K.; St Clair, W.H. Redox-modulated phenomena and radiation therapy: The central role of superoxide dismutases. Antioxid. Redox Signal. 2014, 20, 1567–1589. [Google Scholar] [CrossRef] [PubMed]
- Kinnula, V.L.; Crapo, J.D. Superoxide dismutases in malignant cells and human tumors. Free Radic. Biol. Med. 2004, 36, 718–744. [Google Scholar] [CrossRef] [PubMed]
- Robbins, D.; Zhao, Y. Manganese superoxide dismutase in cancer prevention. Antioxid. Redox Signal. 2014, 20, 1628–1645. [Google Scholar] [CrossRef] [PubMed]
- Tovmasyan, A.; Sheng, H.; Weitner, T.; Arulpragasam, A.; Lu, M.; Warner, D.S.; Vujaskovic, Z.; Spasojevic, I.; Batinic-Haberle, I. Design, mechanism of action, bioavailability and therapeutic effects of Mn porphyrin-based redox modulators. Med. Princ. Pract. 2013, 22, 103–130. [Google Scholar] [CrossRef] [PubMed]
- Batinić-Haberle, I.; Tovmasyan, A.; Roberts, E.R.H.; Vujaskovic, Z.; Leong, K.W.; Spasojevic, I. SOD Therapeutics: Latest Insights into Their Structure-Activity Relationships and Impact on the Cellular Redox-Based Signaling Pathways. Antioxid. Redox Signal. 2014, 20, 2372–2415. [Google Scholar] [CrossRef]
- Tovmasyan, A.; Sampaio, R.S.; Boss, M.K.; Bueno-Janice, J.C.; Bader, B.H.; Thomas, M.; Reboucas, J.S.; Orr, M.; Chandler, J.D.; Go, Y.M.; et al. Anticancer therapeutic potential of Mn porphyrin/ascorbate system. Free Radic. Biol. Med. 2015, 89, 1231–1247. [Google Scholar] [CrossRef] [Green Version]
- Batinić-Haberle, I.; Spasojević, I.; Stevens, R.D.; Hambright, P.; Fridovich, I. Manganese(iii) meso-tetrakis(ortho-N-alkylpyridyl)porphyrins. Synthesis, characterization, and catalysis of O2-dismutation. J. Chem. Soc. Dalton Trans. 2002, 2689. [Google Scholar] [CrossRef]
- Fernandes, A.S.; Serejo, J.; Gaspar, J.; Cabral, F.; Bettencourt, A.F.; Rueff, J.; Castro, M.; Costa, J.; Oliveira, N.G. Oxidative injury in V79 Chinese hamster cells: Protective role of the superoxide dismutase mimetic MnTM-4-PyP. Cell Biol. Toxicol. 2010, 26, 91–101. [Google Scholar] [CrossRef]
- Caparica, R.; Júlio, A.; Baby, A.R.; Araújo, M.E.M.; Fernandes, A.S.; Costa, J.G.; de Almeida, T.S. Choline-amino acid ionic liquids as green functional excipients to enhance drug solubility. Pharmaceutics 2018, 10, 288. [Google Scholar] [CrossRef]
- Guerreiro, P.S.; Estácio, S.G.; Antunes, F.; Fernandes, A.S.; Pinheiro, P.F.; Costa, J.G.; Castro, M.; Miranda, J.P.; Guedes, R.C.; Oliveira, N.G. Structure-based virtual screening toward the discovery of novel inhibitors of the DNA repair activity of the human apurinic/apyrimidinic endonuclease 1. Chem. Biol. Drug Des. 2016, 88, 915–925. [Google Scholar] [CrossRef] [PubMed]
- Guerreiro, P.S.; Corvacho, E.; Costa, J.G.; Saraiva, N.; Fernandes, A.S.; Castro, M.; Miranda, J.P.; Oliveira, N.G. The APE1 redox inhibitor E3330 reduces collective cell migration of human breast cancer cells and decreases chemoinvasion and colony formation when combined with docetaxel. Chem. Biol. Drug Des. 2017, 90, 561–571. [Google Scholar] [CrossRef] [PubMed]
- Silva, C.O.; Molpeceres, J.; Batanero, B.; Fernandes, A.S.; Saraiva, N.; Costa, J.G.; Rijo, P.; Figueiredo, I.V.; Faísca, P.; Reis, C.P. Functionalized diterpene parvifloron D-loaded hybrid nanoparticles for targeted delivery in melanoma therapy. Ther. Deliv. 2016, 7, 521–544. [Google Scholar] [CrossRef] [PubMed]
- Costa, J.G.; Saraiva, N.; Guerreiro, P.S.; Louro, H.; Silva, M.J.; Miranda, J.P.; Castro, M.; Batinic-Haberle, I.; Fernandes, A.S.; Oliveira, N.G. Ochratoxin A-induced cytotoxicity, genotoxicity and reactive oxygen species in kidney cells: An integrative approach of complementary endpoints. Food Chem. Toxicol. 2016, 87, 65–76. [Google Scholar] [CrossRef] [PubMed]
- Fenech, M. Cytokinesis-block micronucleus cytome assay. Nat. Protoc. 2007, 2, 1084–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fenech, M. The in vitro micronucleus technique. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2000, 455, 81–95. [Google Scholar] [CrossRef]
- Liang, C.-C.; Park, A.Y.; Guan, J.-L. In vitro scratch assay: A convenient and inexpensive method for analysis of cell migration in vitro. Nat. Protoc. 2007, 2, 329–333. [Google Scholar] [CrossRef]
- Fernandes, A.S.; Flórido, A.; Saraiva, N.; Cerqueira, S.; Ramalhete, S.; Cipriano, M.; Cabral, M.F.; Miranda, J.P.; Castro, M.; Costa, J.; et al. Role of the Copper(II) Complex Cu[15]pyN5 in Intracellular ROS and Breast Cancer Cell Motility and Invasion. Chem. Biol. Drug Des. 2015, 86, 578–588. [Google Scholar] [CrossRef]
- Albini, A.; Benelli, R. The chemoinvasion assay: A method to assess tumor and endothelial cell invasion and its modulation. Nat. Protoc. 2007, 2, 504–511. [Google Scholar] [CrossRef]
- Helleday, T.; Petermann, E.; Lundin, C.; Hodgson, B.; Sharma, R.A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 2008, 8, 193–204. [Google Scholar] [CrossRef]
- Yan, S.; Liu, L.; Ren, F.; Gao, Q.; Xu, S.; Hou, B.; Wang, Y.; Jiang, X.; Che, Y. Sunitinib induces genomic instability of renal carcinoma cells through affecting the interaction of LC3-II and PARP-1. Cell Death Dis. 2017, 8, e2988. [Google Scholar] [CrossRef] [PubMed]
- Weydert, C.J.; Waugh, T.A.; Ritchie, J.M.; Iyer, K.S.; Smith, J.L.; Li, L.; Spitz, D.R.; Oberley, L.W. Overexpression of manganese or copper-zinc superoxide dismutase inhibits breast cancer growth. Free Radic. Biol. Med. 2006, 41, 226–237. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Chaiswing, L.; Oberley, T.D.; Batinic-Haberle, I.; St. Clair, W.; Epstein, C.J.; St. Clair, D. A mechanism-based antioxidant approach for the reduction of skin carcinogenesis. Cancer Res. 2005, 65, 1401–1405. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Oberley, T.D.; Oberley, L.W.; Zhong, W. Overexpression of manganese superoxide dismutase in DU145 human prostate carcinoma cells has multiple effects on cell phenotype. Prostate 1998, 35, 221–233. [Google Scholar] [CrossRef]
- Behrend, L.; Mohr, A.; Dick, T.; Zwacka, R.M. Manganese Superoxide Dismutase Induces p53-Dependent Senescence in Colorectal Cancer Cells. Mol. Cell. Biol. 2005, 25, 7758–7769. [Google Scholar] [CrossRef] [Green Version]
- Flórido, A.; Saraiva, N.; Cerqueira, S.; Almeida, N.; Parsons, M.; Batinic-Haberle, I.; Miranda, J.P.; Costa, J.G.; Carrara, G.; Castro, M.; et al. The manganese(III) porphyrin MnTnHex-2-PyP5+ modulates intracellular ROS and breast cancer cell migration: Impact on doxorubicin-treated cells. Redox Biol. 2019, 20, 367–378. [Google Scholar] [CrossRef]
- Brodaczewska, K.K.; Szczylik, C.; Fiedorowicz, M.; Porta, C.; Czarnecka, A.M. Choosing the right cell line for renal cell cancer research. Mol. Cancer 2016, 15, 83. [Google Scholar] [CrossRef]
- Mancilla, I.A.; Coatti, G.C.; Biazi, B.I.; Zanetti, T.A.; Baranoski, A.; Marques, L.A.; Corveloni, A.C.; Lepri, S.R.; Mantovani, M.S. Molecular pathways related to the control of proliferation and cell death in 786-O cells treated with plumbagin. Mol. Biol. Rep. 2019. [Google Scholar] [CrossRef]
- Ou, Y.C.; Yang, C.R.; Cheng, C.L.; Raung, S.L.; Hung, Y.Y.; Chen, C.J. Indomethacin induces apoptosis in 786-O renal cell carcinoma cells by activating mitogen-activated protein kinases and AKT. Eur. J. Pharmacol. 2007, 563, 49–60. [Google Scholar] [CrossRef]
- Maolakuerban, N.; Azhati, B.; Tusong, H.; Abula, A.; Yasheng, A.; Xireyazidan, A. MiR-200c-3p inhibits cell migration and invasion of clear cell renal cell carcinoma via regulating SLC6A1. Cancer Biol. Ther. 2018, 19, 282–291. [Google Scholar] [CrossRef] [Green Version]
- Yeh, I.-J.; Chen, S.-C.; Yen, M.-C.; Wu, Y.-H.; Hung, C.-H.; Kuo, P.-L. 6-Shogaol Suppresses 2-Amino-1-Methyl-6-Phenylimidazo [4,5-b] Pyridine (PhIP)-Induced Human 786-O Renal Cell Carcinoma Osteoclastogenic Activity and Metastatic Potential. Nutrients 2019, 11, 2306. [Google Scholar] [CrossRef] [PubMed]
- Zhou, L.; Liu, X.-D.; Sun, M.; Zhang, X.; German, P.; Bai, S.; Ding, Z.; Tannir, N.; Wood, C.G.; Matin, S.F.; et al. Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene 2016, 35, 2687–2697. [Google Scholar] [CrossRef] [PubMed]
- Weitner, T.; Kos, I.; Sheng, H.; Tovmasyan, A.; Reboucas, J.S.; Fan, P.; Warner, D.S.; Vujaskovic, Z.; Batinic-Haberle, I.; Spasojevic, I. Comprehensive pharmacokinetic studies and oral bioavailability of two Mn porphyrin-based SOD mimics, MnTE-2-PyP5+and MnTnHex-2-PyP5+. Free Radic. Biol. Med. 2013, 58, 73–80. [Google Scholar] [CrossRef] [PubMed]
- Ye, X.; Fels, D.; Tovmasyan, A.; Aird, K.M.; Dedeugd, C.; Allensworth, J.L.; Kos, I.; Park, W.; Spasojevic, I.; Devi, G.R.; et al. Cytotoxic effects of Mn(III) N -alkylpyridylporphyrins in the presence of cellular reductant, ascorbate. Free Radic. Res. 2011, 45, 1289–1306. [Google Scholar] [CrossRef]
- Batinic-Haberle, I.; Tovmasyan, A.; Spasojevic, I. Mn porphyrin-based redox-active drugs—Differential effects as cancer therapeutics and protectors of normal tissue against oxidative injury. Antioxid. Redox Signal. 2018, 29, 1691–1724. [Google Scholar] [CrossRef]
- Henderson, L.M.; Chappell, J.B. Dhydrorhodamine 123: A fluorescent probe for superoxide generation? Eur. J. Biochem. 1993, 217, 973–980. [Google Scholar] [CrossRef]
- Tarpey, M.M.M.; Wink, D.A.; Grisham, M.B. Methods for detection of reactive metabolites of oxygen and nitrogen: In vitro and in vivo considerations. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R431–R444. [Google Scholar] [CrossRef]
- Sarto, C.; Frutiger, S.; Cappellano, F.; Sanchez, J.-C.; Doro, G.; Catanzaro, F.; Hughes, G.J.; Hochstrasser, D.F.; Mocarelli, P. Modified expression of plasma glutathione peroxidase and manganese superoxide dismutase in human renal cell carcinoma. Electrophoresis 1999, 20, 3458–3466. [Google Scholar] [CrossRef]
- Tovmasyan, A.; Maia, C.G.C.; Weitner, T.; Carballal, S.; Sampaio, R.S.; Lieb, D.; Ghazaryan, R.; Ivanovic-Burmazovic, I.; Ferrer-Sueta, G.; Radi, R.; et al. A comprehensive evaluation of catalase-like activity of different classes of redox-active therapeutics. Free Radic. Biol. Med. 2015, 86, 308–321. [Google Scholar] [CrossRef] [Green Version]
- Bandarra, S.; Fernandes, A.S.; Magro, I.; Guerreiro, P.S.; Pingarilho, M.; Churchwell, M.I.; Gil, O.M.; Batinić-Haberle, I.; Gonçalves, S.; Rueff, J.; et al. Mechanistic insights into the cytotoxicity and genotoxicity induced by glycidamide in human mammary cells. Mutagenesis 2013, 28, 721–729. [Google Scholar] [CrossRef] [Green Version]
- Gad, S.C.; Sullivan, D.W.; Crapo, J.D.; Spainhour, C.B. A nonclinical safety assessment of MnTE-2-PyP, a manganese porphyrin. Int. J. Toxicol. 2013, 32, 274–287. [Google Scholar] [CrossRef] [PubMed]
- Gad, S.C.; Sullivan, D.W.; Spasojevic, I.; Mujer, C.V.; Spainhour, C.B.; Crapo, J.D. Nonclinical Safety and Toxicokinetics of MnTnBuOE-2-PyP5+ (BMX-001). Int. J. Toxicol. 2016, 35, 438–453. [Google Scholar] [CrossRef] [PubMed]
- Raimondo, F.; Morosi, L.; Corbetta, S.; Chinello, C.; Brambilla, P.; Della Mina, P.; Villa, A.; Albo, G.; Battaglia, C.; Bosari, S.; et al. Differential protein profiling of renal cell carcinoma urinary exosomes. Mol. Biosyst. 2013, 9, 1220–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandes, A.S.; Saraiva, N.; Oliveira, N.G. Redox Therapeutics in Breast Cancer: Role of SOD Mimics. In Redox-Active Therapeutics; Batinic-Haberle, I., Reboucas, J.S., Spasojevic, I., Eds.; Springer: Cham, Switzerland, 2016; pp. 451–467. ISBN 978-3-319-30705-3. [Google Scholar]
- Egea, J.; Fabregat, I.; Frapart, Y.M.; Ghezzi, P.; Görlach, A.; Kietzmann, T.; Kubaichuk, K.; Knaus, U.G.; Lopez, M.G.; Olaso-Gonzalez, G.; et al. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol. 2017, 13, 94–162. [Google Scholar] [CrossRef] [Green Version]
- Kramer, N.; Walzl, A.; Unger, C.; Rosner, M.; Krupitza, G.; Hengstschläger, M.; Dolznig, H. In vitro cell migration and invasion assays. Mutat. Res. 2013, 752, 10–24. [Google Scholar] [CrossRef]
- Finlay, D.K.; Rosenzweig, E.; Sinclair, L.V.; Feijoo-Carnero, C.; Hukelmann, J.L.; Rolf, J.; Panteleyev, A.A.; Okkenhaug, K.; Cantrell, D.A. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 2012, 209, 2441–2453. [Google Scholar] [CrossRef] [Green Version]
- Zong, H.; Yin, B.; Zhou, H.; Cai, D.; Ma, B.; Xiang, Y. Inhibition of mTOR pathway attenuates migration and invasion of gallbladder cancer via EMT inhibition. Mol. Biol. Rep. 2014, 41, 4507–4512. [Google Scholar] [CrossRef]
- Dolcet, X.; Llobet, D.; Pallares, J.; Matias-Guiu, X. NF-kB in development and progression of human cancer. Virchows Arch. 2005, 446, 475–482. [Google Scholar] [CrossRef]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Costa, J.G.; Saraiva, N.; Batinic-Haberle, I.; Castro, M.; Oliveira, N.G.; Fernandes, A.S. The SOD Mimic MnTnHex-2-PyP5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells. Antioxidants 2019, 8, 490. https://doi.org/10.3390/antiox8100490
Costa JG, Saraiva N, Batinic-Haberle I, Castro M, Oliveira NG, Fernandes AS. The SOD Mimic MnTnHex-2-PyP5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells. Antioxidants. 2019; 8(10):490. https://doi.org/10.3390/antiox8100490
Chicago/Turabian StyleCosta, João G., Nuno Saraiva, Ines Batinic-Haberle, Matilde Castro, Nuno G. Oliveira, and Ana S. Fernandes. 2019. "The SOD Mimic MnTnHex-2-PyP5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells" Antioxidants 8, no. 10: 490. https://doi.org/10.3390/antiox8100490
APA StyleCosta, J. G., Saraiva, N., Batinic-Haberle, I., Castro, M., Oliveira, N. G., & Fernandes, A. S. (2019). The SOD Mimic MnTnHex-2-PyP5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells. Antioxidants, 8(10), 490. https://doi.org/10.3390/antiox8100490