Redox-Regulation in Cancer Stem Cells
Abstract
1. Introduction
2. “Side Population” Cells in Cancer
3. Reactive Oxygen Species and Sources in CSC
4. Reactive Oxygen Species in Tumorigenesis and Disease Progression
5. Redox-Signaling in CSCs: Role of NRF2
6. Signaling Pathways Contributing in the Regulation of ROS Levels in CSC
6.1. Wnt-Signaling Pathway
6.2. Sonic Hedgehog Signaling
6.3. CD13/APN in CSC
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Conflicts of Interest
References
- Boesch, M.; Zeimet, A.G.; Fiegl, H.; Wolf, B.; Huber, J.; Klocker, H.; Gastl, G.; Sopper, S.; Wolf, D. High prevalence of side population in human cancer cell lines. Oncoscience 2016, 3, 85–87. [Google Scholar] [CrossRef] [PubMed]
- Zhu, P.; Fan, Z. Cancer stem cells and tumorigenesis. Biophys. Rep. 2018, 4, 178–188. [Google Scholar] [CrossRef] [PubMed]
- Hirschmann-Jax, C.; Foster, A.E.; Wulf, G.G.; Nuchtern, J.G.; Jax, T.W.; Gobel, U.; Goodell, M.A.; Brenner, M.K. A distinct "side population" of cells with high drug efflux capacity in human tumor cells. Proc. Natl. Acad. Sci. USA 2004, 101, 14228–14233. [Google Scholar] [CrossRef] [PubMed]
- Golebiewska, A.; Brons, N.H.; Bjerkvig, R.; Niclou, S.P. Critical appraisal of the side population assay in stem cell and cancer stem cell research. Cell Stem Cell 2011, 8, 136–147. [Google Scholar] [CrossRef] [PubMed]
- Sales-Pardo, I.; Avendano, A.; Martinez-Munoz, V.; Garcia-Escarp, M.; Celis, R.; Whittle, P.; Barquinero, J.; Domingo, J.C.; Marin, P.; Petriz, J. Flow cytometry of the Side Population: Tips & tricks. Cell Oncol. 2006, 28, 37–53. [Google Scholar]
- Afify, S.M.; Seno, M. Conversion of Stem Cells to Cancer Stem Cells: Undercurrent of Cancer Initiation. Cancers 2019, 11, 345. [Google Scholar] [CrossRef]
- Garnier, D.; Milsom, C.; Magnus, N.; Meehan, B.; Weitz, J.; Yu, J.; Rak, J. Role of the tissue factor pathway in the biology of tumor initiating cells. Thromb. Res. 2010, 125 (Suppl. S2), S44–S50. [Google Scholar] [CrossRef]
- Unruh, D.; Horbinski, C. Beyond thrombosis: The impact of tissue factor signaling in cancer. J. Hematol. Oncol. 2020, 13, 93. [Google Scholar] [CrossRef]
- Tuy, K.; Rickenbacker, L.; Hjelmeland, A.B. Reactive oxygen species produced by altered tumor metabolism impacts cancer stem cell maintenance. Redox Biol. 2021, 44, 101953. [Google Scholar] [CrossRef]
- Aramini, B.; Masciale, V.; Arienti, C.; Dominici, M.; Stella, F.; Martinelli, G.; Fabbri, F. Cancer Stem Cells (CSCs), Circulating Tumor Cells (CTCs) and Their Interplay with Cancer Associated Fibroblasts (CAFs): A New World of Targets and Treatments. Cancers 2022, 14, 2408. [Google Scholar] [CrossRef]
- Mirantes, C.; Espinosa, I.; Ferrer, I.; Dolcet, X.; Prat, J.; Matias-Guiu, X. Epithelial-to-mesenchymal transition and stem cells in endometrial cancer. Hum. Pathol. 2013, 44, 1973–1981. [Google Scholar] [CrossRef] [PubMed]
- Mizrak, D.; Brittan, M.; Alison, M. CD133: Molecule of the moment. J. Pathol. 2008, 214, 3–9. [Google Scholar] [CrossRef] [PubMed]
- Nakashima, M.; Watanabe, M.; Nakano, K.; Uchimaru, K.; Horie, R. Differentiation of Hodgkin lymphoma cells by reactive oxygen species and regulation by heme oxygenase-1 through HIF-1α. Cancer Sci. 2021, 112, 2542–2555. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Sun, M.; Zhang, X.; Xu, Z.; Miyamoto, H.; Zheng, Y. Activation of Glucocorticoid Receptor Inhibits the Stem-Like Properties of Bladder Cancer via Inactivating the β-Catenin Pathway. Front Oncol. 2020, 10, 1332. [Google Scholar] [CrossRef]
- Foster, B.A.; Gangavarapu, K.J.; Mathew, G.; Azabdaftari, G.; Morrison, C.D.; Miller, A.; Huss, W.J. Human prostate side population cells demonstrate stem cell properties in recombination with urogenital sinus mesenchyme. PLoS ONE 2013, 8, e55062. [Google Scholar] [CrossRef]
- Huang, B.; Huang, Y.J.; Yao, Z.J.; Chen, X.; Guo, S.J.; Mao, X.P.; Wang, D.H.; Chen, J.X.; Qiu, S.P. Cancer stem cell-like side population cells in clear cell renal cell carcinoma cell line 769P. PLoS ONE 2013, 8, e68293. [Google Scholar] [CrossRef]
- Liu, T.; Xu, H.; Huang, M.; Ma, W.; Saxena, D.; Lustig, R.A.; Alonso-Basanta, M.; Zhang, Z.; O’Rourke, D.M.; Zhang, L.; et al. Circulating Glioma Cells Exhibit Stem Cell-like Properties. Cancer Res. 2018, 78, 6632–6642. [Google Scholar] [CrossRef]
- Ho, M.M.; Ng, A.V.; Lam, S.; Hung, J.Y. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 2007, 67, 4827–4833. [Google Scholar] [CrossRef]
- Platet, N.; Mayol, J.F.; Berger, F.; Herodin, F.; Wion, D. Fluctuation of the SP/non-SP phenotype in the C6 glioma cell line. FEBS Lett. 2007, 581, 1435–1440. [Google Scholar] [CrossRef]
- Xiong, B.; Ma, L.; Hu, X.; Zhang, C.; Cheng, Y. Characterization of side population cells isolated from the colon cancer cell line SW480. Int. J. Oncol. 2014, 45, 1175–1183. [Google Scholar] [CrossRef]
- Yang, Y.; Fan, Y.; Qi, Y.; Liu, D.; Wu, K.; Wen, F.; Zhao, S. Side population cells separated from A549 lung cancer cell line possess cancer stem cell-like properties and inhibition of autophagy potentiates the cytotoxic effect of cisplatin. Oncol. Rep. 2015, 34, 929–935. [Google Scholar] [CrossRef] [PubMed]
- Chua, C.; Zaiden, N.; Chong, K.H.; See, S.J.; Wong, M.C.; Ang, B.T.; Tang, C. Characterization of a side population of astrocytoma cells in response to temozolomide. J. Neurosurg. 2008, 109, 856–866. [Google Scholar] [CrossRef] [PubMed]
- Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef] [PubMed]
- Szotek, P.P.; Pieretti-Vanmarcke, R.; Masiakos, P.T.; Dinulescu, D.M.; Connolly, D.; Foster, R.; Dombkowski, D.; Preffer, F.; Maclaughlin, D.T.; Donahoe, P.K. Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc. Natl. Acad. Sci. USA 2006, 103, 11154–11159. [Google Scholar] [CrossRef]
- Wu, C.; Alman, B.A. Side population cells in human cancers. Cancer Lett. 2008, 268, 1–9. [Google Scholar] [CrossRef]
- Chiba, T.; Kita, K.; Zheng, Y.W.; Yokosuka, O.; Saisho, H.; Iwama, A.; Nakauchi, H.; Taniguchi, H. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology 2006, 44, 240–251. [Google Scholar] [CrossRef]
- Haraguchi, N.; Utsunomiya, T.; Inoue, H.; Tanaka, F.; Mimori, K.; Barnard, G.F.; Mori, M. Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cell. 2006, 24, 506–513. [Google Scholar] [CrossRef]
- Wu, C.; Wei, Q.; Utomo, V.; Nadesan, P.; Whetstone, H.; Kandel, R.; Wunder, J.S.; Alman, B.A. Side population cells isolated from mesenchymal neoplasms have tumor initiating potential. Cancer Res. 2007, 67, 8216–8222. [Google Scholar] [CrossRef]
- Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef]
- Stingl, J.; Eirew, P.; Ricketson, I.; Shackleton, M.; Vaillant, F.; Choi, D.; Li, H.I.; Eaves, C.J. Purification and unique properties of mammary epithelial stem cells. Nature 2006, 439, 993–997. [Google Scholar] [CrossRef]
- Clement, V.; Marino, D.; Cudalbu, C.; Hamou, M.F.; Mlynarik, V.; de Tribolet, N.; Dietrich, P.Y.; Gruetter, R.; Hegi, M.E.; Radovanovic, I. Marker-independent identification of glioma-initiating cells. Nat. Method. 2010, 7, 224–228. [Google Scholar] [CrossRef] [PubMed]
- Panieri, E.; Santoro, M.M. ROS homeostasis and metabolism: A dangerous liason in cancer cells. Cell Death Dis. 2016, 7, e2253. [Google Scholar] [CrossRef] [PubMed]
- D’Autreaux, B.; Toledano, M.B. ROS as signalling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007, 8, 813–824. [Google Scholar] [CrossRef] [PubMed]
- Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
- Zhou, D.; Shao, L.; Spitz, D.R. Reactive oxygen species in normal and tumor stem cells. Adv. Cancer Res. 2014, 122, 1–67. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, I.M.; Aykin-Burns, N.; Sim, J.E.; Walsh, S.A.; Higashikubo, R.; Buettner, G.R.; Venkataraman, S.; Mackey, M.A.; Flanagan, S.W.; Oberley, L.W.; et al. Mitochondrial O2*- and H2O2 mediate glucose deprivation-induced stress in human cancer cells. J. Biol. Chem. 2005, 280, 4254–4263. [Google Scholar] [CrossRef] [PubMed]
- Boveris, A. Mitochondrial production of superoxide radical and hydrogen peroxide. Adv. Exp. Med. Biol. 1977, 78, 67–82. [Google Scholar]
- Tebay, L.E.; Robertson, H.; Durant, S.T.; Vitale, S.R.; Penning, T.M.; Dinkova-Kostova, A.T.; Hayes, J.D. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it at-tenuates degenerative disease. Free Radic. Biol. Med. 2015, 88 Pt B, 108–146. [Google Scholar] [CrossRef]
- Weyemi, U.; Redon, C.E.; Parekh, P.R.; Dupuy, C.; Bonner, W.M. NADPH Oxidases NOXs and DUOXs as putative targets for cancer therapy. Anticancer Agent. Med. Chem. 2013, 13, 502–514. [Google Scholar]
- Mintz, J.; Vedenko, A.; Rosete, O.; Shah, K.; Goldstein, G.; Hare, J.M.; Ramasamy, R.; Arora, H. Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics. Vaccines 2021, 9, 94. [Google Scholar] [CrossRef]
- Switzer, C.H.; Glynn, S.A.; Cheng, R.Y.; Ridnour, L.A.; Green, J.E.; Ambs, S.; Wink, D.A. S-nitrosylation of EGFR and Src activates an oncogenic signaling network in human basal-like breast cancer. Mol. Cancer Res. 2012, 10, 1203–1215. [Google Scholar] [CrossRef] [PubMed]
- Zhou, F.; Gao, S.; Han, D.; Han, W.; Chen, S.; Patalano, S.; Macoska, J.A.; He, H.H.; Cai, C. TMPRSS2-ERG activates NO-cGMP signaling in prostate cancer cells. Oncogene 2019, 38, 4397–4411. [Google Scholar] [CrossRef] [PubMed]
- Belgorosky, D.; Girouard, J.; Langle, Y.V.; Hamelin-Morrissete, J.; Marino, L.; Aguero, E.I.; Malagrino, H.; Reyes-Moreno, C.; Eijan, A.M. Relevance of iNOS expression in tumor growth and maintenance of cancer stem cells in a bladder cancer model. J. Mol. Med. 2020, 98, 1615–1627. [Google Scholar] [CrossRef] [PubMed]
- Charles, N.; Ozawa, T.; Squatrito, M.; Bleau, A.M.; Brennan, C.W.; Hambardzumyan, D.; Holland, E.C. Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell 2010, 6, 141–152. [Google Scholar] [CrossRef] [PubMed]
- Palumbo, P.; Lombardi, F.; Siragusa, G.; Dehcordi, S.R.; Luzzi, S.; Cimini, A.; Cifone, M.G.; Cinque, B. Involvement of NOS2 Activity on Human Glioma Cell Growth, Clonogenic Potential, and Neurosphere Generation. Int. J. Mol. Sci. 2018, 19, 2801. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Li, Y.; Tsung, A.; Huang, H.; Du, Q.; Yang, M.; Deng, M.; Xiong, S.; Wang, X.; Zhang, L.; et al. iNOS promotes CD24(+)CD133(+) liver cancer stem cell phenotype through a TACE/ADAM17-dependent Notch signaling pathway. Proc. Natl. Acad. Sci. USA 2018, 115, E10127–E10136. [Google Scholar] [CrossRef]
- Penarando, J.; Lopez-Sanchez, L.M.; Mena, R.; Guil-Luna, S.; Conde, F.; Hernandez, V.; Toledano, M.; Gudino, V.; Raponi, M.; Billard, C.; et al. A role for endothelial nitric oxide synthase in intestinal stem cell proliferation and mesenchymal colorectal cancer. BMC Biol. 2018, 16, 3. [Google Scholar] [CrossRef]
- Maiuthed, A.; Bhummaphan, N.; Luanpitpong, S.; Mutirangura, A.; Aporntewan, C.; Meeprasert, A.; Rungrotmongkol, T.; Rojanasakul, Y.; Chanvorachote, P. Nitric oxide promotes cancer cell dedifferentiation by disrupting an Oct4:caveolin-1 complex: A new regulatory mechanism for cancer stem cell formation. J. Biol. Chem. 2018, 293, 13534–13552. [Google Scholar] [CrossRef]
- Luanpitpong, S.; Chanvorachote, P. Nitric Oxide and Aggressive Behavior of Lung Cancer Cells. Anticancer Res. 2015, 35, 4585–4592. [Google Scholar]
- Gao, W.; Wang, Y.; Yu, S.; Wang, Z.; Ma, T.; Chan, A.M.; Chiu, P.K.; Ng, C.F.; Wu, D.; Chan, F.L. Endothelial nitric oxide synthase (eNOS)-NO signaling axis functions to promote the growth of prostate cancer stem-like cells. Stem Cell Res. Ther. 2022, 13, 188. [Google Scholar] [CrossRef]
- Xing, F.; Hu, Q.; Qin, Y.; Xu, J.; Zhang, B.; Yu, X.; Wang, W. The Relationship of Redox With Hallmarks of Cancer: The Importance of Homeostasis and Context. Front. Oncol. 2022, 12, 862743. [Google Scholar] [CrossRef] [PubMed]
- Zhou, F.; Shen, Q.; Claret, F.X. Novel roles of reactive oxygen species in the pathogenesis of acute myeloid leukemia. J. Leukoc. Biol. 2013, 94, 423–429. [Google Scholar] [CrossRef] [PubMed]
- Szatrowski, T.P.; Nathan, C.F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 1991, 51, 794–798. [Google Scholar] [PubMed]
- Cadenas, E.; Wefers, H.; Müller, A.; Brigelius, R.; Sies, H. Active oxygen metabolites and their action in the hepatocyte. Studies on chemiluminescence responses and alkane production. Agent. Action. 1982, 11, 203–216. [Google Scholar]
- Jones, D.P. Redefining oxidative stress. Antioxid. Redox Signal. 2020, 8, 1865–1879. [Google Scholar] [CrossRef]
- Jones, D.P. Radical-free biology of oxidative stress. Am. J. Physiol. Cell Physiol. 2008, 295, C849–C868. [Google Scholar] [CrossRef]
- Ghezzi, P. Oxidoreduction of protein thiols in redox regulation. Biochem. Soc. Trans. 2005, 33 Pt 6, 1378–1381. [Google Scholar] [CrossRef]
- Shi, X.; Zhang, Y.; Zheng, J.; Pan, J. Reactive oxygen species in cancer stem cells. Antioxid. Redox Signal. 2012, 16, 1215–1228. [Google Scholar] [CrossRef]
- Ishimoto, T.; Nagano, O.; Yae, T.; Tamada, M.; Motohara, T.; Oshima, H.; Oshima, M.; Ikeda, T.; Asaba, R.; Yagi, H.; et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell 2011, 19, 387–400. [Google Scholar] [CrossRef]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef]
- Ye, X.Q.; Li, Q.; Wang, G.H.; Sun, F.F.; Huang, G.J.; Bian, X.W.; Yu, S.C.; Qian, G.S. Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells. Int. J. Cancer 2011, 129, 820–831. [Google Scholar] [CrossRef] [PubMed]
- Dong, C.; Yuan, T.; Wu, Y.; Wang, Y.; Fan, T.W.; Miriyala, S.; Lin, Y.; Yao, J.; Shi, J.; Kang, T.; et al. Loss of FBP1 by Snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell 2013, 23, 316–331. [Google Scholar] [CrossRef] [PubMed]
- Haraguchi, N.; Ishii, H.; Mimori, K.; Tanaka, F.; Ohkuma, M.; Kim, H.M.; Akita, H.; Takiuchi, D.; Hatano, H.; Nagano, H.; et al. CD13 is a therapeutic target in human liver cancer stem cells. J. Clin. Investig. 2010, 120, 3326–3339. [Google Scholar] [CrossRef]
- Kim, H.M.; Haraguchi, N.; Ishii, H.; Ohkuma, M.; Okano, M.; Mimori, K.; Eguchi, H.; Yamamoto, H.; Nagano, H.; Sekimoto, M.; et al. Increased CD13 expression reduces reactive oxygen species, promoting survival of liver cancer stem cells via an epithelial-mesenchymal transition-like phenomenon. Ann. Surg. Oncol. 2012, 19 (Suppl. S3), S539–S548. [Google Scholar] [CrossRef] [PubMed]
- Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef] [PubMed]
- Sekhar, K.R.; Rachakonda, G.; Freeman, M.L. Cysteine-based regulation of the CUL3 adaptor protein Keap1. Toxicol. Appl. Pharmacol. 2010, 244, 21–26. [Google Scholar] [CrossRef] [PubMed]
- Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Gene. Cell. 2011, 16, 123–140. [Google Scholar] [CrossRef]
- Villeneuve, N.F.; Lau, A.; Zhang, D.D. Regulation of the Nrf2-Keap1 antioxidant response by the ubiquitin proteasome system: An insight into cullin-ring ubiquitin ligases. Antioxid Redox Signal. 2010, 13, 1699–1712. [Google Scholar] [CrossRef]
- Raghunath, A.; Sundarraj, K.; Nagarajan, R.; Arfuso, F.; Bian, J.; Kumar, A.P.; Sethi, G.; Perumal, E. Antioxidant response elements: Discovery, classes, regulation and potential applications. Redox Biol. 2018, 17, 297–314. [Google Scholar] [CrossRef]
- Choi, B.H.; Kim, J.M.; Kwak, M.K. The multifaceted role of NRF2 in cancer progression and cancer stem cells maintenance. Arch. Pharm. Res. 2021, 44, 263–280. [Google Scholar] [CrossRef]
- Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal 2018, 29, 1727–1745. [Google Scholar] [CrossRef]
- Suzuki, M.; Otsuki, A.; Keleku-Lukwete, N.; Yamamoto, M. Overview of redox-regulation by Keap1-Nrf2 system in toxicology and cancer. Curr. Opinion. Toxicol. 2016, 1, 29–36. [Google Scholar] [CrossRef]
- Kwak, M.K.; Wakabayashi, N.; Itoh, K.; Motohashi, H.; Yamamoto, M.; Kensler, T.W. Modulation of gene expression by cancer chemopreventive dithiolethiones through the Keap1-Nrf2 pathway. Identification of novel gene clusters for cell survival. J. Biol. Chem. 2003, 278, 8135–8145. [Google Scholar] [CrossRef]
- Tu, W.; Wang, H.; Li, S.; Liu, Q.; Sha, H. The Anti-Inflammatory and Anti-Oxidant Mechanisms of the Keap1/Nrf2/ARE Signaling Pathway in Chronic Diseases. Aging Dis. 2019, 10, 637–651. [Google Scholar] [CrossRef]
- Wu, S.; Lu, H.; Bai, Y. Nrf2 in cancers: A double-edged sword. Cancer Med. 2019, 8, 2252–2267. [Google Scholar] [CrossRef]
- Mostafazadeh, M.; Kahroba, H.; Haiaty, S.; TazeKand, A.P.; Samadi, N.; Rahbarghazi, R.; Nouri, M. In vitro exosomal transfer of Nrf2 led to the oxaliplatin resistance in human colorectal cancer LS174T cells. Cell Biochem. Funct. 2022, 40, 391–402. [Google Scholar] [CrossRef]
- Wang, L.; Liu, X.; Kang, Q.; Pan, C.; Zhang, T.; Feng, C.; Chen, L.; Wei, S.; Wang, J. Nrf2 Overexpression Decreases Vincristine Chemotherapy Sensitivity Through the PI3K-AKT Pathway in Adult B-Cell Acute Lymphoblastic Leukemia. Front. Oncol. 2022, 12, 876556. [Google Scholar] [CrossRef]
- Hayes, J.D.; McMahon, M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trend. Biochem. Sci. 2009, 34, 176–188. [Google Scholar] [CrossRef]
- Guo, D.; Wu, B.; Yan, J.; Li, X.; Sun, H.; Zhou, D. A possible gene silencing mechanism: Hypermethylation of the Keap1 promoter abrogates binding of the transcription factor Sp1 in lung cancer cells. Biochem. Biophys. Res. Commun. 2012, 428, 80–85. [Google Scholar] [CrossRef]
- Hanada, N.; Takahata, T.; Zhou, Q.; Ye, X.; Sun, R.; Itoh, J.; Ishiguro, A.; Kijima, H.; Mimura, J.; Itoh, K.; et al. Methylation of the KEAP1 gene promoter region in human colorectal cancer. BMC Cancer 2012, 12, 66. [Google Scholar] [CrossRef]
- Sparaneo, A.; Fabrizio, F.P.; la Torre, A.; Graziano, P.; Di Maio, M.; Fontana, A.; Bisceglia, M.; Rossi, A.; Pizzolitto, S.; De Maglio, G.; et al. Effects of KEAP1 Silencing on the Regulation of NRF2 Activity in Neuroendocrine Lung Tumors. Int. J. Mol. Sci. 2019, 20, 2531. [Google Scholar] [CrossRef]
- Wang, D.; Ma, Y.; Yang, X.; Xu, X.; Zhao, Y.; Zhu, Z.; Wang, X.; Deng, H.; Li, C.; Gao, F.; et al. Hypermethylation of the Keap1 gene inactivates its function, promotes Nrf2 nuclear accumulation, and is involved in arsenite-induced human keratinocyte transformation. Free Radic. Biol. Med. 2015, 89, 209–219. [Google Scholar] [CrossRef]
- Wang, R.; An, J.; Ji, F.; Jiao, H.; Sun, H.; Zhou, D. Hypermethylation of the Keap1 gene in human lung cancer cell lines and lung cancer tissues. Biochem. Biophys. Res. Commun. 2008, 373, 151–154. [Google Scholar] [CrossRef]
- Kahroba, H.; Shirmohamadi, M.; Hejazi, M.S.; Samadi, N. The Role of Nrf2 signaling in cancer stem cells: From stemness and self-renewal to tumorigenesis and chemoresistance. Life Sci. 2019, 239, 116986. [Google Scholar] [CrossRef]
- Gao, L.; Morine, Y.; Yamada, S.; Saito, Y.; Ikemoto, T.; Tokuda, K.; Takasu, C.; Miyazaki, K.; Shimada, M. Nrf2 signaling promotes cancer stemness, migration, and expression of ABC transporter genes in sorafenib-resistant hepatocellular carcinoma cells. PLoS ONE 2021, 16, e0256755. [Google Scholar] [CrossRef]
- Kim, D.; Choi, B.H.; Ryoo, I.G.; Kwak, M.K. High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: Inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis. 2018, 9, 896. [Google Scholar] [CrossRef]
- Zhu, J.; Wang, H.; Fan, Y.; Hu, Y.; Ji, X.; Sun, Q.; Liu, H. Knockdown of nuclear factor erythroid 2-related factor 2 by lentivirus induces differentiation of glioma stem-like cells. Oncol. Rep. 2014, 32, 1170–1178. [Google Scholar] [CrossRef]
- Jang, J.; Wang, Y.; Lalli, M.A.; Guzman, E.; Godshalk, S.E.; Zhou, H.; Kosik, K.S. Primary Cilium-Autophagy-Nrf2 (PAN) Axis Activation Commits Human Embryonic Stem Cells to a Neuroectoderm Fate. Cell 2016, 165, 410–420. [Google Scholar] [CrossRef]
- Kim, D.H.; Jang, J.H.; Kwon, O.S.; Cha, H.J.; Youn, H.J.; Chun, K.S.; Surh, Y.J. Nuclear Factor Erythroid-Derived 2-Like 2-Induced Reductive Stress Favors Self-Renewal of Breast Cancer Stem-Like Cells via the FoxO3a-Bmi-1 Axis. Antioxid. Redox Signal. 2020, 32, 1313–1329. [Google Scholar] [CrossRef]
- Chang, Q.; Bi, Z.; Fu, Y.; Rice, M.K.A.; Zhang, Q.; Wadgaonkar, P.; Almutairy, B.; Zhang, W.; Lu, Y.; Xu, L.; et al. Characterization of Arsenic-Induced Cancer Stem-Like Cells. Method. Mol. Biol. 2020, 2117, 293–303. [Google Scholar]
- Waalkes, M.P.; Liu, J.; Germolec, D.R.; Trempus, C.S.; Cannon, R.E.; Tokar, E.J.; Tennant, R.W.; Ward, J.M.; Diwan, B.A. Arsenic exposure in utero exacerbates skin cancer response in adulthood with contemporaneous distortion of tumor stem cell dynamics. Cancer Res. 2008, 68, 8278–8285. [Google Scholar] [CrossRef]
- Bi, Z.; Zhang, Q.; Fu, Y.; Wadgaonkar, P.; Zhang, W.; Almutairy, B.; Xu, L.; Rice, M.; Qiu, Y.; Thakur, C.; et al. Nrf2 and HIF1alpha converge to arsenic-induced metabolic reprogramming and the formation of the cancer stem-like cells. Theranostics 2020, 10, 4134–4149. [Google Scholar] [CrossRef]
- Goto, S.; Kawabata, T.; Li, T.S. Enhanced Expression of ABCB1 and Nrf2 in CD133-Positive Cancer Stem Cells Associates with Doxorubicin Resistance. Stem Cells Int. 2020, 2020, 8868849. [Google Scholar] [CrossRef]
- Noman, A.S.M.; Parag, R.R.; Rashid, M.I.; Rahman, M.Z.; Chowdhury, A.A.; Sultana, A.; Jerin, C.; Siddiqua, A.; Rahman, L.; Shirin, A.; et al. Widespread expression of Sonic hedgehog (Shh) and Nrf2 in patients treated with cisplatin predicts outcome in resected tumors and are potential therapeutic targets for HPV-negative head and neck cancer. Ther. Adv. Med. Oncol. 2020, 12, 1758835920911229. [Google Scholar] [CrossRef]
- Kuo, K.T.; Lin, C.H.; Wang, C.H.; Pikatan, N.W.; Yadav, V.K.; Fong, I.H.; Yeh, C.T.; Lee, W.H.; Huang, W.C. HNMT Upregulation Induces Cancer Stem Cell Formation and Confers Protection against Oxidative Stress through Interaction with HER2 in Non-Small-Cell Lung Cancer. Int. J. Mol. Sci. 2022, 23, 1663. [Google Scholar] [CrossRef]
- von Mach-Szczypinski, J.; Stanosz, S.; Sieja, K.; Stanosz, M. Histamine and its metabolizing enzymes in tissues of primary ductal breast cancer. Eur. J. Gynaecol. Oncol. 2009, 30, 509–511. [Google Scholar]
- Jaganjac, M.; Milkovic, L.; Sunjic, S.B.; Zarkovic, N. The NRF2, Thioredoxin, and Glutathione System in Tumorigenesis and Anticancer Therapies. Antioxidants 2020, 9, 1151. [Google Scholar] [CrossRef]
- Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef]
- Lendeckel, U.; Venz, S.; Wolke, C. Macrophages: Shapes and functions. ChemTexts 2022, 8, 12. [Google Scholar] [CrossRef]
- DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
- Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef]
- Feng, R.; Morine, Y.; Ikemoto, T.; Imura, S.; Iwahashi, S.; Saito, Y.; Shimada, M. Nrf2 activation drive macrophages polarization and cancer cell epithelial-mesenchymal transition during interaction. Cell Commun. Signal. 2018, 16, 54. [Google Scholar] [CrossRef]
- Engblom, C.; Pfirschke, C.; Pittet, M.J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef]
- Ruffell, B.; Coussens, L.M. Macrophages and therapeutic resistance in cancer. Cancer Cell 2015, 27, 462–472. [Google Scholar] [CrossRef]
- Park, J.; Kim, S.K.; Hallis, S.P.; Choi, B.H.; Kwak, M.K. Role of CD133/NRF2 Axis in the Development of Colon Cancer Stem Cell-Like Properties. Front. Oncol. 2021, 11, 808300. [Google Scholar] [CrossRef]
- Peng, L.; Xiong, Y.; Wang, R.; Xiang, L.; Zhou, H.; Fu, Z. The critical role of peroxiredoxin-2 in colon cancer stem cells. Aging 2021, 13, 11170–11187. [Google Scholar] [CrossRef]
- Ryoo, I.G.; Choi, B.H.; Ku, S.K.; Kwak, M.K. High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: Implications for cancer stem cell resistance. Redox Biol. 2018, 17, 246–258. [Google Scholar] [CrossRef]
- MacDonald, B.T.; Tamai, K.; He, X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev. Cell 2009, 17, 9–26. [Google Scholar] [CrossRef]
- Wang, R.; Sun, Q.; Wang, P.; Liu, M.; Xiong, S.; Luo, J.; Huang, H.; Du, Q.; Geller, D.A.; Cheng, B. Notch and Wnt/beta-catenin signaling pathway play important roles in activating liver cancer stem cells. Oncotarget 2016, 7, 5754–5768. [Google Scholar] [CrossRef]
- Fodde, R.; Brabletz, T. Wnt/beta-catenin signaling in cancer stemness and malignant behavior. Curr. Opin. Cell Biol. 2007, 19, 150–158. [Google Scholar] [CrossRef]
- Clevers, H.; Nusse, R. Wnt/beta-catenin signaling and disease. Cell 2012, 149, 1192–1205. [Google Scholar] [CrossRef]
- Wolke, C.; Antileo, E.; Lendeckel, U. WNT signaling in atrial fibrillation. Exp. Biol. Med. 2021, 246, 1112–1120. [Google Scholar] [CrossRef]
- Yamashita, T.; Budhu, A.; Forgues, M.; Wang, X.W. Activation of hepatic stem cell marker EpCAM by Wnt-beta-catenin signaling in hepatocellular carcinoma. Cancer Res. 2007, 67, 10831–10839. [Google Scholar] [CrossRef]
- Yang, W.; Yan, H.X.; Chen, L.; Liu, Q.; He, Y.Q.; Yu, L.X.; Zhang, S.H.; Huang, D.D.; Tang, L.; Kong, X.N.; et al. Wnt/beta-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells. Cancer Res. 2008, 68, 4287–4295. [Google Scholar] [CrossRef]
- Martin-Rufian, M.; Nascimento-Gomes, R.; Higuero, A.; Crisma, A.R.; Campos-Sandoval, J.A.; Gomez-Garcia, M.C.; Cardona, C.; Cheng, T.; Lobo, C.; Segura, J.A.; et al. Both GLS silencing and GLS2 overexpression synergize with oxidative stress against proliferation of glioma cells. J. Mol. Med. 2014, 92, 277–290. [Google Scholar] [CrossRef]
- Ulanet, D.B.; Couto, K.; Jha, A.; Choe, S.; Wang, A.; Woo, H.K.; Steadman, M.; DeLaBarre, B.; Gross, S.; Driggers, E.; et al. Mesenchymal phenotype predisposes lung cancer cells to impaired proliferation and redox stress in response to glutaminase inhibition. PLoS ONE 2014, 9, e115144. [Google Scholar] [CrossRef][Green Version]
- Xiang, Y.; Stine, Z.E.; Xia, J.; Lu, Y.; O’Connor, R.S.; Altman, B.J.; Hsieh, A.L.; Gouw, A.M.; Thomas, A.G.; Gao, P.; et al. Targeted inhibition of tumor-specific glutaminase diminishes cell-autonomous tumorigenesis. J. Clin. Investig. 2015, 125, 2293–2306. [Google Scholar] [CrossRef]
- Li, B.; Cao, Y.; Meng, G.; Qian, L.; Xu, T.; Yan, C.; Luo, O.; Wang, S.; Wei, J.; Ding, Y.; et al. Targeting glutaminase 1 attenuates stemness properties in hepatocellular carcinoma by increasing reactive oxygen species and suppressing Wnt/beta-catenin pathway. EBioMedicine 2019, 39, 239–254. [Google Scholar] [CrossRef]
- Liao, J.; Liu, P.P.; Hou, G.; Shao, J.; Yang, J.; Liu, K.; Lu, W.; Wen, S.; Hu, Y.; Huang, P. Regulation of stem-like cancer cells by glutamine through beta-catenin pathway mediated by redox signaling. Mol. Cancer 2017, 16, 51. [Google Scholar] [CrossRef]
- Fearon, E.R. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 2011, 6, 479–507. [Google Scholar] [CrossRef]
- Pelicci, P.G.; Dalton, P.; Giorgio, M. The other face of ROS: A driver of stem cell expansion in colorectal cancer. Cell Stem Cell 2013, 12, 635–636. [Google Scholar] [CrossRef]
- Myant, K.B.; Cammareri, P.; McGhee, E.J.; Ridgway, R.A.; Huels, D.J.; Cordero, J.B.; Schwitalla, S.; Kalna, G.; Ogg, E.L.; Athineos, D.; et al. ROS production and NF-kappaB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell 2013, 12, 761–773. [Google Scholar] [CrossRef]
- Suh, H.N.; Kim, M.J.; Jung, Y.S.; Lien, E.M.; Jun, S.; Park, J.I. Quiescence Exit of Tert(+) Stem Cells by Wnt/beta-Catenin Is Indispensable for Intestinal Regeneration. Cell Rep. 2017, 21, 2571–2584. [Google Scholar] [CrossRef]
- Povinelli, B.J.; Nemeth, M.J. Wnt5a regulates hematopoietic stem cell proliferation and repopulation through the Ryk receptor. Stem Cell. 2014, 32, 105–115. [Google Scholar] [CrossRef]
- Zhang, J.; Fan, J.; Zeng, X.; Nie, M.; Luan, J.; Wang, Y.; Ju, D.; Yin, K. Hedgehog signaling in gastrointestinal carcinogenesis and the gastrointestinal tumor microenvironment. Acta Pharm. Sin. B 2021, 11, 609–620. [Google Scholar] [CrossRef]
- Yoon, C.; Park, D.J.; Schmidt, B.; Thomas, N.J.; Lee, H.J.; Kim, T.S.; Janjigian, Y.Y.; Cohen, D.J.; Yoon, S.S. CD44 expression denotes a subpopulation of gastric cancer cells in which Hedgehog signaling promotes chemotherapy resistance. Clin. Cancer Res. 2014, 20, 3974–3988. [Google Scholar] [CrossRef]
- Haque, I.; De, A.; Majumder, M.; Mehta, S.; McGregor, D.; Banerjee, S.K.; Van Veldhuizen, P.; Banerjee, S. The matricellular protein CCN1/Cyr61 is a critical regulator of Sonic Hedgehog in pancreatic carcinogenesis. J. Biol. Chem. 2012, 287, 38569–38579. [Google Scholar] [CrossRef]
- Song, L.; Chen, X.; Gao, S.; Zhang, C.; Qu, C.; Wang, P.; Liu, L. Ski modulate the characteristics of pancreatic cancer stem cells via regulating sonic hedgehog signaling pathway. Tumor Biol. 2016, 37, 16115–16125. [Google Scholar] [CrossRef]
- Zhang, Y.; Xue, X.; Zhao, X.; Qin, L.; Shen, Y.; Dou, H.; Sun, J.; Wang, T.; Yang, D.Q. Vasohibin 2 promotes malignant behaviors of pancreatic cancer cells by inducing epithelial-mesenchymal transition via Hedgehog signaling pathway. Cancer Med. 2018, 7, 5567–5576. [Google Scholar] [CrossRef]
- Liang, Y.; Yang, L.; Xie, J. The Role of the Hedgehog Pathway in Chemoresistance of Gastrointestinal Cancers. Cells 2021, 10, 2030. [Google Scholar] [CrossRef]
- Semenza, G.L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2010, 29, 625–634. [Google Scholar] [CrossRef]
- Dengler, V.L.; Galbraith, M.; Espinosa, J.M. Transcriptional regulation by hypoxia inducible factors. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 1–15. [Google Scholar] [CrossRef]
- Mimeault, M.; Batra, S.K. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J. Cell Mol. Med. 2013, 17, 30–54. [Google Scholar] [CrossRef]
- Di Magno, L.; Manzi, D.; D’Amico, D.; Coni, S.; Macone, A.; Infante, P.; Di Marcotullio, L.; De Smaele, E.; Ferretti, E.; Screpanti, I.; et al. Druggable glycolytic requirement for Hedgehog-dependent neuronal and medul-loblastoma growth. Cell Cycle 2014, 13, 3404–3413. [Google Scholar] [CrossRef]
- Lu, L.; Wu, M.; Zhao, F.; Fu, W.; Li, W.; Li, X.; Liu, T. Prognostic and clinicopathological value of Gli-1 expression in gastric cancer: A meta-analysis. Oncotarget 2016, 7, 69087–69096. [Google Scholar] [CrossRef]
- Tang, C.-T.; Lin, X.-L.; Wu, S.; Liang, Q.; Yang, L.; Gao, Y.-J.; Ge, Z.-Z. NOX4-driven ROS formation regulates proliferation and apoptosis of gastric cancer cells through the GLI1 pathway. Cell. Signal. 2018, 46, 52–63. [Google Scholar] [CrossRef]
- Liu, Z.; Tu, K.; Wang, Y.; Yao, B.; Li, Q.; Wang, L.; Dou, C.; Liu, Q.; Zheng, X. Hypoxia Accelerates Aggressiveness of Hepa-tocellular Carcinoma Cells Involving Oxidative Stress, Epithelial-Mesenchymal Transition and Non-Canonical Hedgehog Signaling. Cell. Physiol. Biochem. 2017, 44, 1856–1868. [Google Scholar] [CrossRef]
- Li, T.; Liao, X.; Lochhead, P.; Morikawa, T.; Yamauchi, M.; Nishihara, R.; Inamura, K.; Kim, S.A.; Mima, K.; Sukawa, Y.; et al. SMO expression in colorectal cancer: Associations with clinical, pathological, and molecular features. Ann. Surg. Oncol. 2014, 21, 4164–4173. [Google Scholar] [CrossRef]
- Xu, M.; Li, X.; Liu, T.; Leng, A.; Zhang, G. Prognostic value of hedgehog signaling pathway in patients with colon cancer. Med. Oncol. 2011, 29, 1010–1016. [Google Scholar] [CrossRef]
- Huang, F.-T.; Zhuan-Sun, Y.-X.; Zhuang, Y.-Y.; Wei, S.-L.; Tang, J.; Chen, W.-B.; Zhang, S.-N. Zhang, Inhibition of hedgehog signaling depresses self-renewal of pancreatic cancer stem cells and reverses chemoresistance. Int. J. Oncol. 2012, 41, 1707–1714. [Google Scholar] [CrossRef]
- Lee, D.-H.; Lee, S.-Y.; Oh, S.C. Hedgehog signaling pathway as a potential target in the treatment of advanced gastric cancer. Tumor Biol. 2017, 39, 1–10. [Google Scholar] [CrossRef]
- Bose, C.; Das, U.; Kuilya, T.K.; Mondal, J.; Bhadra, J.; Banerjee, P.; Goswami, R.K.; Sinha, S. Cananginone Abrogates EMT in Breast Cancer Cells through Hedgehog Signaling. Chem. Biodivers. 2022, 19, e202100823. [Google Scholar] [CrossRef]
- Chang, J.; Guo, C.; Li, J.; Liang, Z.; Wang, Y.; Yu, A.; Liu, R.; Guo, Y.; Chen, J.; Huang, S. EN1 Regulates Cell Growth and Pro-liferation in Human Glioma Cells via Hedgehog Signaling. Int. J. Mol. Sci. 2022, 23, 1123. [Google Scholar] [CrossRef] [PubMed]
- Leung, H.W.; Lau, E.Y.T.; Leung, C.O.N.; Lei, M.M.L.; Mok, E.H.K.; Ma, V.W.S.; Cho, W.C.S.; Ng, I.O.L.; Yun, J.P.; Cai, S.H.; et al. NRF2/SHH signaling cascade promotes tumor-initiating cell lineage and drug resistance in hepa-tocellular carcinoma. Cancer Lett. 2020, 476, 856. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Medina, R.; Le Dreau, G.; Ros, M.; Martí, E. Hedgehog activation is required upstream of Wnt signalling to control neural progenitor proliferation. Development 2009, 136, 3301–3309. [Google Scholar] [CrossRef] [PubMed]
- Guo, Q.; Jing, F.J.; Qu, H.J.; Xu, W.; Han, B.; Xing, X.M.; Ji, H.Y.; Jing, F.B. Ubenimex Reverses MDR in Gastric Cancer Cells by Activating Caspase-3-Mediated Apoptosis and Suppressing the Expression of Membrane Transport Proteins. Biomed Res. Int. 2019, 2019, 4390839. [Google Scholar] [CrossRef] [PubMed]
- Guo, Q.; Jing, F.J.; Xu, W.; Li, X.; Li, X.; Sun, J.L.; Xing, X.M.; Zhou, C.K.; Jing, F.B. Ubenimex induces autophagy inhibition and EMT suppression to overcome cisplatin resistance in GC cells by perturbing the CD13/EMP3/PI3K/AKT/NF-kappaB axis. Aging 2019, 12, 80–105. [Google Scholar] [CrossRef]
- Guo, Q.; Sui, Z.G.; Xu, W.; Quan, X.H.; Sun, J.L.; Li, X.; Ji, H.Y.; Jing, F.B. Ubenimex suppresses Pim-3 kinase expression by targeting CD13 to reverse MDR in HCC cells. Oncotarget 2017, 8, 72652–72665. [Google Scholar] [CrossRef]
- Liu, Y.C.; Yeh, C.T.; Lin, K.H. Cancer Stem Cell Functions in Hepatocellular Carcinoma and Comprehensive Therapeutic Strategies. Cells 2020, 9, 1331. [Google Scholar] [CrossRef]
- Yamashita, M.; Wada, H.; Eguchi, H.; Ogawa, H.; Yamada, D.; Noda, T.; Asaoka, T.; Kawamoto, K.; Gotoh, K.; Umeshita, K.; et al. A CD13 inhibitor, ubenimex, synergistically enhances the effects of anticancer drugs in hepatocellular carcinoma. Int. J. Oncol. 2016, 49, 89–98. [Google Scholar] [CrossRef]
- Vu, N.B.; Nguyen, T.T.; Tran, L.C.; Do, C.D.; Nguyen, B.H.; Phan, N.K.; Pham, P.V. Doxorubicin and 5-fluorouracil resistant hepatic cancer cells demonstrate stem-like properties. Cytotechnology 2013, 65, 491–503. [Google Scholar] [CrossRef]
- Sun, L.; Zhang, L.; Chen, J.; Li, C.; Sun, H.; Wang, J.; Xiao, H. Activation of Tyrosine Metabolism in CD13+ Cancer Stem Cells Drives Relapse in Hepatocellular Carcinoma. Cancer Res. Treat. 2020, 52, 604–621. [Google Scholar] [CrossRef]
- Lu, C.; Amin, M.A.; Fox, D.A. CD13/Aminopeptidase N Is a Potential Therapeutic Target for Inflammatory Disorders. J. Immunol. 2020, 204, 3–11. [Google Scholar] [CrossRef] [PubMed]
- Hashida, H.; Takabayashi, A.; Kanai, M.; Adachi, M.; Kondo, K.; Kohno, N.; Yamaoka, Y.; Miyake, M. Aminopeptidase N is involved in cell motility and angiogenesis: Its clinical significance in human colon cancer. Gastroenterology 2002, 122, 376–386. [Google Scholar] [CrossRef] [PubMed]
- Bhagwat, S.V.; Lahdenranta, J.; Giordano, R.; Arap, W.; Pasqualini, R.; Shapiro, L.H. CD13/APN is activated by angiogenic signals and is essential for capillary tube formation. Blood 2001, 97, 652–659. [Google Scholar] [CrossRef] [PubMed]
- Cui, S.X.; Zhang, H.L.; Xu, W.F.; Qu, X.J. 13F-1, a novel 5-fluorouracil prodrug containing an Asn-Gly-Arg (NO2) COOCH3 tripeptide, inhibits human colonic carcinoma growth by targeting Aminopeptidase N (APN/CD13). Eur. J. Pharmacol. 2014, 734, 50–59. [Google Scholar] [CrossRef]
- Pasqualini, R.; Koivunen, E.; Kain, R.; Lahdenranta, J.; Sakamoto, M.; Stryhn, A.; Ashmun, R.A.; Shapiro, L.H.; Arap, W.; Ruoslahti, E. Aminopeptidase N is a receptor for tumor-homing peptides and a target for inhibiting angiogenesis. Cancer Res. 2000, 60, 722–727. [Google Scholar]
- Seidi, K.; Jahanban-Esfahlan, R.; Monhemi, H.; Zare, P.; Minofar, B.; Daei Farshchi Adli, A.; Farajzadeh, D.; Behzadi, R.; Mesgari Abbasi, M.; Neubauer, H.A.; et al. NGR (Asn-Gly-Arg)-targeted delivery of coagulase to tumor vasculature arrests cancer cell growth. Oncogene 2018, 37, 3967–3980. [Google Scholar] [CrossRef]
- Meng, Y.; Zhang, Z.; Liu, K.; Ye, L.; Liang, Y.; Gu, W. Aminopeptidase N (CD13) targeted MR and NIRF dual-modal imaging of ovarian tumor xenograft. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 93, 968–974. [Google Scholar] [CrossRef]
- Kanamori, H.; Takasaki, H.; Takabayashi, M.; Yamaji, S.; Koharazawa, H.; Fujimaki, K.; Taguchi, J.; Ishigatsubo, Y. Long-term cytogenetic remission with ubenimex monotherapy in a case of chronic myeloid leukemia. Anticancer Drug. 2004, 15, 729–731. [Google Scholar] [CrossRef]
- Fujii, H.; Yosizawa, K.; Maruyama, S.; Abe, F. Growth inhibitory effects of ubenimex on leukemic cell lines resistant to chemotherapeutic agents. Jpn. J. Antibiot. 1996, 49, 1109–1115. [Google Scholar]
- Usuka, Y.; Saito, Y. Bestatin treatment of myelodysplastic syndromes and chronic myelogenous leukemia. Biomed. Pharmacother. 1991, 45, 87–93. [Google Scholar] [CrossRef]
- Cardinale, V.; Renzi, A.; Carpino, G.; Torrice, A.; Bragazzi, M.C.; Giuliante, F.; DeRose, A.M.; Fraveto, A.; Onori, P.; Napoletano, C.; et al. Profiles of cancer stem cell subpopulations in cholangiocarcinomas. Am. J. Pathol. 2015, 185, 1724–1739. [Google Scholar] [CrossRef] [PubMed]
- Castelli, G.; Pelosi, E.; Testa, U. Liver Cancer: Molecular Characterization, Clonal Evolution and Cancer Stem Cells. Cancers 2017, 9, 127. [Google Scholar] [CrossRef] [PubMed]
- Haraguchi, N.; Inoue, H.; Tanaka, F.; Mimori, K.; Utsunomiya, T.; Sasaki, A.; Mori, M. Cancer stem cells in human gastrointestinal cancers. Hum. Cell 2006, 19, 24–29. [Google Scholar] [CrossRef]
- Mu, X.; Espanol-Suner, R.; Mederacke, I.; Affo, S.; Manco, R.; Sempoux, C.; Lemaigre, F.P.; Adili, A.; Yuan, D.; Weber, A.; et al. Hepatocellular carcinoma originates from hepatocytes and not from the progenitor/biliary compartment. J. Clin. Investig. 2015, 125, 3891–3903. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Fang, C.; Qu, M.; Wu, H.; Wang, X.; Zhang, H.; Ma, H.; Zhang, Z.; Huang, Y.; Shi, L.; et al. CD13 Inhibition Enhances Cytotoxic Effect of Chemotherapy Agents. Front. Pharmacol. 2018, 9, 1042. [Google Scholar] [CrossRef]
- Xing, X.; Li, F.; Hu, Y.; Zhang, L.; Hui, Q.; Qin, H.; Jiang, Q.; Jiang, W.; Fang, C.; Zhang, L. Discovery of Novel Tetrahydro-beta-carboline Containing Aminopeptidase N Inhibitors as Cancer Chemosensitizers. Front. Oncol. 2022, 12, 894842. [Google Scholar] [CrossRef]
- Sun, Z.P.; Zhang, J.; Shi, L.H.; Zhang, X.R.; Duan, Y.; Xu, W.F.; Dai, G.; Wang, X.J. Aminopeptidase N inhibitor 4cc synergizes antitumor effects of 5-fluorouracil on human liver cancer cells through ROS-dependent CD13 inhibition. Biomed. Pharmacother. 2015, 76, 65–72. [Google Scholar] [CrossRef]
- Hu, B.; Xu, Y.; Li, Y.C.; Huang, J.F.; Cheng, J.W.; Guo, W.; Yin, Y.; Gao, Y.; Wang, P.X.; Wu, S.Y.; et al. CD13 promotes hepatocellular carcinogenesis and sorafenib resistance by activating HDAC5-LSD1-NF-kappaB oncogenic signaling. Clin. Transl. Med. 2020, 10, e233. [Google Scholar] [CrossRef]
- Carballo, G.B.; Honorato, J.R.; de Lopes, G.P.F.; Spohr, T. A highlight on Sonic hedgehog pathway. Cell Commun. Signal. 2018, 16, 11. [Google Scholar] [CrossRef]
- Laukkanen, M.O.; Castellone, M.D. Hijacking the Hedgehog Pathway in Cancer Therapy. Anticancer Agent. Med. Chem. 2016, 16, 309–317. [Google Scholar] [CrossRef]
- Cui, Q.; Wang, J.Q.; Assaraf, Y.G.; Ren, L.; Gupta, P.; Wei, L.; Ashby, C.R., Jr.; Yang, D.H.; Chen, Z.S. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist. Updat. 2018, 41, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Kesavardhana, S.; Kanneganti, T.D. Stressed-out ROS take a silent death route. Nat. Immunol. 2018, 19, 103–105. [Google Scholar] [CrossRef] [PubMed]
- Lendeckel, U.; Kahne, T.; Arndt, M.; Frank, K.; Ansorge, S. Inhibition of alanyl aminopeptidase induces MAP-kinase p42/ERK2 in the human T cell line KARPAS-299. Biochem. Biophys. Res. Commun. 1998, 252, 5–9. [Google Scholar] [CrossRef] [PubMed]
- Nishikawa, S.; Ishii, H.; Haraguchi, N.; Kano, Y.; Fukusumi, T.; Ohta, K.; Ozaki, M.; Sakai, D.; Satoh, T.; Nagano, H.; et al. Genotoxic therapy stimulates error-prone DNA repair in dormant hepatocellular cancer stem cells. Exp. Ther. Med. 2012, 3, 959–962. [Google Scholar] [CrossRef][Green Version]
- Haraguchi, N.; Ishii, H.; Nagano, H.; Doki, Y.; Mori, M. The future prospects and subject of the liver cancer stem cells study for the clinical application. Gastroenterology 2011, 140, 1355. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lendeckel, U.; Wolke, C. Redox-Regulation in Cancer Stem Cells. Biomedicines 2022, 10, 2413. https://doi.org/10.3390/biomedicines10102413
Lendeckel U, Wolke C. Redox-Regulation in Cancer Stem Cells. Biomedicines. 2022; 10(10):2413. https://doi.org/10.3390/biomedicines10102413
Chicago/Turabian StyleLendeckel, Uwe, and Carmen Wolke. 2022. "Redox-Regulation in Cancer Stem Cells" Biomedicines 10, no. 10: 2413. https://doi.org/10.3390/biomedicines10102413
APA StyleLendeckel, U., & Wolke, C. (2022). Redox-Regulation in Cancer Stem Cells. Biomedicines, 10(10), 2413. https://doi.org/10.3390/biomedicines10102413