Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1648 2022-12-15 11:14:07 |
2 format correct + 3 word(s) 1651 2022-12-19 06:04:40 | |
3 format correct Meta information modification 1651 2022-12-19 06:13:58 | |
4 format correct + 2 word(s) 1653 2022-12-22 04:51:43 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Iacono, M.L.;  Gaggianesi, M.;  Bianca, P.;  Brancato, O.R.;  Muratore, G.;  Modica, C.;  Roozafzay, N.;  Shams, K.;  Colarossi, L.;  Colarossi, C.; et al. CSCs is The Main Players in Drug Resistance. Encyclopedia. Available online: https://encyclopedia.pub/entry/38817 (accessed on 04 July 2024).
Iacono ML,  Gaggianesi M,  Bianca P,  Brancato OR,  Muratore G,  Modica C, et al. CSCs is The Main Players in Drug Resistance. Encyclopedia. Available at: https://encyclopedia.pub/entry/38817. Accessed July 04, 2024.
Iacono, Melania Lo, Miriam Gaggianesi, Paola Bianca, Ornella Roberta Brancato, Giampaolo Muratore, Chiara Modica, Narges Roozafzay, Kimiya Shams, Lorenzo Colarossi, Cristina Colarossi, et al. "CSCs is The Main Players in Drug Resistance" Encyclopedia, https://encyclopedia.pub/entry/38817 (accessed July 04, 2024).
Iacono, M.L.,  Gaggianesi, M.,  Bianca, P.,  Brancato, O.R.,  Muratore, G.,  Modica, C.,  Roozafzay, N.,  Shams, K.,  Colarossi, L.,  Colarossi, C.,  Memeo, L.,  Turdo, A.,  Veschi, V.,  Franco, S.D.,  Todaro, M., & Stassi, G. (2022, December 15). CSCs is The Main Players in Drug Resistance. In Encyclopedia. https://encyclopedia.pub/entry/38817
Iacono, Melania Lo, et al. "CSCs is The Main Players in Drug Resistance." Encyclopedia. Web. 15 December, 2022.
CSCs is The Main Players in Drug Resistance
Edit

Drug resistance is doubtless the main challenge of treatment in cancer patients. It is possible to distinguish two categories of drug resistance: intrinsic resistance and acquired resistance after drug treatment. Compelling evidence highlights that intratumoral heterogeneity is one of the major hurdles involved in intrinsic drug resistance, in which the cancer stem cells (CSCs) represent the main players due to their self-renewal and differentiation abilities. 

natural products cancer stem cells drug resistance alkaloids

1. Introduction

Despite prominent advances in the field of cancer prevention and early diagnosis, it is expected that one in five people will develop cancer during their lifespan. One of the greatest challenges in translational oncology is limiting the onset of primary or acquired drug resistance, which is boosted by cancer stem cells (CSCs). CSCs represent a pluripotent heterogeneous population within tumor bulk, with self-renewal and differentiation abilities, contributing to the failure of conventional therapies and, therefore, to disease relapse and metastasis [1]. Increasing evidence points out that different natural products (NPs) can modulate the CSCs’ hallmarks and sensitize them to conventional treatment [2]. NPs show minimal side effects in comparison with chemotherapeutics, and many studies have demonstrated their emerging role as adjuvant agents in cancer treatment.

2. Cancer Stem Cells: The Main Players in Drug Resistance

Drug resistance is doubtless the main challenge of treatment in cancer patients. It is possible to distinguish two categories of drug resistance: intrinsic resistance and acquired resistance after drug treatment [3]. Compelling evidence highlights that intratumoral heterogeneity is one of the major hurdles involved in intrinsic drug resistance, in which the CSCs represent the main players due to their self-renewal and differentiation abilities [4][5]. The presence of CSCs has been characterized in different tumors, such as thyroid, colorectal, breast, prostate, and other solid tumors [6][7]. CSCs are identified and can be isolated by the expression of specific surface markers such as CD133 [8], CD44 [9], CD44v6 [10], EpCAM [11], or enzyme activity such as aldehyde dehydrogenase (ALDH) [12]. CSCs are also defined as tumor-initiating cells, as they can generate tumor xenografts in immunocompromised mice models [13]. Moreover, the failure of conventional therapies, based on the use of radiotherapy and chemotherapy to induce DNA damage in highly proliferative cells and to eradicate tumor mass, is strictly due to the presence of CSCs, which are characterized by multiple survival mechanisms [14]. In particular, the mechanisms through which CSCs escape chemotherapeutic treatments are different, such as i) drug export (the aberrant expression of ATP-binding cassette, ABC, drug pumps); ii) high survival (the inhibition of antiapoptotic processes, the high expression of proteins involved in DNA-damage repair, high telomerase activity); iii) reactive oxygen species (ROS) decrease (high ALDH activity, high expression of detoxification enzymes);and iv) the aberrant activation of pathways involved in stemness [15][16].

2.1. Drug Export in CSCs

It is common knowledge that the high expression of ABC proteins contributes to chemotherapy resistance and that CSCs overexpress different drug-transporter pumps, including ABCB1, ABCG2, and ABCC1 [15][17]. The Hoechst 33342 side population assay is a useful method to identify and isolate the CSC subpopulation in solid and hematopoietic tumors [18]. Yin et al. reported that CD133+ EPCAM+ liver CSCs express high levels of ABCG2 and ABCB1 and are highly resistant to doxorubicin treatment. The use of specific ABC inhibitors increases doxorubicin intracellular efflux, decreasing the sphere-forming capacity and viability of CSCs [19]. Other ABC transporters, including multidrug resistance protein 1 (MRP1, ABCC1), breast cancer resistance protein (BCRP), and MRP5/ABCC5, are reported as multidrug resistance transporters in solid and hematopoietic tumors [20][21][22]. Moreover, CD133+ melanoma CSCs expressed higher levels of ABCB5 compared to CD113 cells and are resistant to the antiapoptotic activity of the natural compound caffeic acid phenethyl ester [23]. In the lung, the high expression levels of ABCB1 in CSCs mediated the resistance to PHA-665752 and crizotinib, a MET inhibitor [24]. Although the targeting of ABC transporters could be an effective strategy to target CSCs, the use of specific inhibitors causes many side effects, due to the expression of the same targets in normal cells, as well [24].

2.2. Enhanced Survival Ability in CSCs

CSCs can also circumvent the toxic effects induced by chemotherapeutic treatment activating DNA damage response (DDR) by the ATM(ataxia-telangiectasia-mutated)- and ATR (ATM- and RAD3-related)-dependent phosphorylation of targets such as Check-1, Check-2, or H2A.X (known as γH2A.X when phosphorylated) [25]. Manic and co-workers demonstrated that in colorectal CSCs the treatment with chemotherapeutic agents induces the activation of the DDR players, such as PARP1, RAD51, and/or MRE11, resulting in higher DNA damage repair machinery [26]. In breast cancer, both BRCA1wt and BRCA1mut CSCs were highly resistant to PARP inhibitors, due to the high expression of Rad51 and Sam68, and the inhibition of this critical signaling axis hampered CSC viability [27][28]. In addition, CD133+ glioma CSCs displayed resistance to radiotherapy treatment by the activation of DNA-damage repair mechanisms, where Check-1 and Check-2 are the main players. The inhibition of these two effectors reverted the radioresistance in glioma-CSCs, suggesting that targeting DNA damage could be a promising therapeutic approach for brain cancer treatment [29].
The deregulation of apoptotic pathways is another mechanism underlying CSC-mediated chemoresistance. A weak expression of death receptors, such as TRAIL and FAS, and the overexpression of inhibitor apoptosis proteins (IAPs) have been described in CSCs compared to differentiated tumor cells [6][30]. In CSCs, IAPs are often overexpressed and impair the activation of the apoptosis cascade by mediating pro-apoptotic protein degradation [31]. CD133+ colorectal CSCs highly resistant to 5-fluorouracil (5-FU) treatment expressed high levels of SURVIVIN, and the use of a specific aptamer-SURVIVIN siRNA enhanced the in vitro and in vivo 5-FU efficacy [32][33]. Moreover, in nasopharyngeal CSCs, XIAPs increased the stability of SOX2, and the use of an inhibitor of the IAP family in combination with 5-FU impaired tumor growth [34]. It has been reported that an aberrant expression of BCL-2 family members in CSCs contributes to drug resistance [20][35]. BCL-2 is overexpressed in leukemia stem cells and the use of a specific inhibitor of BCL-2, venetoclax, combined with azacitidine, resulted in disease remission in acute myeloid leukemia by CSC targeting [36][37]. In gastro-esophageal cancers, the use of the small molecule AT-101, which inhibits the BCL-2 family, decreased the expression of CSC markers (YAP1/SOX9) [38] (NCT00561197).
One of the effects of radiotherapy is the induction of DNA damage through the production of ROS and water-derived radicals. In CSCs, the presence of ROS is dramatically reduced due to the increase in ROS scavengers, limiting apoptosis induction and DDR mechanism activation [39]. The CSCs can also escape from anticancer therapies by increasing aldehyde dehydrogenase (ALDH) activity, which acts by reducing intracellular ROS levels. In turn, ROS generated from radio- and chemotherapy enhance the cytosolic expression of aldehydes, such as ALDH1A and 3A1 [40]. High levels of drug-metabolizing enzymes, such as ALDH1a1 and bleomycin hydrolase (BLMH1), have been characterized in the secretome of colorectal CSCs, increasing chemoresistance [41]. Moreover, several studies have pointed out that CSCs isolated from different tumor types display high ALDH expression levels and activity, which boost their chemoresistance [12][42]. Therefore, the upregulation of ROS levels could be an efficient strategy to counteract CSC features and sensitize CSCs to treatments.

2.3. Stemness Induction in CSCs by Different Signaling Pathways

Several signaling pathways, among which are Notch, Sonic-Hedgehog (SHH), Wnt/β-catenin, PI3K/Akt/mTOR (mTORC1 and mTORC2), TGF-β, JACK/STAT, and Hippo-YAP/TAZ, are aberrantly activated or deregulated in CSCs compared to normal stem cells [43].
The Wnt signaling pathway plays a key role during embryogenesis, and in many cancers, such as breast, colorectal, thyroid, and esophageal cancers, its activation promotes CSC growth and chemoresistance [43][44]. It has been demonstrated that the Wnt pathway is crucial for the maintenance of intestinal crypt homeostasis, and the APC mutation in transgenic mice increased the presence of LGR5+ stem cells at the bottom of crypts, boosting the transformation in microadenoma [45]. Vermeulen et al. demonstrated that the Wnt pathway is highly activated in CD133+ colorectal CSCs and can be influenced by extrinsic factors secreted by TME cells [46]. In hepatocellular carcinoma, the activation of the Wnt signaling pathway, induced by protein tyrosine kinase-2 (PTK2), boosted CSC tumorigenic potential and contributed to sorafenib resistance [47]. In endometrial CSCs, Lu and co-workers showed that SPARC-related modular calcium binding 2 (SMOC-2) interacts with Fzd6 and LRP6 (LDL-receptor-related protein 6) receptors and activates the Wnt/β-catenin pathway, increasing cisplatin and placlitaxel resistance [48].
The SHH signaling pathway is involved in normal embryogenesis development and plays a key role in the promotion of tumor growth and in drug resistance, upregulating the genes involved in CSC maintenance, such as CD44, CCND2, c-MYC, NANOG, OCT4, and ALDH1 [49][50]. The SHH signaling pathway is involved in chemoresistance mechanisms by the modulation of the ABCG2 transporter and ALDH activity [51][52].
The dual TGF-β role in tumor progression has been extensively studied [53]. In fact, TGF-β is a key regulator of stemness, promoting EMT and radio-/chemoresistance [54][55]. Moreover, it has been shown that the cooperation of TGF-β with other signaling pathways increases CSC features. TGF-β and tumor necrosis factor alpha (TNF-α) induced a mesenchymal phenotype in breast CSCs by decreasing CLDN3-4-7 gene expression and, in turn, increasing in vivo tumorigenesis and resistance to oxaliplatin, etoposide, and paclitaxel [56]. In leukemia stem cells, TGF-β regulated the activation of AKT and induced FOXO3a nuclear localization, boosting sphere-forming ability and tumor growth [57].
In addition to the mechanisms described above, other intrinsic and extrinsic factors contribute to drug resistance in CSCs. Increasing evidence sheds new light on the role of epigenetic alterations in increasing intratumoral heterogeneity and in the failure of standard therapies [58]. Several molecular mechanisms, such as DNA methylation, chromatin remodeling, regulation by non-coding RNAs, and the modification of histone proteins, contribute to the aberrant expression of ABC transporters in solid and hematological tumors [1][59]. Furthermore, numerous studies point out that the crosstalk between CSCs and the tumor microenvironment (TME) influences the plasticity of CSCs, promoting drug resistance [6][60].
To overcome this challenge in cancer treatment, many researchers have focused on the development of therapeutic approaches targeting CSCs and the different mechanisms involved in drug resistance. In this regard, NPs could be considered eligible candidates.

References

  1. Turdo, A.; Veschi, V.; Gaggianesi, M.; Chinnici, A.; Bianca, P.; Todaro, M.; Stassi, G. Meeting the Challenge of Targeting Cancer Stem Cells. Front. Cell Dev. Biol. 2019, 7, 16.
  2. Moselhy, J.; Srinivasan, S.; Ankem, M.K.; Damodaran, C. Natural Products That Target Cancer Stem Cells. Anticancer Res. 2015, 35, 5773–5788.
  3. Schmidt, F.; Efferth, T. Tumor Heterogeneity, Single-Cell Sequencing, and Drug Resistance. Pharmaceuticals 2016, 9, 33.
  4. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem. Cells Int. 2018, 2018, 5416923.
  5. Hausser, J.; Alon, U. Tumour heterogeneity and the evolutionary trade-offs of cancer. Nat. Rev. Cancer 2020, 20, 247–257.
  6. Gaggianesi, M.; Di Franco, S.; Pantina, V.D.; Porcelli, G.; D’Accardo, C.; Verona, F.; Veschi, V.; Colarossi, L.; Faldetta, N.; Pistone, G.; et al. Messing Up the Cancer Stem Cell Chemoresistance Mechanisms Supported by Tumor Microenvironment. Front. Oncol. 2021, 11, 702642.
  7. Veschi, V.; Verona, F.; Lo Iacono, M.; D’Accardo, C.; Porcelli, G.; Turdo, A.; Gaggianesi, M.; Forte, S.; Giuffrida, D.; Memeo, L.; et al. Cancer Stem Cells in Thyroid Tumors: From the Origin to Metastasis. Front. Endocrinol. 2020, 11, 566.
  8. Ricci-Vitiani, L.; Lombardi, D.G.; Pilozzi, E.; Biffoni, M.; Todaro, M.; Peschle, C.; De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007, 445, 111–115.
  9. 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.
  10. Todaro, M.; Gaggianesi, M.; Catalano, V.; Benfante, A.; Iovino, F.; Biffoni, M.; Apuzzo, T.; Sperduti, I.; Volpe, S.; Cocorullo, G.; et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 2014, 14, 342–356.
  11. Loening-Baucke, V. Lichen sclerosus et atrophicus in children. Am. J. Dis. Child. 1991, 145, 1058–1061.
  12. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007, 1, 555–567.
  13. Clarke, M.F.; Dick, J.E.; Dirks, P.B.; Eaves, C.J.; Jamieson, C.H.; Jones, D.L.; Visvader, J.; Weissman, I.L.; Wahl, G.M. Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006, 66, 9339–9344.
  14. Zhou, H.M.; Zhang, J.G.; Zhang, X.; Li, Q. Targeting cancer stem cells for reversing therapy resistance: Mechanism, signaling, and prospective agents. Signal Transduct. Target Ther. 2021, 6, 62.
  15. Begicevic, R.R.; Falasca, M. ABC Transporters in Cancer Stem Cells: Beyond Chemoresistance. Int. J. Mol. Sci. 2017, 18, 2362.
  16. Rezayatmand, H.; Razmkhah, M.; Razeghian-Jahromi, I. Drug resistance in cancer therapy: The Pandora’s Box of cancer stem cells. Stem Cell Res.Ther. 2022, 13, 181.
  17. Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Rev. Cancer 2010, 10, 147–156.
  18. 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.
  19. Yin, W.; Xiang, D.; Wang, T.; Zhang, Y.; Pham, C.V.; Zhou, S.; Jiang, G.; Hou, Y.; Zhu, Y.; Han, Y.; et al. The inhibition of ABCB1/MDR1 or ABCG2/BCRP enables doxorubicin to eliminate liver cancer stem cells. Sci. Rep. 2021, 11, 10791.
  20. Safa, A.R. Resistance to drugs and cell death in cancer stem cells (CSCs). J. Transl. Sci. 2020, 6, 341.
  21. Lu, J.F.; Pokharel, D.; Bebawy, M. MRP1 and its role in anticancer drug resistance. Drug Metab. Rev. 2015, 47, 406–419.
  22. Jaramillo, A.C.; Cloos, J.; Lemos, C.; Stam, R.W.; Kaspers, G.J.L.; Jansen, G.; Peters, G.J. Ex vivo resistance in childhood acute lymphoblastic leukemia: Correlations between BCRP, MRP1, MRP4 and MRP5 ABC transporter expression and intracellular methotrexate polyglutamate accumulation. Leuk. Res. 2019, 79, 45–51.
  23. El-Khattouti, A.; Sheehan, N.T.; Monico, J.; Drummond, H.A.; Haikel, Y.; Brodell, R.T.; Megahed, M.; Hassan, M. CD133(+) melanoma subpopulation acquired resistance to caffeic acid phenethyl ester-induced apoptosis is attributed to the elevated expression of ABCB5: Significance for melanoma treatment. Cancer Lett. 2015, 357, 83–104.
  24. Sugano, T.; Seike, M.; Noro, R.; Soeno, C.; Chiba, M.; Zou, F.; Nakamichi, S.; Nishijima, N.; Matsumoto, M.; Miyanaga, A.; et al. Inhibition of ABCB1 Overcomes Cancer Stem Cell-like Properties and Acquired Resistance to MET Inhibitors in Non-Small Cell Lung Cancer. Mol. Cancer Ther. 2015, 14, 2433–2440.
  25. Maugeri-Sacca, M.; Bartucci, M.; De Maria, R. DNA damage repair pathways in cancer stem cells. Mol. Cancer Ther. 2012, 11, 1627–1636.
  26. Manic, G.; Musella, M.; Corradi, F.; Sistigu, A.; Vitale, S.; Soliman Abdel Rehim, S.; Mattiello, L.; Malacaria, E.; Galassi, C.; Signore, M.; et al. Control of replication stress and mitosis in colorectal cancer stem cells through the interplay of PARP1, MRE11 and RAD51. Cell Death Differ. 2021, 28, 2060–2082.
  27. Liu, Y.; Burness, M.L.; Martin-Trevino, R.; Guy, J.; Bai, S.; Harouaka, R.; Brooks, M.D.; Shang, L.; Fox, A.; Luther, T.K.; et al. RAD51 Mediates Resistance of Cancer Stem Cells to PARP Inhibition in Triple-Negative Breast Cancer. Clin. Cancer Res. 2017, 23, 514–522.
  28. Turdo, A.; Gaggianesi, M.; Di Franco, S.; Veschi, V.; D’Accardo, C.; Porcelli, G.; Lo Iacono, M.; Pillitteri, I.; Verona, F.; Militello, G.; et al. Effective targeting of breast cancer stem cells by combined inhibition of Sam68 and Rad51. Oncogene 2022, 41, 2196–2209.
  29. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760.
  30. Grzybowska-Izydorczyk, O.; Cebula, B.; Robak, T.; Smolewski, P. Expression and prognostic significance of the inhibitor of apoptosis protein (IAP) family and its antagonists in chronic lymphocytic leukaemia. Eur. J. Cancer 2010, 46, 800–810.
  31. Eckelman, B.P.; Salvesen, G.S.; Scott, F.L. Human inhibitor of apoptosis proteins: Why XIAP is the black sheep of the family. EMBO Rep. 2006, 7, 988–994.
  32. Lee, M.R.; Ji, S.Y.; Mia-Jan, K.; Cho, M.Y. Chemoresistance of CD133(+) colon cancer may be related with increased survivin expression. Biochem. Biophys. Res. Commun. 2015, 463, 229–234.
  33. AlShamaileh, H.; Wang, T.; Xiang, D.; Yin, W.; Tran, P.H.; Barrero, R.A.; Zhang, P.Z.; Li, Y.; Kong, L.; Liu, K.; et al. Aptamer-mediated survivin RNAi enables 5-fluorouracil to eliminate colorectal cancer stem cells. Sci. Rep. 2017, 7, 5898.
  34. Ji, J.; Yu, Y.; Li, Z.L.; Chen, M.Y.; Deng, R.; Huang, X.; Wang, G.F.; Zhang, M.X.; Yang, Q.; Ravichandran, S.; et al. XIAP Limits Autophagic Degradation of Sox2 and Is A Therapeutic Target in Nasopharyngeal Carcinoma Stem Cells. Theranostics 2018, 8, 1494–1510.
  35. Wang, Y.H.; Scadden, D.T. Harnessing the apoptotic programs in cancer stem-like cells. EMBO Rep. 2015, 16, 1084–1098.
  36. Lagadinou, E.D.; Sach, A.; Callahan, K.; Rossi, R.M.; Neering, S.J.; Minhajuddin, M.; Ashton, J.M.; Pei, S.; Grose, V.; O’Dwyer, K.M.; et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell 2013, 12, 329–341.
  37. Pollyea, D.A.; Stevens, B.M.; Jones, C.L.; Winters, A.; Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Culp-Hill, R.; Riemondy, K.A.; Gillen, A.E.; et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018, 24, 1859–1866.
  38. Song, S.; Chen, Q.; Li, Y.; Lei, G.; Scott, A.; Huo, L.; Li, C.Y.; Estrella, J.S.; Correa, A.; Pizzi, M.P.; et al. Targeting cancer stem cells with a pan-BCL-2 inhibitor in preclinical and clinical settings in patients with gastroesophageal carcinoma. Gut 2021, 70, 2238–2248.
  39. 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.
  40. Zanoni, M.; Bravaccini, S.; Fabbri, F.; Arienti, C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front. Med. 2022, 9, 795762.
  41. Emmink, B.L.; Verheem, A.; Van Houdt, W.J.; Steller, E.J.; Govaert, K.M.; Pham, T.V.; Piersma, S.R.; Borel Rinkes, I.H.; Jimenez, C.R.; Kranenburg, O. The secretome of colon cancer stem cells contains drug-metabolizing enzymes. J. Proteomics 2013, 91, 84–96.
  42. Gan, C.; Pierscianek, D.; El Hindy, N.; Ahmadipour, Y.; Keyvani, K.; Sure, U.; Zhu, Y. The predominant expression of cancer stem cell marker ALDH1A3 in tumor infiltrative area is associated with shorter overall survival of human glioblastoma. BMC Cancer 2020, 20, 672.
  43. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target Ther. 2020, 5, 8.
  44. De Sousa, E.M.F.; Vermeulen, L. Wnt Signaling in Cancer Stem Cell Biology. Cancers 2016, 8, 60.
  45. Barker, N.; Ridgway, R.A.; van Es, J.H.; van de Wetering, M.; Begthel, H.; van den Born, M.; Danenberg, E.; Clarke, A.R.; Sansom, O.J.; Clevers, H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 2009, 457, 608–611.
  46. Vermeulen, L.; De Sousa, E.M.F.; van der Heijden, M.; Cameron, K.; de Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476.
  47. Fan, Z.; Duan, J.; Wang, L.; Xiao, S.; Li, L.; Yan, X.; Yao, W.; Wu, L.; Zhang, S.; Zhang, Y.; et al. PTK2 promotes cancer stem cell traits in hepatocellular carcinoma by activating Wnt/beta-catenin signaling. Cancer Lett. 2019, 450, 132–143.
  48. Lu, H.; Ju, D.D.; Yang, G.D.; Zhu, L.Y.; Yang, X.M.; Li, J.; Song, W.W.; Wang, J.H.; Zhang, C.C.; Zhang, Z.G.; et al. Targeting cancer stem cell signature gene SMOC-2 Overcomes chemoresistance and inhibits cell proliferation of endometrial carcinoma. EBioMedicine 2019, 40, 276–289.
  49. 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.
  50. Zhu, R.; Gires, O.; Zhu, L.; Liu, J.; Li, J.; Yang, H.; Ju, G.; Huang, J.; Ge, W.; Chen, Y.; et al. TSPAN8 promotes cancer cell stemness via activation of sonic Hedgehog signaling. Nat. Commun. 2019, 10, 2863.
  51. Bai, X.Y.; Zhang, X.C.; Yang, S.Q.; An, S.J.; Chen, Z.H.; Su, J.; Xie, Z.; Gou, L.Y.; Wu, Y.L. Blockade of Hedgehog Signaling Synergistically Increases Sensitivity to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small-Cell Lung Cancer Cell Lines. PLoS ONE 2016, 11, e0149370.
  52. Zhang, M.; Mathur, A.; Zhang, Y.; Xi, S.; Atay, S.; Hong, J.A.; Datrice, N.; Upham, T.; Kemp, C.D.; Ripley, R.T.; et al. Mithramycin represses basal and cigarette smoke-induced expression of ABCG2 and inhibits stem cell signaling in lung and esophageal cancer cells. Cancer Res. 2012, 72, 4178–4192.
  53. Massague, J. TGFbeta in Cancer. Cell 2008, 134, 215–230.
  54. Tang, Y.A.; Chen, Y.F.; Bao, Y.; Mahara, S.; Yatim, S.; Oguz, G.; Lee, P.L.; Feng, M.; Cai, Y.; Tan, E.Y.; et al. Hypoxic tumor microenvironment activates GLI2 via HIF-1alpha and TGF-beta2 to promote chemoresistance in colorectal cancer. Proc. Natl. Acad. Sci. USA 2018, 115, E5990–E5999.
  55. Akhurst, R.J.; Hata, A. Targeting the TGFbeta signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811.
  56. Asiedu, M.K.; Ingle, J.N.; Behrens, M.D.; Radisky, D.C.; Knutson, K.L. TGFbeta/TNF(alpha)-mediated epithelial-mesenchymal transition generates breast cancer stem cells with a claudin-low phenotype. Cancer Res. 2011, 71, 4707–4719.
  57. Naka, K.; Hoshii, T.; Muraguchi, T.; Tadokoro, Y.; Ooshio, T.; Kondo, Y.; Nakao, S.; Motoyama, N.; Hirao, A. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature 2010, 463, 676–680.
  58. Verona, F.; Pantina, V.D.; Modica, C.; Lo Iacono, M.; D’Accardo, C.; Porcelli, G.; Cricchio, D.; Turdo, A.; Gaggianesi, M.; Di Franco, S.; et al. Targeting epigenetic alterations in cancer stem cells. Front. Mol. Med. 2022, 2, 882.
  59. Wajapeyee, N.; Gupta, R. Epigenetic Alterations and Mechanisms That Drive Resistance to Targeted Cancer Therapies. Cancer Res. 2021, 81, 5589–5595.
  60. Di Franco, S.; Bianca, P.; Sardina, D.S.; Turdo, A.; Gaggianesi, M.; Veschi, V.; Nicotra, A.; Mangiapane, L.R.; Lo Iacono, M.; Pillitteri, I.; et al. Adipose stem cell niche reprograms the colorectal cancer stem cell metastatic machinery. Nat. Commun. 2021, 12, 5006.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , , , , ,
View Times: 453
Revisions: 4 times (View History)
Update Date: 22 Dec 2022
1000/1000
Video Production Service