Autophagy Regulation on Cancer Stem Cell Maintenance, Metastasis, and Therapy Resistance
Abstract
:Simple Summary
Abstract
1. Cancer Stem Cells
2. Autophagy
3. Pro-Survival Autophagy Promotes Stemness Maintenance
4. Pro-Survival Hypoxia-Induced Autophagy Promotes Metastasis
5. Pro-Survival Autophagy Promotes Treatment Resistance
6. Lethal Autophagy and Cancer Stem Cells
7. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Baisiwala, S.; Budhiraja, S.; Goel, C.; Nandoliya, K.R.; Saathoff, M.R.; Ahmed, A.U. Spelling Out CICs: A Multi-Organ Examination of the Contributions of Cancer Initiating Cells’ Role in Tumor Progression. Stem Cell Rev. Rep. 2021, 1–13. [Google Scholar] [CrossRef]
- Pagotto, A.; Pilotto, G.; Mazzoldi, E.L.; Nicoletto, M.O.; Frezzini, S.; Pastò, A.; Amadori, A. Autophagy inhibition reduces chemoresistance and tumorigenic potential of human ovarian cancer stem cells. Cell Death Dis. 2017, 8, e2943. [Google Scholar] [CrossRef] [Green Version]
- Fabris, L.; Sato, K.; Alpini, G.; Strazzabosco, M. The Tumor Microenvironment in Cholangiocarcinoma Progression. Hepatology 2021, 73 (Suppl. S1), 75–85. [Google Scholar] [CrossRef]
- Brunel, A.; Hombourger, S.; Barthout, E.; Battu, S.; Kögel, D.; Antonietti, P.; Deluche, E.; Saada, S.; Durand, S.; Lalloué, F.; et al. Autophagy inhibition reinforces stemness together with exit from dormancy of polydisperse glioblastoma stem cells. Aging 2021, 13, 18106–18130. [Google Scholar] [CrossRef]
- Zhu, H.; Wang, D.; Zhang, L.; Xie, X.; Wu, Y.; Liu, Y.; Shao, G.; Su, Z. Upregulation of autophagy by hypoxia-inducible factor-1α promotes EMT and metastatic ability of CD133+ pancreatic cancer stem-like cells during intermittent hypoxia. Oncol. Rep. 2014, 32, 935–942. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Huang, S.; Chen, S.; Chen, J.; Wang, Z.; Wang, Y.; Zheng, H. SOX2 promotes chemoresistance, cancer stem cells properties, and epithelial-mesenchymal transition by β-catenin and Beclin1/autophagy signaling in colorectal cancer. Cell Death Dis. 2021, 12, 449. [Google Scholar] [CrossRef]
- Zhang, M.; Yang, H.; Wan, L.; Wang, Z.; Wang, H.; Ge, C.; Liu, Y.; Hao, Y.; Zhang, D.; Shi, G.; et al. Single-cell transcriptomic architecture and intercellular crosstalk of human intrahepatic cholangiocarcinoma. J. Hepatol. 2020, 73, 1118–1130. [Google Scholar] [CrossRef]
- Nazio, F.; Bordi, M.; Cianfanelli, V.; Locatelli, F.; Cecconi, F. Autophagy and cancer stem cells: Molecular mechanisms and therapeutic applications. Cell Death Differ. 2019, 26, 690–702. [Google Scholar] [CrossRef] [Green Version]
- Afify, S.M.; Sanchez Calle, A.; Hassan, G.; Kumon, K.; Nawara, H.M.; Zahra, M.H.; Mansour, H.M.; Khayrani, A.C.; Alam, M.J.; Du, J.; et al. A novel model of liver cancer stem cells developed from induced pluripotent stem cells. Br. J. Cancer 2020, 122, 1378–1390. [Google Scholar] [CrossRef] [Green Version]
- Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef]
- Codony-Servat, J.; Rosell, R. Cancer stem cells and immunoresistance: Clinical implications and solutions. Transl. Lung Cancer Res. 2015, 4, 689–703. [Google Scholar]
- Lei, M.M.L.; Lee, T.K.W. Cancer Stem Cells: Emerging Key Players in Immune Evasion of Cancers. Front. Cell Dev. Biol. 2021, 9, 692940. [Google Scholar] [CrossRef]
- Bayik, D.; Lathia, J.D. Cancer stem cell-immune cell crosstalk in tumour progression. Nat. Rev. Cancer 2021, 21, 526–536. [Google Scholar] [CrossRef]
- Yuan, S.; Norgard, R.J.; Stanger, B.Z. Cellular Plasticity in Cancer. Cancer Discov. 2019, 9, 837–851. [Google Scholar] [CrossRef] [Green Version]
- Babaei, G.; Aziz, S.G.; Jaghi, N.Z.Z. EMT, cancer stem cells and autophagy; The three main axes of metastasis. Biomed. Pharmacother. 2021, 133, 110909. [Google Scholar] [CrossRef]
- Yamashita, T.; Wang, X.W. Cancer stem cells in the development of liver cancer. J. Clin. Investig. 2013, 123, 1911–1918. [Google Scholar] [CrossRef]
- Favaro, R.; Appolloni, I.; Pellegatta, S.; Sanga, A.B.; Pagella, P.; Gambini, E.; Pisati, F.; Ottolenghi, S.; Foti, M.; Finocchiaro, G.; et al. Sox2 is required to maintain cancer stem cells in a mouse model of high-grade oligodendroglioma. Cancer Res. 2014, 74, 1833–1844. [Google Scholar] [CrossRef] [Green Version]
- Lee, T.K.; Castilho, A.; Cheung, V.C.; Tang, K.H.; Ma, S.; Ng, I.O. CD24(+) liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell Stem Cell 2011, 9, 50–63. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Wang, X. Targeting the Wnt/β-catenin signaling pathway in cancer. J. Hematol. Oncol. 2020, 13, 165. [Google Scholar] [CrossRef]
- Chen, J.-F.; Luo, X.; Xiang, L.-S.; Li, H.-T.; Zha, L.; Li, N.; He, J.-M.; Xie, G.-F.; Xie, X.; Liang, H.-J. EZH2 promotes colorectal cancer stem-like cell expansion by activating p21cip1-Wnt/β-catenin signaling. Oncotarget 2016, 7, 41540–41558. [Google Scholar] [CrossRef]
- Artavanis-Tsakonas, S.; Rand, M.D.; Lake, R.J. Notch signaling: Cell fate control and signal integration in development. Science 1999, 284, 770–776. [Google Scholar] [CrossRef] [Green Version]
- Stylianou, S.; Clarke, R.B.; Brennan, K. Aberrant activation of notch signaling in human breast cancer. Cancer Res. 2006, 66, 1517–1525. [Google Scholar] [CrossRef] [Green Version]
- Harrison, H.; Farnie, G.; Howell, S.J.; Rock, R.E.; Stylianou, S.; Brennan, K.R.; Bundred, N.J.; Clarke, R.B. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res. 2010, 70, 709–718. [Google Scholar] [CrossRef] [Green Version]
- Ding, J.; Li, H.-Y.; Zhang, L.; Zhou, Y.; Wu, J. Hedgehog Signaling, a Critical Pathway Governing the Development and Progression of Hepatocellular Carcinoma. Cells 2021, 10, 123. [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]
- Li, C.; Du, Y.; Yang, Z.; He, L.; Wang, Y.; Hao, L.; Ding, M.; Yan, R.; Wang, J.; Fan, Z. GALNT1-Mediated Glycosylation and Activation of Sonic Hedgehog Signaling Maintains the Self-Renewal and Tumor-Initiating Capacity of Bladder Cancer Stem Cells. Cancer Res. 2016, 76, 1273–1283. [Google Scholar] [CrossRef] [Green Version]
- Po, A.; Silvano, M.; Miele, E.; Capalbo, C.; Eramo, A.; Salvati, V.; Todaro, M.; Besharat, Z.M.; Catanzaro, G.; Cucchi, D.; et al. Noncanonical GLI1 signaling promotes stemness features and in vivo growth in lung adenocarcinoma. Oncogene 2017, 36, 4641–4652. [Google Scholar] [CrossRef] [Green Version]
- Staudt, L.M. Oncogenic activation of NF-kappaB. Cold Spring Harb. Perspect. Biol. 2010, 2, a000109. [Google Scholar] [CrossRef]
- Poma, P. NF-κB and Disease. Int. J. Mol. Sci. 2020, 21, 9181. [Google Scholar] [CrossRef]
- Vazquez-Santillan, K.; Melendez-Zajgla, J.; Jimenez-Hernandez, L.E.; Gaytan-Cervantes, J.; Muñoz-Galindo, L.; Piña-Sanchez, P.; Martinez-Ruiz, G.; Torres, J.; Garcia-Lopez, P.; Gonzalez-Torres, C.; et al. NF-kappaΒ-inducing kinase regulates stem cell phenotype in breast cancer. Sci. Rep. 2016, 6, 37340. [Google Scholar] [CrossRef] [Green Version]
- Bolli, R.; Dawn, B.; Xuan, Y.-T. Role of the JAK-STAT pathway in protection against myocardial ischemia/reperfusion injury. Trends Cardiovasc. Med. 2003, 13, 72–79. [Google Scholar] [CrossRef]
- Xin, P.; Xu, X.; Deng, C.; Liu, S.; Wang, Y.; Zhou, X.; Ma, H.; Wei, D.; Sun, S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int. Immunopharmacol. 2020, 80, 106210. [Google Scholar] [CrossRef]
- Kanno, H.; Sato, H.; Yokoyama, T.-A.; Yoshizumi, T.; Yamada, S. The VHL tumor suppressor protein regulates tumorigenicity of U87-derived glioma stem-like cells by inhibiting the JAK/STAT signaling pathway. Int. J. Oncol. 2013, 42, 881–886. [Google Scholar] [CrossRef]
- Cutry, A.F.; Kinniburgh, A.J.; Twardzik, D.R.; Wenner, C.E. Transforming growth factor alpha (TGF alpha) induction of C-FOS and C-MYC expression in C3H 10T1/2 cells. Biochem. Biophys. Res. Commun. 1988, 152, 216–222. [Google Scholar] [CrossRef]
- Zhou, L.; Lopes, J.E.; Chong, M.M.; Ivanov, I.I.; Min, R.; Victora, G.D.; Shen, Y.; Du, J.; Rubtsov, Y.P.; Rudensky, A.Y.; et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature 2008, 453, 236–240. [Google Scholar] [CrossRef] [Green Version]
- Xia, W.; Lo, C.M.; Poon, R.Y.C.; Cheung, T.T.; Chan, A.C.Y.; Chen, L.; Yang, S.; Tsao, G.S.W.; Wang, X.Q. Smad inhibitor induces CSC differentiation for effective chemosensitization in cyclin D1- and TGF-β/Smad-regulated liver cancer stem cell-like cells. Oncotarget 2017, 8, 38811–38824. [Google Scholar] [CrossRef]
- Justice, R.W.; Zilian, O.; Woods, D.F.; Noll, M.; Bryant, P.J. The Drosophila tumor suppressor gene warts encodes a homolog of human myotonic dystrophy kinase and is required for the control of cell shape and proliferation. Genes Dev. 1995, 9, 534–546. [Google Scholar] [CrossRef] [Green Version]
- Zhu, P.; Wang, Y.; Wu, J.; Huang, G.; Liu, B.; Ye, B.; Du, Y.; Gao, G.; Tian, Y.; He, L.; et al. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat. Commun. 2016, 7, 13608. [Google Scholar] [CrossRef] [Green Version]
- Song, S.; Ajani, J.A.; Honjo, S.; Maru, D.M.; Chen, Q.; Scott, A.W.; Heallen, T.R.; Xiao, L.; Hofstetter, W.L.; Weston, B.; et al. Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res. 2014, 74, 4170–4182. [Google Scholar] [CrossRef] [Green Version]
- Ooki, A.; Del Carmen Rodriguez Pena, M.; Marchionni, L.; Dinalankara, W.; Begum, A.; Hahn, N.M.; VandenBussche, C.J.; Rasheed, Z.A.; Mao, S.; Netto, G.J.; et al. YAP1 and COX2 Coordinately Regulate Urothelial Cancer Stem-like Cells. Cancer Res. 2018, 78, 168–181. [Google Scholar] [CrossRef] [Green Version]
- Massa, A.; Varamo, C.; Vita, F.; Tavolari, S.; Peraldo-Neia, C.; Brandi, G.; Rizzo, A.; Cavalloni, G.; Aglietta, M. Evolution of the Experimental Models of Cholangiocarcinoma. Cancers 2020, 12, 2308. [Google Scholar] [CrossRef]
- Vicent, S.; Lieshout, R.; Saborowski, A.; Verstegen, M.M.A.; Raggi, C.; Recalcati, S.; Invernizzi, P.; van der Laan, L.J.W.; Alvaro, D.; Calvisi, D.F.; et al. Experimental models to unravel the molecular pathogenesis, cell of origin and stem cell properties of cholangiocarcinoma. Liver Int. 2019, 39 (Suppl. S1), 79–97. [Google Scholar] [CrossRef] [Green Version]
- Menendez, J.A.; Joven, J.; Cufí, S.; Corominas-Faja, B.; Oliveras-Ferraros, C.; Cuyàs, E.; Martín-Castillo, B.; López-Bonet, E.; Alarcón, T.; Vazquez-Martin, A. The Warburg effect version 2.0: Metabolic reprogramming of cancer stem cells. Cell Cycle 2013, 12, 1166–1179. [Google Scholar] [CrossRef] [Green Version]
- Huang, F.; Wang, B.-R.; Wang, Y.-G. Role of autophagy in tumorigenesis, metastasis, targeted therapy and drug resistance of hepatocellular carcinoma. World J. Gastroenterol. 2018, 24, 4643–4651. [Google Scholar] [CrossRef]
- Persano, L.; Rampazzo, E.; Basso, G.; Viola, G. Glioblastoma cancer stem cells: Role of the microenvironment and therapeutic targeting. Biochem. Pharmacol. 2013, 85, 612–622. [Google Scholar] [CrossRef] [PubMed]
- Vitale, I.; Manic, G.; Dandrea, V.; De Maria, R. Role of autophagy in the maintenance and function of cancer stem cells. Int. J. Dev. Biol. 2015, 59, 95–108. [Google Scholar] [CrossRef] [Green Version]
- Stavoe, A.K.H.; Holzbaur, E.L.F. Neuronal autophagy declines substantially with age and is rescued by overexpression of WIPI2. Autophagy 2020, 16, 371–372. [Google Scholar] [CrossRef]
- Ghosh, A.K.; Mau, T.; O’Brien, M.; Garg, S.; Yung, R. Impaired autophagy activity is linked to elevated ER-stress and inflammation in aging adipose tissue. Aging 2016, 8, 2525–2537. [Google Scholar] [CrossRef] [Green Version]
- Oshima, M.; Seki, T.; Kurauchi, Y.; Hisatsune, A.; Katsuki, H. Reciprocal Regulation of Chaperone-Mediated Autophagy/Microautophagy and Exosome Release. Biol. Pharm. Bull. 2019, 42, 1394–1401. [Google Scholar] [CrossRef] [Green Version]
- Tao, Z.; Li, T.; Ma, H.; Yang, Y.; Zhang, C.; Hai, L.; Liu, P.; Yuan, F.; Li, J.; Yi, L.; et al. Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death Dis. 2018, 9, 1063. [Google Scholar] [CrossRef]
- Barthet, V.J.A.; Brucoli, M.; Ladds, M.; Nössing, C.; Kiourtis, C.; Baudot, A.D.; O’Prey, J.; Zunino, B.; Müller, M.; May, S.; et al. Autophagy suppresses the formation of hepatocyte-derived cancer-initiating ductular progenitor cells in the liver. Sci. Adv. 2021, 7, eabf9141. [Google Scholar] [CrossRef]
- Zhou, Q.; Cui, F.; Lei, C.; Ma, S.; Huang, J.; Wang, X.; Qian, H.; Zhang, D.; Yang, Y. ATG7-mediated autophagy involves in miR-138-5p regulated self-renewal and invasion of lung cancer stem-like cells derived from A549 cells. Anti-Cancer Drugs 2021, 32, 376–385. [Google Scholar] [CrossRef]
- Cao, W.; Li, J.; Yang, K.; Cao, D. An overview of autophagy: Mechanism, regulation and research progress. Bull. Cancer 2021, 108, 304–322. [Google Scholar] [CrossRef]
- Mari, M.; Griffith, J.; Rieter, E.; Krishnappa, L.; Klionsky, D.J.; Reggiori, F. An Atg9-containing compartment that functions in the early steps of autophagosome biogenesis. J. Cell Biol. 2010, 190, 1005–1022. [Google Scholar] [CrossRef] [Green Version]
- Laplante, M.; Sabatini, D.M. mTOR signaling in growth control and disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef] [Green Version]
- Pattingre, S.; Tassa, A.; Qu, X.; Garuti, R.; Liang, X.H.; Mizushima, N.; Packer, M.; Schneider, M.D.; Levine, B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005, 122, 927–939. [Google Scholar] [CrossRef] [Green Version]
- Jin, L.; Chen, Y.; Cheng, D.; He, Z.; Shi, X.; Du, B.; Xi, X.; Gao, Y.; Guo, Y. YAP inhibits autophagy and promotes progression of colorectal cancer via upregulating Bcl-2 expression. Cell Death Dis. 2021, 12, 457. [Google Scholar] [CrossRef]
- Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999, 402, 672–676. [Google Scholar] [CrossRef]
- Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.-L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
- Shen, Y.; Li, D.-D.; Wang, L.-L.; Deng, R.; Zhu, X.-F. Decreased expression of autophagy-related proteins in malignant epithelial ovarian cancer. Autophagy 2008, 4, 1067–1068. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Liu, D.; Sun, Z.; Ye, T.; Li, J.; Zeng, B.; Zhao, Q.; Rosie Xing, H. Autophagy augments the self-renewal of lung cancer stem cells by the degradation of ubiquitinated p53. Cell Death Dis. 2021, 12, 98. [Google Scholar] [CrossRef]
- Sharif, T.; Martell, E.; Dai, C.; Kennedy, B.E.; Murphy, P.; Clements, D.R.; Kim, Y.; Lee, P.W.; Gujar, S.A. Autophagic homeostasis is required for the pluripotency of cancer stem cells. Autophagy 2017, 13, 264–284. [Google Scholar] [CrossRef] [Green Version]
- Kitanaka, C.; Sato, A.; Okada, M. JNK Signaling in the Control of the Tumor-Initiating Capacity Associated with Cancer Stem Cells. Genes Cancer 2013, 4, 388–396. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Hu, S.B.; Wang, L.Y.; Zhang, X.; Zhou, X.; Yang, B.; Li, J.H.; Xiong, J.; Liu, N.; Li, Y.; et al. Autophagy-dependent generation of Axin2+ cancer stem-like cells promotes hepatocarcinogenesis in liver cirrhosis. Oncogene 2017, 36, 6725–6737. [Google Scholar] [CrossRef] [Green Version]
- Lin, C.J.; Chen, T.L.; Tseng, Y.Y.; Wu, G.J.; Hsieh, M.H.; Lin, Y.W.; Chen, R.M. Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway. Toxicol. Appl. Pharmacol. 2016, 304, 59–69. [Google Scholar] [CrossRef]
- Tasdemir, E.; Maiuri, M.C.; Galluzzi, L.; Vitale, I.; Djavaheri-Mergny, M.; D’Amelio, M.; Criollo, A.; Morselli, E.; Zhu, C.; Harper, F.; et al. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 2008, 10, 676–687. [Google Scholar] [CrossRef] [Green Version]
- Youle, R.J.; Narendra, D.P. Mechanisms of mitophagy. Nat. Rev. Mol. Cell Biol. 2011, 12, 9–14. [Google Scholar] [CrossRef]
- Boya, P.; Codogno, P.; Rodriguez-Muela, N. Autophagy in stem cells: Repair, remodelling and metabolic reprogramming. Development 2018, 145, dev146506. [Google Scholar] [CrossRef] [Green Version]
- Vazquez-Martin, A.; Van den Haute, C.; Cufí, S.; Corominas-Faja, B.; Cuyàs, E.; López-Bonet, E.; Rodriguez-Gallego, E.; Fernández-Arroyo, S.; Joven, J.; Baekelandt, V.; et al. Mitophagy-driven mitochondrial rejuvenation regulates stem cell fate. Aging 2016, 8, 1330–1352. [Google Scholar] [CrossRef] [Green Version]
- Gautier, C.A.; Kitada, T.; Shen, J. Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc. Natl. Acad. Sci. USA 2008, 105, 11364–11369. [Google Scholar] [CrossRef] [Green Version]
- Liu, K.; Lee, J.; Kim, J.Y.; Wang, L.; Tian, Y.; Chan, S.T.; Cho, C.; Machida, K.; Chen, D.; Ou, J.J. Mitophagy Controls the Activities of Tumor Suppressor p53 to Regulate Hepatic Cancer Stem Cells. Mol. Cell 2017, 68, 281–292.e5. [Google Scholar] [CrossRef] [Green Version]
- Digomann, D.; Kurth, I.; Tyutyunnykova, A.; Chen, O.; Löck, S.; Gorodetska, I.; Peitzsch, C.; Skvortsova, I.-I.; Negro, G.; Aschenbrenner, B.; et al. The CD98 Heavy Chain Is a Marker and Regulator of Head and Neck Squamous Cell Carcinoma Radiosensitivity. Clin. Cancer Res. 2019, 25, 3152–3163. [Google Scholar] [CrossRef]
- Yang, G.; Lu, C.; Mei, Z.; Sun, X.; Han, J.; Qian, J.; Liang, Y.; Pan, Z.; Kong, D.; Xu, S.; et al. Association of Cancer Stem Cell Radio-Resistance Under Ultra-High Dose Rate FLASH Irradiation With Lysosome-Mediated Autophagy. Front. Cell Dev. Biol. 2021, 9, 672693. [Google Scholar] [CrossRef]
- Ma, X.; Mao, G.; Chang, R.; Wang, F.; Zhang, X.; Kong, Z. Down-regulation of autophagy-associated protein increased acquired radio-resistance bladder cancer cells sensitivity to taxol. Int. J. Radiat. Biol. 2021, 97, 507–516. [Google Scholar] [CrossRef]
- Liu, X.; Suo, H.; Zhou, S.; Hou, Z.; Bu, M.; Liu, X.; Xu, W. Afatinib induces pro-survival autophagy and increases sensitivity to apoptosis in stem-like HNSCC cells. Cell Death Dis. 2021, 12, 728. [Google Scholar] [CrossRef]
- Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef]
- Gilkes, D.M.; Semenza, G.L. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 2013, 9, 1623–1636. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.; Dong, J.; Haiech, J.; Kilhoffer, M.-C.; Zeniou, M. Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy. Stem Cells Int. 2016, 2016, 1740936. [Google Scholar] [CrossRef] [Green Version]
- Lewerenz, J.; Hewett, S.J.; Huang, Y.; Lambros, M.; Gout, P.W.; Kalivas, P.W.; Massie, A.; Smolders, I.; Methner, A.; Pergande, M.; et al. The cystine/glutamate antiporter system x(c)(-) in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid. Redox Signal. 2013, 18, 522–555. [Google Scholar] [CrossRef] [Green Version]
- Pommier, Y. Drugging topoisomerases: Lessons and challenges. ACS Chem. Biol. 2013, 8, 82–95. [Google Scholar] [CrossRef] [Green Version]
- Jordan, M.A.; Wilson, L. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 2004, 4, 253–265. [Google Scholar] [CrossRef]
- Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacinto, F.V.; Esteller, M. MGMT hypermethylation: A prognostic foe, a predictive friend. DNA Repair 2007, 6, 1155–1160. [Google Scholar] [CrossRef] [PubMed]
- Moitra, K.; Lou, H.; Dean, M. Multidrug efflux pumps and cancer stem cells: Insights into multidrug resistance and therapeutic development. Clin. Pharmacol. Ther. 2011, 89, 491–502. [Google Scholar] [CrossRef]
- Kathawala, R.J.; Gupta, P.; Ashby, C.R., Jr.; Chen, Z.-S. The modulation of ABC transporter-mediated multidrug resistance in cancer: A review of the past decade. Drug Resist. Updates 2015, 18, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Li, G.-M.; Li, L.; Li, M.-Q.; Chen, X.; Su, Q.; Deng, Z.-J.; Liu, H.-B.; Li, B.; Zhang, W.-H.; Jia, Y.-X.; et al. DAPK3 inhibits gastric cancer progression via activation of ULK1-dependent autophagy. Cell Death Differ. 2021, 28, 952–967. [Google Scholar] [CrossRef] [PubMed]
- Lei, W.; Huo, Z. Jervine inhibits non-small cell lung cancer (NSCLC) progression by suppressing Hedgehog and AKT signaling via triggering autophagy-regulated apoptosis. Biochem. Biophys. Res. Commun. 2020, 533, 397–403. [Google Scholar] [CrossRef] [PubMed]
- Sato, K.; Marzioni, M.; Meng, F.; Francis, H.; Glaser, S.; Alpini, G. Ductular Reaction in Liver Diseases: Pathological Mechanisms and Translational Significances. Hepatology 2019, 69, 420–430. [Google Scholar] [CrossRef] [Green Version]
- Xie, Q.; Liu, Y.; Li, X. The interaction mechanism between autophagy and apoptosis in colon cancer. Transl. Oncol. 2020, 13, 100871. [Google Scholar] [CrossRef]
- Heckmann, B.L.; Green, D.R. LC3-associated phagocytosis at a glance. J. Cell Sci. 2019, 132, jcs222984. [Google Scholar] [CrossRef] [Green Version]
- Matthew-Onabanjo, A.N.; Janusis, J.; Mercado-Matos, J.; Carlisle, A.E.; Kim, D.; Levine, F.; Cruz-Gordillo, P.; Richards, R.; Lee, M.J.; Shaw, L.M. Beclin 1 Promotes Endosome Recruitment of Hepatocyte Growth Factor Tyrosine Kinase Substrate to Suppress Tumor Proliferation. Cancer Res. 2020, 80, 249–262. [Google Scholar] [CrossRef] [PubMed]
Identifier | Cancer Type | Intervention | Phase | CSC Specific? |
---|---|---|---|---|
NCT03979651 | Melanoma | Chloroquine & kinase inhibitor | NA | NO |
NCT03513211 | Prostate Cancer | Hydroxychloroquine & antifungal drug | II | NO |
NCT03037437 | HCC | Hydroxychloroquine & kinase inhibitor | II | NO |
NCT04841148 | Breast Cancer | Hydroxychloroquine & kinase inhibitor | II | NO |
NCT04735068 | NSCLC | Hydroxychloroquine & kinase inhibitor | II | NO |
NCT04132505 | PAAD | Hydroxychloroquine & kinase inhibitor | I | NO |
NCT04316169 | Breast Cancer | Hydroxychloroquine & kinase inhibitor | I | NO |
NCT04214418 | GIC | Hydroxychloroquine & kinase inhibitor & ICB | I/II | NO |
NCT04524702 | PAAD | Hydroxychloroquine & Vitamin D analog | II | NO |
NCT04341207 | Cancer & COVID-19 | Hydroxychloroquine & antibiotic | II | NO |
NCT03774472 | Breast Cancer | Hydroxychloroquine & kinase inhibitor & aromatase inhibitor | I/II | NO |
NCT03825289 | Pancreatic Cancer | Hydroxychloroquine & kinase inhibitor | I | NO |
NCT04145297 | GIC | Hydroxychloroquine & kinase inhibitor | I | NO |
NCT04566133 | Biliary Cancer | Hydroxychloroquine & kinase inhibitor | II | NO |
NCT03377179 | CCA | Hydroxychloroquine & kinase inhibitor | II | NO |
NCT04593758 | CCS | Hydroxychloroquine & anti-mitochondrial drug | I/II | NO |
NCT04911816 | PAAD | Hydroxychloroquine & FOLFIRINOX | I/II | NO |
NCT03598595 | Osteosarcoma | Hydroxychloroquine & taxane & nucleoside | I/II | NO |
NCT04201457 | Glioma | Hydroxychloroquine & enzyme/kinase inhibitors | I/II | NO |
NCT03979651 | Melanoma | Hydroxychloroquine & kinase inhibitor | NA | NO |
NCT03008148 | Glioblastoma | Hydroxychloroquine & alkylating agent & radiotherapy | II/III | NO |
NCT04375813 | Bladder Cancer | Rapamycin | II | NO |
NCT03439462 | CRC | Rapamycin | I/II | NO |
NCT02389309 | Brain Cancer | Rapamycin & alkylating agent & kinase inhibitor | I | NO |
NCT03662412 | Pancreatic Cancer | Rapamycin | I/II | NO |
NCT03433183 | MPNST, NF | Rapamycin & kinase inhibitor | II | NO |
NCT03571438 | Kidney Cancer | Rapamycin & kinase inhibitor | NA | NO |
NCT00700258 | RCC, MCL, GIC | Rapamycin & kinase inhibitor | NA | NO |
NCT02642094 | Breast cancer | Rapamycin | II | YES |
Author | Mechanism of Action | Cell Line | Animal | Results |
---|---|---|---|---|
Sharif et al., (2017) [62] | NAMPT inhibition | Teratocarcinoma CSCs | NO | ↓ POU5F1, Nanog, & SOX2 expression |
Sharif et al., (2017) [62] | ATG12 KD & ATG7 KD | HNSCC CSCs | NO | ↓ Stemness ↑ Differentiation |
Pagotto et al., (2017) [2] | ATG5 KO, CQ | Ovarian CSCs | NO | ↓ Spheroid formation ↓ Stemness markers |
Li et al., (2017) [64] | ATG3 KD, ATG7 KD & CQ | Axin2+CD90+ CSCs | NO | ↓ HGF expression ↓ Stemness markers |
Wang et al., (2021) [61] | 3-MA, BafA1 & Rapamycin | Lung CSC A549 | NO | ↑ CSC stemness ↑ Spheroid/tumor formation |
Vazquez-Martinet al., (2016) [69] | PINK1 KD | iPSC | NO | ↓ Mitochondrial rejuvenation ↑ Differentiation |
Liu et al., (2017) [71] | ATG5 KD, ATG7 KD, 3-MA | Hepatic CSCs | NO | ↑ Phosphorylated p53 ↓ Nanog expression |
Zhu et al., (2014) [5] | HIF-1A siRNA, 3-MA | Pancreatic CSCs | NO | ↓ Vimentin & MMP-9 ↑ Epithelial phenotype ↑ E-cadherin expression |
Digomann et al., (2019) [72] | ATG5 KD & BafA1 | HNSCC CSCs | NO | ↑ Radiosensitivity ↑ Apoptosis |
Yang et al., (2021) [73] | Irradiation | Breast CSCs | NO | ↑ Autophagic vesicles ↓ Initial apoptosis induction |
Zhu et al., (2021) [6] | Irinotecan (chemotherapy) w/ CQ | NO | Mouse | ↓ Tumor size |
Ma et al., (2021) [74] | Taxol w/ Beclin1/ATG5 KD | Radio-resistant Bladder CSCs | NO | ↑ Apoptosis |
Brunel et al., (2021) [4] | Temozolomide (chemotherapy) w/ Beclin1 KD | GSCs | NO | ↓ Proliferation No change in apoptosis |
Zhu et al., (2021) [6] | Rapamycin | CSCs | NO | ↑ Chemoresistance & stemness Sox2 upregulates ABC transporters |
Zhu et al., (2021) [6] | 2-MA | CSCs | NO | ↓ Malignant cancer phenotype |
Liu et al., (2021) [75] | Afatinib (RTK inhibitor) w/ 3-MA or CQ | HNSCC CSCs | NO | ↑ Afatinib-induced apoptosis with coincubation |
Zhuang et al., (2012) [76] | Curcumin | GSCs | NO | ↓ Self-renewal ↑ Induction of differentiation |
Tao et al., (2018) [50] | AZD8055 or rapamycin | GSCs | NO | ↓ Self-renewal ↓Tumorigenicity |
Tao et al., (2018) [50] | mTOR inhibition | NO | Mouse | ↓ Tumor size and prolonged survival |
Tao et al., (2018) [50] | Beclin KD, ATG5 KD & CQ | GSCs | NO | ↑ Stemness markers ↑ Proliferation & clonogenicity |
Barthet et al., (2021) [51] | ATG5KD /ATG7 KD | LPCs | Mouse | ↑ TAZ & YAP co-expression ↑ Ductular cell formation ↑ Carcinogenesis |
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
Wang, X.; Lee, J.; Xie, C. Autophagy Regulation on Cancer Stem Cell Maintenance, Metastasis, and Therapy Resistance. Cancers 2022, 14, 381. https://doi.org/10.3390/cancers14020381
Wang X, Lee J, Xie C. Autophagy Regulation on Cancer Stem Cell Maintenance, Metastasis, and Therapy Resistance. Cancers. 2022; 14(2):381. https://doi.org/10.3390/cancers14020381
Chicago/Turabian StyleWang, Xin, Jihye Lee, and Changqing Xie. 2022. "Autophagy Regulation on Cancer Stem Cell Maintenance, Metastasis, and Therapy Resistance" Cancers 14, no. 2: 381. https://doi.org/10.3390/cancers14020381
APA StyleWang, X., Lee, J., & Xie, C. (2022). Autophagy Regulation on Cancer Stem Cell Maintenance, Metastasis, and Therapy Resistance. Cancers, 14(2), 381. https://doi.org/10.3390/cancers14020381