1. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176:11-42. https://doi.org/10.1016/j.cell.2018.09.048.
2. Mizushima N, Levine B. Autophagy in human diseases. N Engl J Med. 2020;383:1564-76. https://doi.org/10.1056/NEJMra2022774.
3. Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P et al. Autophagy in major human diseases. EMBO J. 2021:e108863. https://doi.org/10.15252/embj.2021108863.
4. Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19:349-64. https://doi.org/10.1038/s41580-018-0003-4.
5. Galluzzi L, Bravo-San Pedro JM, Levine B, Green DR, Kroemer G. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat Rev Drug Discov. 2017;16:487-511. https://doi.org/10.1038/nrd.2017.22.
6. Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17:528-42. https://doi.org/10.1038/nrc.2017.53.
7. Amaravadi RK, Kimmelman AC, Debnath J. Targeting autophagy in cancer: recent advances and future directions. Cancer Discov. 2019;9:1167-81. https://doi.org/10.1158/2159-8290.CD-19-0292.
8. Wang RC, Levine B. Autophagy in cellular growth control. FEBS Lett. 2010;584:1417-26. https://doi.org/10.1016/j.febslet.2010.01.009.
9. Neufeld TP. Autophagy and cell growth--the yin and yang of nutrient responses. J Cell Sci. 2012;125:2359-68. https://doi.org/10.1242/jcs.103333.
10. Mathiassen SG, De Zio D, Cecconi F. Autophagy and the cell cycle: a complex landscape. Front Oncol. 2017;7:51. https://doi.org/10.3389/fonc.2017.00051.
11. Zheng K, He Z, Kitazato K, Wang Y. Selective autophagy regulates cell cycle in cancer therapy. Theranostics. 2019;9:104-25. https://doi.org/10.7150/thno.30308.
12. Ben-Sahra I, Manning BD. mTORC1 signaling and the metabolic control of cell growth. Curr Opin Cell Biol. 2017;45:72-82. https://doi.org/10.1016/j.ceb.2017.02.012.
13. Szwed A, Kim E, Jacinto E. Regulation and metabolic functions of mTORC1 and mTORC2. Physiol Rev. 2021;101:1371-426. https://doi.org/10.1152/physrev.00026.2020.
14. Li Z, Zhang X. Kinases involved in both autophagy and mitosis. Int J Mol Sci. 2017;18:1884. https://doi.org/10.3390/ijms18091884.
15. Nowosad A, Jeannot P, Callot C, Creff J, Perchey RT, Joffre C et al. p27 controls Ragulator and mTOR activity in amino acid-deprived cells to regulate the autophagy-lysosomal pathway and coordinate cell cycle and cell growth. Nat Cell Biol. 2020;22:1076-90. https://doi.org/10.1038/s41556-020-0554-4.
16. Vijayaraghavan S, Karakas C, Doostan I, Chen X, Bui T, Yi M et al. CDK4/6 and autophagy inhibitors synergistically induce senescence in Rb positive cytoplasmic cyclin E negative cancers. Nat Commun. 2017;8:15916. https://doi.org/10.1038/ncomms15916.
17. Yin Q, Jian Y, Xu M, Huang X, Wang N, Liu Z et al. CDK4/6 regulate lysosome biogenesis through TFEB/TFE3. J Cell Biol. 2020;219:e201911036. https://doi.org/10.1083/jcb.201911036.
18. Vessoni AT, Filippi-Chiela EC, Menck CF, Lenz G. Autophagy and genomic integrity. Cell Death Differ. 2013;20:1444-54. https://doi.org/10.1038/cdd.2013.103.
19. Roos WP, Thomas AD, Kaina B. DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer. 2016;16:20-33. https://doi.org/10.1038/nrc.2015.2.
20. Hewitt G, Korolchuk VI. Repair, reuse, recycle: the expanding role of autophagy in genome maintenance. Trends Cell Biol. 2017;27:340-51. https://doi.org/10.1016/j.tcb.2016.11.011.
21. Gomes LR, Menck CFM, Leandro GS. Autophagy roles in the modulation of DNA repair pathways. Int J Mol Sci. 2017;18:2351. https://doi.org/10.3390/ijms18112351.
22. Kenzelmann Broz D, Spano Mello S, Bieging KT, Jiang D, Dusek RL, Brady CA et al. Global genomic profiling reveals an extensive p53-regulated autophagy program contributing to key p53 responses. Genes Dev. 2013;27:1016-31. https://doi.org/10.1101/gad.212282.112.
23. Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Biol. 2015;16:393-405. https://doi.org/10.1038/nrm4007.
24. Eskelinen EL, Prescott AR, Cooper J, Brachmann SM, Wang L, Tang X et al. Inhibition of autophagy in mitotic animal cells. Traffic. 2002;3:878-93. https://doi.org/10.1034/j.1600-0854.2002.31204.x.
25. Furuya T, Kim M, Lipinski M, Li J, Kim D, Lu T et al. Negative regulation of Vps34 by Cdk mediated phosphorylation. Mol Cell. 2010;38:500-11. https://doi.org/10.1016/j.molcel.2010.05.009.
26. Odle RI, Walker SA, Oxley D, Kidger AM, Balmanno K, Gilley R et al. An mTORC1-to-CDK1 switch maintains autophagy suppression during mitosis. Mol Cell. 2020;77:228-40. https://doi.org/10.1016/j.molcel.2019.10.016.
27. Tasdemir E, Maiuri MC, Tajeddine N, Vitale I, Criollo A, Vicencio JM et al. Cell cycle-dependent induction of autophagy, mitophagy and reticulophagy. Cell Cycle. 2007;6:2263-7. https://doi.org/10.4161/cc.6.18.4681.
28. Kang KB, Zhu C, Yong SK, Gao Q, Wong MC. Enhanced sensitivity of celecoxib in human glioblastoma cells: induction of DNA damage leading to p53-dependent G1 cell cycle arrest and autophagy. Mol Cancer. 2009;8:66. https://doi.org/10.1186/1476-4598-8-66.
29. Wang Y, Xu W, Yan Z, Zhao W, Mi J, Li J et al. Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. J Exp Clin Cancer Res. 2018;37:63. https://doi.org/10.1186/s13046-018-0731-5.
30. Yu H, Yin S, Zhou S, Shao Y, Sun J, Pang X et al. Magnolin promotes autophagy and cell cycle arrest via blocking LIF/Stat3/Mcl-1 axis in human colorectal cancers. Cell Death Dis. 2018;9:702. https://doi.org/10.1038/s41419-018-0660-4.
31. Kaluzki I, Hailemariam-Jahn T, Doll M, Kaufmann R, Balermpas P, Zöller N et al. Dimethylfumarate inhibits colorectal carcinoma cell proliferation: evidence for cell cycle arrest, apoptosis and autophagy. Cells. 2019;8:1329. https://doi.org/10.3390/cells8111329.
32. Liu L, Xie R, Nguyen S, Ye M, McKeehan WL. Robust autophagy/mitophagy persists during mitosis. Cell Cycle. 2009;8:1616-20. https://doi.org/10.4161/cc.8.10.8577.
33. Domenech E, Maestre C, Esteban-Martinez L, Partida D, Pascual R, Fernandez-Miranda G et al. AMPK and PFKFB3 mediate glycolysis and survival in response to mitophagy during mitotic arrest. Nat Cell Biol. 2015;17:1304-16. https://doi.org/10.1038/ncb3231.
34. Li Z, Ji X, Wang D, Liu J, Zhang X. Autophagic flux is highly active in early mitosis and differentially regulated throughout the cell cycle. Oncotarget. 2016;7:39705-18. https://doi.org/10.18632/oncotarget.9451.
35. Li Z, Tian X, Ji X, Wang J, Chen H, Wang D et al. ULK1-ATG13 and their mitotic phospho-regulation by CDK1 connect autophagy to cell cycle. PLoS Biol. 2020;18:e3000288. https://doi.org/10.1371/journal.pbio.3000288.
36. Kuo PL, Hsu YL, Cho CY. Plumbagin induces G2-M arrest and autophagy by inhibiting the AKT/mammalian target of rapamycin pathway in breast cancer cells. Mol Cancer Ther. 2006;5:3209-21. https://doi.org/10.1158/1535-7163.MCT-06-0478.
37. Filippi-Chiela EC, Villodre ES, Zamin LL, Lenz G. Autophagy interplay with apoptosis and cell cycle regulation in the growth inhibiting effect of resveratrol in glioma cells. PLoS One. 2011;6:e20849. https://doi.org/10.1371/journal.pone.0020849.
38. Chen K, Shou LM, Lin F, Duan WM, Wu MY, Xie X et al. Artesunate induces G2/M cell cycle arrest through autophagy induction in breast cancer cells. Anticancer Drugs. 2014;25:652-62. https://doi.org/10.1097/CAD.0000000000000089.
39. Pathania AS, Guru SK, Kumar S, Kumar A, Ahmad M, Bhushan S et al. Interplay between cell cycle and autophagy induced by boswellic acid analog. Sci Rep. 2016;6:33146. https://doi.org/10.1038/srep33146.
40. Liang N, Liu X, Zhang S, Sun H. The role of Beclin 1 in IR-induced crosstalk between autophagy and G2/M cell cycle arrest. Cell Signal. 2019;62:109353. https://doi.org/10.1016/j.cellsig.2019.109353.
41. Huang R, Gao S, Han Y, Ning H, Zhou Y, Guan H et al. BECN1 promotes radiation-induced G2/M arrest through regulation CDK1 activity: a potential role for autophagy in G2/M checkpoint. Cell Death Discov. 2020;6:70. https://doi.org/10.1038/s41420-020-00301-2.
42. Sarraf SA, Sideris DP, Giagtzoglou N, Ni L, Kankel MW, Sen A et al. PINK1/Parkin influences cell cycle by sequestering TBK1 at damaged mitochondria, inhibiting mitosis. Cell Rep. 2019;29:225-35 e5. https://doi.org/10.1016/j.celrep.2019.08.085.
43. Kaminskyy V, Abdi A, Zhivotovsky B. A quantitative assay for the monitoring of autophagosome accumulation in different phases of the cell cycle. Autophagy. 2011;7:83-90. https://doi.org/10.4161/auto.7.1.13893.
44. Chan LL, Shen D, Wilkinson AR, Patton W, Lai N, Chan E et al. A novel image-based cytometry method for autophagy detection in living cells. Autophagy. 2012;8:1371-82. https://doi.org/10.4161/auto.21028.
45. Oeste CL, Seco E, Patton WF, Boya P, Perez-Sala D. Interactions between autophagic and endo-lysosomal markers in endothelial cells. Histochem Cell Biol. 2013;139:659-70. https://doi.org/10.1007/s00418-012-1057-6.
46. Guo S, Liang Y, Murphy SF, Huang A, Shen H, Kelly DF et al. A rapid and high content assay that measures cyto-ID-stained autophagic compartments and estimates autophagy flux with potential clinical applications. Autophagy. 2015;11:560-72. https://doi.org/10.1080/15548627.2015.1017181.
47. Zhao H, Traganos F, Dobrucki J, Wlodkowic D, Darzynkiewicz Z. Induction of DNA damage response by the supravital probes of nucleic acids. Cytometry A. 2009;75:510-9. https://doi.org/10.1002/cyto.a.20727.
48. Montecucco A, Zanetta F, Biamonti G. Molecular mechanisms of etoposide. EXCLI J. 2015;14:95-108. https://doi.org/10.17179/excli2015-561.
49. Clifford B, Beljin M, Stark GR, Taylor WR. G2 arrest in response to topoisomerase II inhibitors: the role of p53. Cancer Res. 2003;63:4074-81.
50. Schonn I, Hennesen J, Dartsch DC. Cellular responses to etoposide: cell death despite cell cycle arrest and repair of DNA damage. Apoptosis. 2010;15:162-72. https://doi.org/10.1007/s10495-009-0440-9.
51. Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB et al. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 2004;6:1221-8. https://doi.org/10.1038/ncb1192.
52. Nishida Y, Arakawa S, Fujitani K, Yamaguchi H, Mizuta T, Kanaseki T et al. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature. 2009;461:654-8. https://doi.org/10.1038/nature08455.
53. Kaizuka T, Morishita H, Hama Y, Tsukamoto S, Matsui T, Toyota Y et al. An autophagic flux probe that releases an internal control. Mol Cell. 2016;64:835-49. https://doi.org/10.1016/j.molcel.2016.09.037.
54. Huang HL, Hsing HW, Lai TC, Chen YW, Lee TR, Chan HT et al. Trypsin-induced proteome alteration during cell subculture in mammalian cells. J Biomed Sci. 2010;17:36. https://doi.org/10.1186/1423-0127-17-36.
55. Cheng J, Haas M. Frequent mutations in the p53 tumor suppressor gene in human leukemia T-cell lines. Mol Cell Biol. 1990;10:5502-9. https://doi.org/10.1128/mcb.10.10.5502.
56. Kojima K, Konopleva M, Samudio IJ, Shikami M, Cabreira-Hansen M, McQueen T et al. MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood. 2005;106:3150-9. https://doi.org/10.1182/blood-2005-02-0553.
57. Klionsky DJ, Abdel-Aziz AK, Abdelfatah S, Abdellatif M, Abdoli A, Abel S et al. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition). Autophagy. 2021:1-382. https://doi.org/10.1080/15548627.2020.1797280.
58. Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: from discovery to therapy. Cancer Discov. 2016;6:353-67. https://doi.org/10.1158/2159-8290.CD-15-0894.
59. Füllgrabe J, Klionsky DJ, Joseph B. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol. 2014;15:65-74. https://doi.org/10.1038/nrm3716.
60. Füllgrabe J, Ghislat G, Cho DH, Rubinsztein DC. Transcriptional regulation of mammalian autophagy at a glance. J Cell Sci. 2016;129:3059-66. https://doi.org/10.1242/jcs.188920.
61. Bordi M, De Cegli R, Testa B, Nixon RA, Ballabio A, Cecconi F. A gene toolbox for monitoring autophagy transcription. Cell Death Dis. 2021;12:1044. https://doi.org/10.1038/s41419-021-04121-9.
62. Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabolic control of autophagy. Cell. 2014;159:1263-76. https://doi.org/10.1016/j.cell.2014.11.006.
63. Kimmelman AC, White E. Autophagy and tumor metabolism. Cell Metab. 2017;25:1037-43. https://doi.org/10.1016/j.cmet.2017.04.004.
64. Lahiri V, Hawkins WD, Klionsky DJ. Watch what you (self-) eat: autophagic mechanisms that modulate metabolism. Cell Metab. 2019;29:803-26. https://doi.org/10.1016/j.cmet.2019.03.003.
65. Ferrick DA, Neilson A, Beeson C. Advances in measuring cellular bioenergetics using extracellular flux. Drug Discov Today. 2008;13:268-74. https://doi.org/10.1016/j.drudis.2007.12.008.
66. Williams GH, Stoeber K. Cell cycle markers in clinical oncology. Curr Opin Cell Biol. 2007;19:672-9. https://doi.org/10.1016/j.ceb.2007.10.005.
67. Warnes G. Flow cytometric assays for the study of autophagy. Methods. 2015;82:21-8. https://doi.org/10.1016/j.ymeth.2015.03.027.
68. Tasdemir E, Maiuri MC, Orhon I, Kepp O, Morselli E, Criollo A et al. p53 represses autophagy in a cell cycle-dependent fashion. Cell Cycle. 2008;7:3006-11. https://doi.org/10.4161/cc.7.19.6702.
69. Gump JM, Staskiewicz L, Morgan MJ, Bamberg A, Riches DW, Thorburn A. Autophagy variation within a cell population determines cell fate through selective degradation of Fap-1. Nat Cell Biol. 2014;16:47-54. https://doi.org/10.1038/ncb2886.
70. Gump JM, Thorburn A. Sorting cells for basal and induced autophagic flux by quantitative ratiometric flow cytometry. Autophagy. 2014;10:1327-34. https://doi.org/10.4161/auto.29394.
71. Ben-David U, Siranosian B, Ha G, Tang H, Oren Y, Hinohara K et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature. 2018;560:325-30. https://doi.org/10.1038/s41586-018-0409-3.
72. Thomas HE, Zhang Y, Stefely JA, Veiga SR, Thomas G, Kozma SC et al. Mitochondrial complex I activity is required for maximal autophagy. Cell Rep. 2018;24:2404-17. https://doi.org/10.1016/j.celrep.2018.07.101.