Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting
Abstract
:1. Introduction
2. The Autophagy Process and Its Regulation
Molecular Mechanism for Autophagy
3. Autophagy in Hematopoiesis
3.1. Hematopoietic Stem Cells (HSCs)
3.2. Erythrocytes
3.3. Megakaryocytes and Platelets
3.4. Granulocytes
3.5. Monocytes
4. Autophagy in AML
4.1. Autophagy and AML Stem Cells
4.2. Regulation of Autophagy Genes in AML Cells
4.3. Autophagic Biomarkers
4.4. Autophagy and Genetic Alterations in AML
4.4.1. Fusion Genes in AML and Autophagy
4.4.2. Genetic Mutations in AML and Autophagy
- FLT3
- KIT
- NPM1
- P53
- IDH1/2 (isocitrate dehydrogenase)
- DNMT3A
5. Autophagy and Tumor Microenvironment (TME) in AML
6. Autophagy Affects AML Metabolism
7. Autophagy and Chemotherapy in AML
Name | Mechanism | References |
---|---|---|
Cytotoxic | ||
Dihydroartemisinin | mTOR/p7056k-inhibition/AMPK-activation | [207] |
AC-73 | CD147 inhibitor | [190] |
Syrosingopine | MCT4 inhibitor | [189] |
Statins | Cholesterol biosynthesis | [208] |
S100A8-S100A9 | BECN1 | [209] |
Arginase | Arginine-depleting enzyme | [210] |
dendrogenin A (DDA) | Metabolite of cholesterol | [215] |
Arsenic Trioxide (ATO) | Fusion protein degradation | [126] |
All-trans retinoic acid (ATRA) | Fusion protein degradation | [126] |
Cytoprotective | ||
Rapamycin | mTOR inhibitor | [127] |
Sorafenib | Multi-targeted RTK | [134,136] |
Azacitidine (AZA) | DNA methyltransferase | [135,217] |
BIX-01294 | Histone methyltransferase G9a | [214] |
Cytosyne arabinoside (ARA-C) | DNA synthesis | [16] |
Daunorubicin (DNR) | DNA synthesis | [216] |
BET inhibitors | Decrease MYC, BCL2, and BCL6 | [213] |
Venetoclax | BCL2 inhibitors | [107] |
Name | Mechanism | Target Point | References |
---|---|---|---|
3-Methyladenine | PI3-K inhibitor | Autophagosome | [210] |
LY294002 | PI3-K/mTOR inhibitor | Autophagosome | [203] |
SBI0206965 | ULK1 inhibitor | Autophagosome | [216] |
Bafilomycin A1 | V-ATPase inhibitor | Autophagolysosome | [201,202] |
VPS34-IN1 | VSP34 inhibitor | Autophagolysosome | [204,205] |
Roc325 | Unknow | Lysosome | [217] |
Lys05 | Unknow | Lysosome | [136] |
Cloroquine | Unknow | Lysosome | [203] |
Hydroxychloroquine | Unknow | Lysosome | [198] |
XRK3F2 | SQSTM1/p62 | Mitophagy | [15] |
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
- Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
- Swaminathan, M.; Wang, E.S. Novel therapies for AML: A round-up for clinicians. Expert Rev. Clin. Pharmacol. 2020, 13, 1389–1400. [Google Scholar] [CrossRef] [PubMed]
- Stanchina, M.; Soong, D.; Zheng-Lin, B.; Watts, J.M.; Taylor, J. Advances in Acute Myeloid Leukemia: Recently Approved Therapies and Drugs in Development. Cancers 2020, 12, 3225. [Google Scholar] [CrossRef]
- Visconte, V.; Przychodzen, B.; Han, Y.; Nawrocki, S.T.; Thota, S.; Kelly, K.R.; Patel, B.J.; Hirsch, C.; Advani, A.S.; Carraway, H.E.; et al. Complete mutational spectrum of the autophagy interactome: A novel class of tumor suppressor genes in myeloid neoplasms. Leukemia 2017, 31, 505–510. [Google Scholar] [CrossRef]
- Shahrabi, S.; Paridar, M.; Zeinvand-Lorestani, M.; Jalili, A.; Zibara, K.; Abdollahi, M.; Khosravi, A. Autophagy regulation and its role in normal and malignant hematopoiesis. J. Cell Physiol. 2019, 234, 21746–21757. [Google Scholar] [CrossRef]
- Stergiou, I.E.; Kapsogeorgou, E.K. Autophagy and Metabolism in Normal and Malignant Hematopoiesis. Int. J. Mol. Sci. 2021, 22, 8540. [Google Scholar] [CrossRef]
- Du, W.; Xu, A.; Huang, Y.; Cao, J.; Zhu, H.; Yang, B.; Shao, X.; He, Q.; Ying, M. The role of autophagy in targeted therapy for acute myeloid leukemia. Autophagy 2021, 17, 2665–2679. [Google Scholar] [CrossRef]
- Deretic, V.; Kroemer, G. Autophagy in metabolism and quality control: Opposing, complementary or interlinked functions? Autophagy 2022, 18, 283–292. [Google Scholar] [CrossRef]
- Jung, S.; Jeong, H.; Yu, S.W. Autophagy as a decisive process for cell death. Exp. Mol. Med. 2020, 52, 921–930. [Google Scholar] [CrossRef]
- Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
- Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and autophagy-related pathways in cancer. Nat. Rev. Mol. Cell Biol. 2023; Epub ahead of print. [Google Scholar] [CrossRef]
- Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
- Lamark, T.; Johansen, T. Mechanisms of Selective Autophagy. Annu. Rev. Cell Dev. Biol. 2021, 37, 143–169. [Google Scholar] [CrossRef]
- Li, Y.; Yin, J.; Wang, C.; Yang, M.; Gu, J.; He, M.; Xu, H.; Fu, W.; Zhang, W.; Ru, Y.; et al. A mitophagy inhibitor targeting p62 attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Lett. 2021, 510, 24–36. [Google Scholar] [CrossRef]
- Shibutani, S.T.; Yoshimori, T. A current perspective of autophagosome biogenesis. Cell Res. 2014, 24, 58–68. [Google Scholar] [CrossRef]
- Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef]
- Zhao, Y.; Wang, Z.; Zhang, W.; Zhang, L. MicroRNAs play an essential role in autophagy regulation in various disease phenotypes. BioFactors 2019, 45, 844–856. [Google Scholar] [CrossRef]
- Kumar, A.; Girisa, S.; Alqahtani, M.S.; Abbas, M.; Hegde, M.; Sethi, G.; Kunnumakkara, A.B. Targeting Autophagy Using Long Non-Coding RNAs (LncRNAs): New Landscapes in the Arena of Cancer Therapeutics. Cells 2023, 12, 810. [Google Scholar] [CrossRef]
- Morel, E. Endoplasmic Reticulum Membrane and Contact Site Dynamics in Autophagy Regulation and Stress Response. Front. Cell Dev. Biol. 2020, 8, 343. [Google Scholar] [CrossRef]
- Hollenstein, D.M.; Kraft, C. Autophagosomes are formed at a distinct cellular structure. Curr. Opin Cell Biol. 2020, 65, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Grasso, D.; Renna, F.J.; Vaccaro, M.I. Initial Steps in Mammalian Autophagosome Biogenesis. Front. Cell Dev. Biol. 2018, 6, 146. [Google Scholar] [CrossRef] [PubMed]
- Hardie, D.G. AMPK and autophagy get connected. EMBO J. 2011, 30, 634–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, L.; Liao, M.; Zhen, Y.; Zhu, S.; Chena, X.; Zhang, J.; Hao, Y.; Liu, B. Autophagy and beyond: Unraveling the complexity of UNC-51-like kinase 1 (ULK1) from biological functions to therapeutic implications. Acta Pharm. Sin. B 2022, 12, 3743–3782. [Google Scholar] [CrossRef]
- Ktistakis, N.T.; Tooze, S.A. Digesting the Expanding Mechanisms of Autophagy. Trends Cell Biol. 2016, 26, 624–635. [Google Scholar] [CrossRef]
- 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]
- Satoo, K.; Noda, N.N.; Kumeta, H.; Fujioka, Y.; Mizushima, N.; Ohsumi, Y.; Inagaki, F. The structure of Atg4B-LC3 complex reveals the mechanism of LC3 processing and delipidation during autophagy. EMBO J. 2009, 28, 1341–1350. [Google Scholar] [CrossRef]
- Mizushima, N. A brief history of autophagy from cell biology to physiology and disease. Nat. Cell Biol. 2018, 20, 521–527. [Google Scholar] [CrossRef]
- Levine, B.; Kroemer, G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 2019, 176, 11–42. [Google Scholar] [CrossRef] [Green Version]
- Gubas, A.; Dikic, I. A guide to the regulation of selective autophagy receptors. FEBS J. 2022, 289, 75–89. [Google Scholar] [CrossRef]
- Kirkin, V.; Rogov, V.V. A Diversity of Selective Autophagy Receptors Determines the Specificity of the Autophagy Pathway. Mol. Cell 2019, 76, 268–285. [Google Scholar] [CrossRef]
- Yorimitsu, T.; Klionsky, D. Autophagy: Molecular machinery for self-eating. Cell Death Differ. 2005, 12 (Suppl. S2), 1542–1552. [Google Scholar] [CrossRef] [Green Version]
- Folkerts, H.; Hilgendorf, S.; Vellenga, E.; Bremer, E.; Wiersma, V.R. The multifaceted role of autophagy in cancer and the microenvironment. Med. Res. Rev. 2019, 39, 517–560. [Google Scholar] [CrossRef] [Green Version]
- Patergnani, S.; Missiroli, S.; Morciano, G.; Perrone, M.; Mantovani, C.M.; Anania, G.; Fiorica, F.; Pinton, P.; Giorgi, C. Understanding the Role of Autophagy in Cancer Formation and Progression Is a Real Opportunity to Treat and Cure Human Cancers. Cancers 2021, 13, 5622. [Google Scholar] [CrossRef]
- Kohli, L.; Passegué, E. Surviving change: The metabolic journey of hematopoietic stem cells. Trends Cell Biol. 2014, 24, 479–487. [Google Scholar] [CrossRef] [Green Version]
- Ito, K.; Bonora, M.; Ito, K. Metabolism as master of hematopoietic stem cell fate. Int. J. Hematol. 2019, 109, 18–27. [Google Scholar] [CrossRef] [Green Version]
- Takubo, K.; Nagamatsu, G.; Kobayashi, C.I.; Nakamura-Ishizu, A.; Kobayashi, H.; Ikeda, E.; Goda, N.; Rahimi, Y.; Johnson, R.S.; Soga, T.; et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 2013, 12, 49–61. [Google Scholar] [CrossRef] [Green Version]
- Morganti, C.; Cabezas-Wallscheid, N.; Ito, K. Metabolic Regulation of Hematopoietic Stem Cells. Hemasphere 2022, 6, e740. [Google Scholar] [CrossRef]
- Warr, M.R.; Passegué, E. Metabolic makeover for HSCs. Cell Stem Cell 2013, 12, 1–3. [Google Scholar] [CrossRef] [Green Version]
- Qiu, J.; Ghaffari, S. Mitochondrial Deep Dive into Hematopoietic Stem Cell Dormancy: Not Much Glycolysis but Plenty of Sluggish Lysosomes. Exp. Hematol. 2022, 114, 1–8. [Google Scholar] [CrossRef]
- Bigarella, C.L.; Liang, R.; Ghaffari, S. Stem cells and the impact of ROS signalling. Development 2014, 141, 4206–4218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Filippi, M.D.; Ghaffari, S. Mitochondria in the maintenance of hematopoietic stem cells: New perspectives and opportunities. Blood 2019, 133, 1943–1952. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Liu, Y.; Liu, R.; Ikenoue, T.; Guan, K.-L.; Liu, Y.; Zheng, P. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J. Exp. Med. 2008, 205, 2397–2408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirao, A.; Hoshii, T. Mechanistic/mammalian target protein of rapamycin signaling in hematopoietic stem cells and leukemia. Cancer Sci. 2013, 104, 977–982. [Google Scholar] [CrossRef] [Green Version]
- Nakada, D.; Saunders, T.L.; Morrison, S.J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 2010, 468, 653–658. [Google Scholar] [CrossRef] [Green Version]
- Khayati, K.; Bhatt, V.; Hu, Z.S.; Fahumy, S.; Luo, X.; Guo, J.Y. Autophagy compensates for Lkb1 loss to maintain adult mice homeostasis and survival. eLife 2020, 9, 62377. [Google Scholar] [CrossRef]
- Liu, F.; Lee, J.Y.; Wei, H.; Tanabe, O.; Engel, J.D.; Morrison, S.J.; Guan, J.L. FIP200 is required for the cell-autonomous maintenance of fetal hematopoietic stem cells. Blood 2010, 116, 4806–4814. [Google Scholar] [CrossRef] [Green Version]
- Wei, H.; Liu, L.; Chen, Q. Selective removal of mitochondria via mitophagy: Distinct pathways for different mitochondrial stresses. Biochim. Biophys. Acta 2015, 1853 Pt B, 2784–2790. [Google Scholar] [CrossRef] [Green Version]
- Ito, K.; Turcotte, R.; Cui, J.; Zimmerman, S.E.; Pinho, S.; Mizoguchi, T.; Arai, F.; Runnels, J.M.; Alt, C.; Teruya-Feldstein, J.; et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science 2016, 354, 1156–1160. [Google Scholar] [CrossRef] [Green Version]
- Jin, G.; Xu, C.; Zhang, X.; Long, J.; Rezaeian, A.H.; Liu, C.; Furth, M.E.; Kridel, S.; Pasche, B.; Bian, X.W.; et al. Atad3a suppresses Pink1-dependent mitophagy to maintain homeostasis of hematopoietic progenitor cells. Nat. Immunol. 2018, 19, 29–40. [Google Scholar] [CrossRef]
- Jung, H.E.; Shim, Y.R.; Oh, J.E.; Oh, D.S.; Lee, H.K. The autophagy Protein Atg5 Plays a Crucial Role in the Maintenance and Reconstitution Ability of Hematopoietic Stem Cells. Immune Netw. 2019, 19, 12. [Google Scholar] [CrossRef]
- Ho, T.T.; Warr, M.R.; Adelman, E.R.; Lansinger, O.M.; Flach, J.; Verovskaya, E.V.; Figueroa, M.E.; Passegué, E. Autophagy maintains the metabolism and function of young and old stem cells. Nature 2017, 543, 205–210. [Google Scholar] [CrossRef] [Green Version]
- Mortensen, M.; Soilleux, E.J.; Djordjevic, G.; Tripp, R.; Lutteropp, M.; Sadighi-Akha, E.; Stranks, A.J.; Glanville, J.; Knight, S.; Jacobsen, S.W.; et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011, 208, 455–467. [Google Scholar] [CrossRef] [Green Version]
- Wei, Q.; Pinho, S.; Dong, S.; Pierce, H.; Li, H.; Nakahara, F.; Xu, J.; Xu, C.; Boulais, P.E.; Zhang, D.; et al. MAEA is an E3 ubiquitin ligase promoting autophagy and maintenance of haematopoietic stem cells. Nat. Commun. 2021, 12, 2522. [Google Scholar] [CrossRef]
- Liang, R.; Arif, T.; Kalmykova, S.; Kasianov, A.; Lin, M.; Menon, V.; Qiu, J.; Bernitz, J.M.; Moore, K.; Lin, F.; et al. Restraining Lysosomal Activity Preserves Hematopoietic Stem Cell Quiescence and Potency. Cell Stem Cell 2020, 26, 359–376. [Google Scholar] [CrossRef]
- Arif, T. Lysosomes and Their Role in Regulating the Metabolism of Hematopoietic Stem Cells. Biology 2022, 11, 1410. [Google Scholar] [CrossRef]
- Zhang, J.; Wu, K.; Xiao, X.; Liao, J.; Hu, Q.; Chen, H.; Liu, J.; An, X. Autophagy as a regulatory component of erythropoiesis. Int. J. Mol. Sci. 2015, 16, 4083–4094. [Google Scholar] [CrossRef] [Green Version]
- Kundu, M.; Lindsten, T.; Yang, C.Y.; Wu, J.; Zhao, F.; Zhang, J.; Selak, M.A.; Ney, P.A.; Thompson, C.B. Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood 2008, 112, 1493–1502. [Google Scholar] [CrossRef] [Green Version]
- Mortensen, M.; Ferguson, D.J.P.; Edelmann, M.; Kessler, B.; Morten, K.J.; Komatsu, M.; Simon, A.K. Loss of autophagy in erythroid cells leads to defective removal of mitochondria and severe anemia in vivo. Proc. Nat. Acad. Sci. USA 2010, 107, 832–837. [Google Scholar] [CrossRef] [Green Version]
- Novak, I.; Kirkin, V.; McEwan, D.G.; Zhang, J.; Wild, P.; Rozenknop, A.; Rogov, V.; Löhr, F.; Popovic, D.; Occhipinti, A.; et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010, 11, 45–51. [Google Scholar] [CrossRef] [Green Version]
- Sandoval, H.; Thiagarajan, P.; Dasgupta, S.K.; Schumacher, A.; Prchal, J.T.; Chen, M.; Wang, J. Essential role for Nix in autophagic maturation of erythroid cells. Nature 2008, 454, 232–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Honda, S.; Arakawa, S.; Nishida, Y.; Yamaguchi, H.; Ishii, E.; Shimizu, S. Ulk1-mediated Atg5-independent macroautophagy mediates elimination of mitochondria from embryonic reticulocytes. Nat. Commun. 2014, 5, 4004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Platt, F.M.; Boland, B.; Van der Spoel, A.C. The cell biology of disease: Lysosomal storage disorders: The cellular impact of lysosomal dysfunction. J. Cell Biol. 2012, 199, 723–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stolla, M.C.; Reilly, A.; Bergantinos, R.; Stewart, S.; Thom, N.; Clough, C.A.; Wellington, R.C.; Stolitenko, R.; Abkowitz, J.L.; Doulatov, S. ATG4A regulates human erythroid maturation and mitochondrial clearance. Blood Adv. 2022, 6, 3579–3589. [Google Scholar] [CrossRef]
- Kang, Y.A.; Sanalkumar, R.; O’Geen, H.; Linnemann, A.K.; Chang, C.J.; Bouhassira, E.E.; Farnham, P.J.; Keles, S.; Bresnick, E.H. Autophagy driven by a master regulator of hematopoiesis. Mol. Cell Biol. 2012, 32, 226–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- You, T.; Wang, Q.; Zhu, L. Role of autophagy in megakaryocyte differentiation and platelet formation. Int. J. Physiol. Pathophysiol. Pharmacol. 2016, 8, 28–34. [Google Scholar] [PubMed]
- Cao, Y.; Cai, J.; Zhang, S.; Yuan, N.; Li, X.; Fang, Y.; Song, L.; Shang, M.; Liu, S.; Zhao, W.; et al. Loss of autophagy leads to failure in megakaryopoiesis, megakaryocyte differentiation, and thrombopoiesis in mice. Exp. Hematol. 2015, 43, 488–494. [Google Scholar] [CrossRef]
- Noetzli, L.J.; French, S.L.; Machlus, K.R. New Insights into the Differentiation of Megakaryocytes from Hematopoietic Progenitors. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1288–1300. [Google Scholar] [CrossRef]
- Ouseph, M.M.; Huang, Y.; Banerjee, M.; Joshi, S.; MacDonald, L.; Zhong, Y.; Liu, H.; Li, X.; Xiang, B.; Zhang, G.; et al. Autophagy is induced upon platelet activation and is essential for hemostasis and thrombosis. Blood 2015, 126, 2072. [Google Scholar] [CrossRef] [Green Version]
- Feng, W.; Chang, C.; Luo, D.; Su, H.; Yu, S.; Hua, W.; Chen, Z.; Hu, H.; Liu, W. Dissection of autophagy in human platelets. Autophagy 2014, 10, 642–651. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Ma, Q.; Siraj, S.; Ney, P.A.; Liu, J.; Liao, X.; Yuan, Y.; Li, W.; Liu, L.; Chen, Q. Nix-mediated mitophagy regulates platelet activation and life span. Blood Adv. 2019, 3, 2342–2354. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Tan, P.; Wang, X.; Yi, Y.; Hu, Y.; Wang, D.; Wang, F. Transcriptomic insights into temporal expression pattern of autophagy genes during monocytic and granulocytic differentiation. Autophagy 2018, 14, 558–559. [Google Scholar] [CrossRef]
- Bhattacharya, A.; Wei, Q.; Shin, J.N.; Abdel Fattah, E.; Bonilla, D.L.; Xiang, Q.; Eissa, N.T. Autophagy is required for neutrophil-mediated inflammation. Cell Rep. 2015, 12, 1731–1739. [Google Scholar] [CrossRef] [Green Version]
- Germic, N.; Hosseini, A.; Stojkov, D.; Oberson, K.; Claus, M.; Benarafa, C.; Calzavarini, S.; Angelillo-Scherrer, A.; Arnold, I.C.; Müller, A.; et al. ATG5 promotes eosinopoiesis but inhibits eosinophil effector functions. Blood 2021, 137, 2958–2969. [Google Scholar] [CrossRef]
- Dhuriya, Y.K.; Sharma, D. Necroptosis: A regulated inflammatory mode of cell death. J. Neuroinflamm. 2018, 15, 199. [Google Scholar] [CrossRef] [Green Version]
- Ueki, S.; Tokunaga, T.; Fujieda, S.; Honda, K.; Hirokawa, M.; Spencer, L.A.; Weller, P.F. Eosinophil ETosis and DNA Traps: A New Look at Eosinophilic Inflammation. Curr. Allergy Asthma Rep. 2016, 16, 54. [Google Scholar] [CrossRef] [Green Version]
- Radonjic-Hoesli, S.; Wang, X.; de Graauw, E.; Stoeckle, C.; Styp-Rekowska, B.; Hlushchuk, R.; Simon, D.; Spaeth, P.J.; Yousefi, S.; Simon, H.U. Adhesion-induced eosinophil cytolysis requires the receptor-interacting protein kinase 3 (RIPK3)-mixed lineage kinase-like (MLKL) signaling pathway, which is counterregulated by autophagy. J. Allergy Clin. Immunol. 2017, 140, 1632–1642. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.U.; O’Sullivan, K.M. The Expanding Role of Extracellular Traps in Inflammation and Autoimmunity: The New Players in Casting Dark Webs. Int. J. Mol. Sci. 2022, 23, 3793. [Google Scholar] [CrossRef]
- Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef]
- Zhang, Y.; Morgan, M.J.; Chen, K.; Choksi, S.; Liu, Z.G. Induction of autophagy is essential for monocyte-macrophage differentiation. Blood 2012, 119, 2895–2905. [Google Scholar] [CrossRef] [Green Version]
- Xu, Y.; Jagannath, C.; Liu, X.D.; Sharafkhaneh, A.; Kolodziejska, K.E.; Eissa, N.T. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity 2007, 27, 135–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Travassos, L.H.; Carneiro, L.A.M.; Ramjeet, M.; Hussey, S.; Kim, Y.G.; Magalhães, J.G.; Yuan, L.; Soares, F.; Chea, E.; Le Bourhis, L.; et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat. Immunol. 2010, 11, 55–62. [Google Scholar] [CrossRef] [PubMed]
- Wen, J.H.; Li, D.Y.; Liang, S.; Yang, C.; Tang, J.X.; Liu, H.F. Macrophage autophagy in macrophage polarization, chronic inflammation and organ fibrosis. Front. Immunol. 2022, 13, 946832. [Google Scholar] [CrossRef] [PubMed]
- Auberger, P.; Puissant, A. Autophagy, a key mechanism of oncogenesis and resistance in leukemia. Blood 2017, 129, 547–552. [Google Scholar] [CrossRef]
- Chen, Y.; Li, J.; Xu, L.; Găman, M.A.; Zou, Z. The genesis and evolution of acute myeloid leukemia stem cells in the microenvironment: From biology to therapeutic targeting. Cell Death Discov. 2022, 8, 397. [Google Scholar] [CrossRef]
- Seo, W.; Silwal, P.; Song, I.C.; Jo, E.K. The dual role of autophagy in acute myeloid leukemia. J. Hematol. Oncol. 2022, 15, 51. [Google Scholar] [CrossRef]
- Altman, J.K.; Szilard, A.; Goussetis, D.J.; Sassano, A.; Colamonici, M.; Gounaris, E.; Frankfurt, O.; Giles, F.J.; Eklund, E.A.; Beauchamp, E.M.; et al. Autophagy is a survival mechanism of acute myelogenous leukemia precursors during dual mTORC2/mTORC1 targeting. Clin. Cancer Res. 2014, 20, 2400–2409. [Google Scholar] [CrossRef] [Green Version]
- Sumitomo, Y.; Koya, J.; Nakazaki, K.; Kataoka, K.; Tsuruta-Kishino, T.; Morita, K.; Sato, T.; Kurokawa, M. Cytoprotective autophagy maintains leukemia-initiating cells in murine myeloid leukemia. Blood 2016, 128, 1614–1624. [Google Scholar] [CrossRef] [Green Version]
- Torgersen, M.L.; Simonsen, A. Autophagy: Friend or foe in the treatment of fusion protein-associated leukemias? Autophagy 2013, 9, 2175–2177. [Google Scholar] [CrossRef] [Green Version]
- Khan, A.; Singh, V.K.; Thakral, D.; Gupta, R. Autophagy in acute myeloid leukemia: A paradoxical role in chemoresistance. Clin. Transl. Oncol. 2022, 24, 1459–1469. [Google Scholar] [CrossRef]
- Bednarczyk, M.; Kociszewska, K.; Grosicka, O.; Grosicki, S. The role of autophagy in acute myeloid leukemia development. Expert Rev. Anticancer Ther. 2023, 23, 5–18. [Google Scholar] [CrossRef]
- Maynard, R.S.; Hellmich, C.; Bowles, K.M.; Rushworth, S.A. Acute Myeloid Leukaemia Drives Metabolic Changes in the Bone Marrow Niche. Front. Oncol. 2022, 12, 924567. [Google Scholar] [CrossRef]
- Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
- Castro, I.; Sampaio-Marques, B.; Ludovico, P. Targeting Metabolic Reprogramming in Acute Myeloid Leukemia. Cells 2019, 8, 967. [Google Scholar] [CrossRef] [Green Version]
- Lin, Q.; Chen, J.; Gu, L.; Dan, X.; Zhang, C.; Yang, Y. New insights into mitophagy and stem cells. Stem Cell Res. Ther. 2021, 12, 452. [Google Scholar] [CrossRef]
- Pei, S.; Minhajuddin, M.; Adane, B.; Khan, N.; Stevens, B.M.; Mack, S.C.; Sisi, L.; Rich, J.N.; Inguva, A.; Shannon, K.M.; et al. AMPK/FIS1-Mediated Mitophagy Is Required for Self-Renewal of Human AML Stem Cells. Cell Stem Cell 2018, 23, 86–100. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.D.; Shaid, S.; Vakhrusheva, O.; Koschade, S.E.; Klann, K.; Thölken, M.; Baker, F.; Zhang, J.; Oellerich, T.; Sürün, D.; et al. Loss of the selective autophagy receptor p62 impairs murine myeloid leukemia progression and mitophagy. Blood 2019, 133, 168–179, Erratum in: Blood 2019, 134, 94.. [Google Scholar] [CrossRef] [Green Version]
- White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef] [Green Version]
- Gewirtz, D.A. The four faces of autophagy: Implications for cancer therapy. Cancer Res. 2014, 74, 647–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, H.Y.; White, E. Role of autophagy in cancer prevention. Cancer Prev. Res. 2011, 4, 973–983. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Clark, J.; Wunderlich, M.; Fan, C.; Davis, A.; Chen, S.; Guan, J.L.; Mulloy, J.C.; Kumar, A.; Zheng, Y. Autophagy is dispensable for Kmt2a/Mll-Mllt3/Af9 AML maintenance and anti-leukemic effect of chloroquine. Autophagy 2017, 13, 955–966. [Google Scholar] [CrossRef] [Green Version]
- Porter, H.A.; Leveque-El Mouttie, L.; Vu, T.; Bruedigam, C.; Sutton, J.; Jacquelin, S.; Hill, G.R.; MacDonald, K.P.A.; Lane, S.W. Acute myeloid leukemia stem cell function is preserved in the absence of autophagy. Haematologica 2017, 102, 344–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watson, A.S.; Riffelmacher, T.; Stranks, A.; Williams, O.; De Boer, J.; Cain, K.; MacFarlane, M.; McGouran, J.; Kessler, B.; Khandwala, S.; et al. Autophagy limits proliferation and glycolytic metabolism in acute myeloid leukemia. Cell Death Discov. 2015, 1, 15008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shu, F.; Xiao, H.; Li, Q.N.; Ren, X.S.; Liu, Z.G.; Hu, B.W.; Wang, H.S.; Wang, H.; Jiang, G.M. Epigenetic and post-translational modifications in autophagy: Biological functions and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 32. [Google Scholar] [CrossRef]
- Djavaheri-Mergny, M.; Giuriato, S.; Tschan, M.P.; Humbert, M. Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019, 8, 103. [Google Scholar] [CrossRef] [Green Version]
- Piya, S.; Kornblau, S.M.; Ruvolo, V.R.; Mu, H.; Ruvolo, P.P.; McQueen, T.; Davis, R.E.; Hail, N., Jr.; Kantarjian, H.; Andreeff, M.; et al. Atg7 suppression enhances chemotherapeutic agent sensitivity and overcomes stroma-mediated chemoresistance in acute myeloid leukemia. Blood 2016, 128, 1260–1269. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.; Mei, S.; Meng, W.; Xue, S.; Jiang, L.; Yang, Y.; Hui, L.; Chen, Y.; Guan, M.X. CXCR4-mediated signaling regulates autophagy and influences acute myeloid leukemia cell survival and drug resistance. Cancer Lett. 2018, 425, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.J.; Bao, L.; Keefer, K.; Shanmughapriya, S.; Chen, L.; Lee, J.; Wang, J.F.; Zhang, X.Q.; Hirschler-Laszkiewicz, I.; Merali, S.; et al. Transient receptor potential ion channel TRPM2 promotes AML proliferation and survival through modulation of mitochondrial function, ROS, and autophagy. Cell Death Dis. 2020, 11, 247. [Google Scholar] [CrossRef] [Green Version]
- Folkerts, H.; Wierenga, A.T.; van den Heuvel, F.A.; Woldhuis, R.R.; Kluit, D.S.; Jaques, J.; Schuringa, J.J.; Vellenga, E. Elevated VMP1 expression in acute myeloid leukemia amplifies autophagy and is protective against venetoclax-induced apoptosis. Cell Death Dis. 2019, 10, 421. [Google Scholar] [CrossRef] [Green Version]
- Jin, J.; Britschgi, A.; Schläfli, A.M.; Humbert, M.; Shan-Krauer, D.; Batliner, J.; Federzoni, E.A.; Ernst, M.; Torbett, B.E.; Yousefi, S.; et al. Low Autophagy (ATG) Gene Expression Is Associated with an Immature AML Blast Cell Phenotype and Can Be Restored during AML Differentiation Therapy. Oxid. Med. Cell Longev. 2018, 2018, 1482795. [Google Scholar] [CrossRef]
- Gabra, M.M.; Salmena, L. microRNAs and Acute Myeloid Leukemia Chemoresistance: A Mechanistic Overview. Front. Oncol. 2017, 7, 255. [Google Scholar] [CrossRef] [PubMed]
- Fischer, J.; Rossetti, S.; Datta, A.; Eng, K.; Beghini, A.; Sacchi, N. miR-17 deregulates a core RUNX1-miRNA mechanism of CBF acute myeloid leukemia. Mol. Cancer. 2015, 14, 7. [Google Scholar] [CrossRef] [Green Version]
- Mi, S.; Li, Z.; Chen, P.; He, C.; Cao, D.; Elkahloun, A.; Lu, J.; Pelloso, L.A.; Wunderlich, M.; Huang, H.; et al. Aberrant overexpression and function of the miR-17-92 cluster in MLL-rearranged acute leukemia. Proc. Natl. Acad. Sci. USA 2010, 107, 3710–3715. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Liu, J.; Chen, K.; Wang, J.; Dong, Q.; Xie, J.; Yuan, Y. Vitamin D promotes autophagy in AML cells by inhibiting miR-17-5p-induced Beclin-1 overexpression. Mol. Cell Biochem. 2021, 476, 3951–3962. [Google Scholar] [CrossRef]
- Ganesan, S.; Palani, H.K.; Lakshmanan, V.; Balasundaram, N.; Alex, A.A.; David, S.; Venkatraman, A.; Korula, A.; George, B.; Balasubramanian, P.; et al. Stromal cells downregulate miR-23a-5p to activate protective autophagy in acute myeloid leukemia. Cell Death Dis. 2019, 10, 736. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Kang, J.; Liu, L.; Chen, L.; Ren, S.; Tao, Y. MicroRNA-143 sensitizes acute myeloid leukemia cells to cytarabine via targeting ATG7- and ATG2B-dependent autophagy. Aging 2020, 12, 20111–20126. [Google Scholar] [CrossRef]
- Bollaert, E.; Claus, M.; Vandewalle, V.; Lenglez, S.; Essaghir, A.; Demoulin, J.B.; Havelange, V. MiR-15a-5p Confers Chemoresistance in Acute Myeloid Leukemia by Inhibiting Autophagy Induced by Daunorubicin. Int. J. Mol. Sci. 2021, 22, 5153. [Google Scholar] [CrossRef]
- Chen, X.X.; Li, Z.P.; Zhu, J.H.; Xia, H.T.; Zhou, H. Systematic Analysis of Autophagy-Related Signature Uncovers Prognostic Predictor for Acute Myeloid Leukemia. DNA Cell Biol. 2020, 39, 1595–1605. [Google Scholar] [CrossRef]
- Huang, L.; Lin, L.; Fu, X.; Meng, C. Development and validation of a novel survival model for acute myeloid leukemia based on autophagy-related genes. PeerJ 2021, 9, 11968. [Google Scholar] [CrossRef]
- Fu, D.; Zhang, B.; Wu, S.; Zhang, Y.; Xie, J.; Ning, W.; Jiang, H. Prognosis and Characterization of Immune Microenvironment in Acute Myeloid Leukemia Through Identification of an Autophagy-Related Signature. Front. Immunol. 2021, 12, 695865. [Google Scholar] [CrossRef]
- Gu, S.; Zi, J.; Han, Q.; Song, C.; Ge, Z. The Autophagy-Related Long Non-Coding RNA Signature for Acute Myeloid Leukemia. 16 April 2020. Available online: https://ssrn.com/abstract=3578746 (accessed on 16 April 2020).
- Zhao, C.; Wang, Y.; Tu, F.; Zhao, S.; Ye, X.; Liu, J.; Zhang, J.; Wang, Z. A Prognostic Autophagy-Related Long Non-coding RNA (ARlncRNA) Signature in Acute Myeloid Leukemia (AML). Front. Genet. 2021, 12, 681867. [Google Scholar] [CrossRef]
- Nazha, A.; Zarzour, A.; Al-Issa, K.; Radivoyevitch, T.; Carraway, H.E.; Hirsch, C.M.; Przychodzen, B.; Pate, B.J.; Clemente, M.; Sanikommu, S.R.; et al. The complexity of interpreting genomic data in patients with acute myeloid leukemia. Blood Cancer J. 2016, 6, 510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Torgersen, M.L.; Engedal, N.; Bøe, S.O.; Hokland, P.; Simonsen, A. Targeting autophagy potentiates the apoptotic effect of histone deacetylase inhibitors in t(8;21) AML cells. Blood 2013, 122, 2467–2476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakatani, K.; Matsuo, H.; Harata, Y.; Higashitani, M.; Koyama, A.; Noura, M.; Nishinaka-Arai, Y.; Kamikubo, Y.; Adachi, S. Inhibition of CDK4/6 and autophagy synergistically induces apoptosis in t(8;21) acute myeloid leukemia cells. Int. J. Hematol. 2021, 113, 243–253. [Google Scholar] [CrossRef] [PubMed]
- Mannan, A.; Muhsen, N.I.; Barragán, E.; Sanz, M.A.; Mohty, M.; Hashmi, S.K.; Aljurf, M. Genotypic and Phenotypic Characteristics of Acute Promyelocytic Translocation Variants. Hematol. Oncol. Stem Cell Ther. 2020, 13, 189–201. [Google Scholar] [CrossRef]
- De Thé, H.; Pandolfi, P.P.; Chen, Z. Acute Promyelocytic Leukemia: A Paradigm for Oncoprotein-Targeted Cure. Cancer Cell 2017, 32, 552–560. [Google Scholar] [CrossRef] [Green Version]
- Moosavi, M.A.; Djavaheri-Mergny, M. Autophagy: New Insights into Mechanisms of Action and Resistance of Treatment in Acute Promyelocytic leukemia. Int. J. Mol. Sci. 2019, 20, 3559. [Google Scholar] [CrossRef] [Green Version]
- Li, T.; Ma, R.; Zhang, Y.; Mo, H.; Yang, X.; Hu, S.; Wang, L.; Novakovic, V.A.; Chen, H.; Kou, J.; et al. Arsenic trioxide promoting ETosis in acute promyelocytic leukemia through mTOR-regulated autophagy. Cell Death Dis. 2018, 9, 75. [Google Scholar] [CrossRef] [Green Version]
- Meyer, C.; Kowarz, E.; Hofmann, J.; Renneville, A.; Zuna, J.; Trka, J.; Ben Abdelali, R.; Macintyre, E.; De Braekeleer, E.; De Braekeleer, M.; et al. New insights to the MLL recombinome of acute leukemias. Leukemia 2009, 23, 1490–1499. [Google Scholar] [CrossRef]
- Manara, E.; Baron, E.; Tregnago, C.; Aveic, S.; Bisio, V.; Bresolin, S.; Masetti, R.; Locatelli, F.; Basso, G.; Pigazzi, M. MLL-AF6 fusion oncogene sequesters AF6 into the nucleus to trigger RAS activation in myeloid leukemia. Blood 2014, 124, 263–272. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.; Chen, L.; Atkinson, J.M.; Claxton, D.F.; Wang, H.G. Atg5-dependent autophagy contributes to the development of acute myeloid leukemia in an MLL-AF9-driven mouse model. Cell Death Dis. 2016, 7, 2361. [Google Scholar] [CrossRef] [Green Version]
- Müller, A.M.; Duque, J.; Shizuru, J.A.; Lübbert, M. Complementing mutations in core binding factor leukemias: From mouse models to clinical applications. Oncogene 2008, 27, 5759–5773. [Google Scholar] [CrossRef] [Green Version]
- Man, N.; Tan, Y.; Sun, X.J.; Liu, F.; Cheng, G.; Greenblatt, S.M.; Martinez, C.; Karl, D.L.; Ando, K.; Sun, M.; et al. Caspase-3 controls AML1-ETO-driven leukemogenesis via autophagy modulation in a ULK1-dependent manner. Blood 2017, 129, 2782–2792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sakaguchi, M.; Yamaguchi, H.; Najima, Y.; Usuki, K.; Ueki, T.; Oh, I.; Mori, S.; Kawata, E.; Uoshima, N.; Kobayashi, Y.; et al. Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2018, 2, 2744–2754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heydt, Q.; Larrue, C.; Saland, E.; Bertoli, S.; Sarry, J.E.; Besson, A.; Manenti, S.; Joffre, C.; Mansat-De Mas, V. Oncogenic FLT3-ITD supports autophagy via ATF4 in acute myeloid leukemia. Oncogene 2018, 37, 787–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, D.; Chen, Y.; Yang, Y.; Yin, Z.; Huang, C.; Wang, Q.; Jiang, L.; Jiang, X.; Yin, C.; Liu, Q.; et al. Autophagy activation mediates resistance to FLT3 inhibitors in acute myeloid leukemia with FLT3-ITD mutation. J. Transl. Med. 2022, 20, 300. [Google Scholar] [CrossRef]
- Qiu, S.; Kumar, H.; Yan, C.; Li, H.; Paterson, A.J.; Anderson, N.R.; He, J.; Yang, J.; Xie, M.; Crossman, D.K.; et al. Autophagy inhibition impairs leukemia stem cell function in FLT3-ITD AML but has antagonistic interactions with tyrosine kinase inhibition. Leukemia 2022, 36, 2621–2633. [Google Scholar] [CrossRef]
- Rudat, S.; Pfaus, A.; Cheng, Y.Y.; Holtmann, J.; Ellegast, J.M.; Bühler, C.; Marcantonio, D.D.; Martinez, E.; Göllner, S.; Wickenhauser, C.; et al. RET-mediated autophagy suppression as targetable co-dependence in acute myeloid leukemia. Leukemia 2018, 32, 2189–2202. [Google Scholar] [CrossRef]
- Dany, M.; Gencer, S.; Nganga, R.; Thomas, R.J.; Oleinik, N.; Baron, K.D.; Szulc, Z.M.; Ruvolo, P.; Kornblau, S.; Andreeff, M.; et al. Targeting FLT3-ITD signaling mediates ceramide-dependent mitophagy and attenuates drug resistance in AML. Blood 2016, 128, 1944–1958. [Google Scholar] [CrossRef] [Green Version]
- Zalpoor, H.; Bakhtiyari, M.; Akbari, A.; Aziziyan, F.; Shapourian, H.; Liaghat, M.; Zare-Badie, Z.; Yahyazadeh, S.; Tarhriz, V.; Ganjalikhani-Hakemi, M. Potential role of autophagy induced by FLT3-ITD and acid ceramidase in acute myeloid leukemia chemo-resistance: New insights. Cell Commun. Signal. 2022, 20, 172. [Google Scholar] [CrossRef]
- Ishikawa, Y.; Kawashima, N.; Atsuta, Y.; Sugiura, I.; Sawa, M.; Dobashi, N.; Yokoyama, H.; Doki, N.; Tomita, A.; Kiguchi, T.; et al. Prospective evaluation of prognostic impact of KIT mutations on acute myeloid leukemia with RUNX1-RUNX1T1 and CBFB-MYH11. Blood Adv. 2020, 4, 66–75. [Google Scholar] [CrossRef]
- Ayatollahi, H.; Shajiei, A.; Sadeghian, M.H.; Sheikhi, M.; Yazdandoust, E.; Ghazanfarpour, M.; Shams, S.F.; Shakeri, S. Prognostic Importance of C-KIT Mutations in Core Binding Factor Acute Myeloid Leukemia: A Systematic Review. Hematol. Oncol. Stem Cell Ther. 2017, 10, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Larrue, C.; Heydt, Q.; Saland, E.; Boutzen, H.; Kaoma, T.; Sarry, J.E.; Joffre, C.; Récher, C. Oncogenic KIT mutations induce STAT3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 2019, 8, 39. [Google Scholar] [CrossRef] [Green Version]
- Xu, J.; Zheng, J.; Fu, X.; Wu, W.; Tao, L.; Li, D.; Lin, D. Inhibition of N822K T>A mutation-induced constitutive c-KIT activation in AML cells triggers apoptotic and autophagic pathways leading to death. Int. J. Med. Sci. 2019, 16, 757–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Hu, J. Nucleophosmin1 (NPM1) abnormality in hematologic malignancies, and therapeutic targeting of mutant NPM1 in acute myeloid leukemia. Ther. Adv. Hematol. 2020, 11, 2040620719899818. [Google Scholar] [CrossRef]
- Zou, Q.; Tan, S.; Yang, Z.; Zhan, Q.; Jin, H.; Xian, J.; Zhang, S.; Yang, L.; Wang, L.; Zhang, L. NPM1 Mutant Mediated PML Delocalization and Stabilization Enhances Autophagy and Cell Survival in Leukemic Cells. Theranostics 2017, 7, 2289–2304. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Yang, L.; Yang, Z.; Tang, Y.; Tao, Y.; Zhan, Q.; Lei, L.; Jing, Y.; Jiang, X.; Jin, H.; et al. Glycolytic Enzyme PKM2 Mediates Autophagic Activation to Promote Cell Survival in NPM1-Mutated Leukemia. Int. J. Biol. Sci. 2019, 15, 882–894. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Tao, Y.; Wang, L.; Yang, L.; Jing, Y.; Jiang, X.; Lei, L.; Yang, Z.; Wang, X.; Peng, M.; et al. NPM1 mutant maintains ULK1 protein stability via TRAF6-dependent ubiquitination to promote autophagic cell survival in leukemia. FASEB J. 2021, 35, 21192. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Pan, C.; Zhang, X.; Zhao, A.; Dong, Y. Mutant NPM1 maintains RASGRP3 protein stability via interaction with MID1 to promote acute myeloid leukemia cell proliferation and autophagy. J. Leukoc. Biol. 2023, 113, 504–517. [Google Scholar] [CrossRef]
- Ok, C.Y.; Patel, K.P.; Garcia-Manero, G.; Routbort, M.J.; Peng, J.; Tang, G.; Goswami, M.; Young, K.H.; Singh, R.; Medeiros, L.J.; et al. TP53 mutation characteristics in therapy-related myelodysplastic syndromes and acute myeloid leukemia is similar to de novo diseases. J. Hematol. Oncol. 2015, 8, 45. [Google Scholar] [CrossRef] [Green Version]
- Prokocimer, M.; Molchadsky, A.; Rotter, V. Dysfunctional diversity of p53 proteins in adult acute myeloid leukemia: Projections on diagnostic workup and therapy. Blood 2017, 130, 699–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Folkerts, H.; Hilgendorf, S.; Wierenga, A.T.J.; Jaques, J.; Mulder, A.B.; Coffer, P.J.; Schuringa, J.J.; Vellenga, E. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis. 2017, 8, 2927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomez-Puerto, M.C.; Folkerts, H.; Wierenga, A.T.; Schepers, K.; Schuringa, J.J.; Coffer, P.J.; Vellenga, E. Autophagy proteins ATG5 and ATG7 are essential for the maintenance of human CD34 (+) hematopoietic stem-progenitor cells. Stem Cells 2016, 34, 1651–1663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allende-Vega, N.; Villalba, M. Metabolic stress controls mutant p53 R248Q stability in acute myeloid leukemia cells. Sci. Rep. 2019, 9, 5637. [Google Scholar] [CrossRef] [Green Version]
- Maiuri, M.C.; Galluzzi, L.; Morselli, E.; Kepp, O.; Malik, S.A.; Kroemer, G. Autophagy regulation by p53. Curr. Opin. Cell Biol. 2010, 22, 181–185. [Google Scholar] [CrossRef]
- Crighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134. [Google Scholar] [CrossRef] [Green Version]
- Feng, Z.; Zhang, H.; Levine, A.J.; Jin, S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. USA 2005, 102, 8204–8209. [Google Scholar] [CrossRef] [Green Version]
- Vu, T.T.; Stölzel, F.; Wang, K.W.; Röllig, C.; Tursky, M.L.; Molloy, T.J.; Ma, D.D. miR-10a as a therapeutic target and predictive biomarker for MDM2 inhibition in acute myeloid leukemia. Leukemia 2021, 35, 1933–1948. [Google Scholar] [CrossRef]
- Burgess, A.; Chia, K.M.; Haupt, S.; Thomas, D.; Haupt, Y.; Lim, E. Clinical Overview of MDM2/X-Targeted Therapies. Front. Oncol. 2016, 6, 7. [Google Scholar] [CrossRef] [Green Version]
- Nakazawa, S.; Sakata, K.I.; Liang, S.; Yoshikawa, K.; Iizasa, H.; Tada, M.; Hamada, J.I.; Kashiwazaki, H.; Kitagawa, Y.; Yamazaki, Y. Dominant-negative p53 mutant R248Q increases the motile and invasive activities of oral squamous cell carcinoma cells. BioMed. Res. 2019, 40, 37–49. [Google Scholar] [CrossRef] [Green Version]
- Yao, Y.; Chai, X.; Gong, C.; Zou, L. WT1 inhibits AML cell proliferation in a p53-dependent manner. Cell Cycle 2021, 20, 1552–1560. [Google Scholar] [CrossRef]
- Wilson, E.R.; Helton, N.M.; Heath, S.E.; Fulton, R.S.; Payton, J.E.; Welch, J.S.; Walter, M.J.; Westervelt, P.; DiPersio, J.F.; Link, D.C.; et al. Focal disruption of DNA methylation dynamics at enhancers in IDH-mutant AML cells. Leukemia 2022, 36, 935–945. [Google Scholar] [CrossRef]
- Tulstrup, M.; Soerensen, M.; Hansen, J.W.; Gillberg, L.; Needhamsen, M.; Kaastrup, K.; Helin, K.; Christensen, K.; Weischenfeldt, J.; Grønbæk, K. TET2 mutations are associated with hypermethylation at key regulatory enhancers in normal and malignant hematopoiesis. Nat. Commun. 2021, 12, 6061. [Google Scholar] [CrossRef] [PubMed]
- Chaudry, S.F.; Chevassut, T.J.T. Epigenetic Guardian: A Review of the DNA Methyltransferase DNMT3A in Acute Myeloid Leukaemia and Clonal Haematopoiesis. BioMed. Res. Int. 2017, 2017, 5473197. [Google Scholar] [CrossRef]
- Nawrocki, S.T.; Han, Y.; Visconte, V.; Przychodzen, B.; Espitia, C.M.; Phillips, J.; Anwer, F.; Advani, A.; Carraway, H.E.; Kelly, K.R.; et al. The novel autophagy inhibitor ROC-325 augments the antileukemic activity of azacitidine. Leukemia 2019, 33, 2971–2974. [Google Scholar] [CrossRef]
- Dai, Y.-J.; Hu, F.; He, S.-Y.; Wang, Y.-Y. Epigenetic landscape analysis of lncRNAs in acute myeloid leukemia with DNMT3A mutations. Ann. Transl. Med. 2020, 8, 318. [Google Scholar] [CrossRef]
- Testa, U.; Labbaye, C.; Castelli, G.; Pelosi, E. Oxidative stress and hypoxia in normal and leukemic stem cells. Exp. Hematol. 2016, 44, 540–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kokkaliaris, K.D.; Scadden, D.T. Cell interactions in the bone marrow microenvironment affecting myeloid malignancies. Blood Adv. 2020, 4, 3795–3803. [Google Scholar] [CrossRef]
- Poillet-Perez, L.; Sarry, J.C.; Joffre, C. Autophagy is a major metabolic regulator involved in cancer therapy resistance. Cell Rep. 2021, 36, 109528. [Google Scholar] [CrossRef] [PubMed]
- Bustos, S.O.; Antunes, F.; Rangel, M.C.; Chammas, R. Emerging Autophagy Functions Shape the Tumor Microenvironment and Play a Role in Cancer Progression-Implications for Cancer Therapy. Front. Oncol. 2020, 10, 606436. [Google Scholar] [CrossRef]
- Li, Y.; Jiang, Y.; Cheng, J.; Ma, J.; Li, Q.; Pang, T. ATG5 regulates mesenchymal stem cells differentiation and mediates chemosensitivity in acute myeloid leukemia. Biochem. Biophys. Res. Commun. 2020, 525, 398–405. [Google Scholar] [CrossRef] [PubMed]
- Joffre, C.; Ducau, C.; Poillet-Perez, L.; Courdy, C.; Mansat-De Mas, V. Autophagy a Close Relative of AML Biology. Biology 2021, 10, 552. [Google Scholar] [CrossRef]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.L.; Wang, J.H.; Zhao, A.H.; Xu, X.; Wang, Y.H.; Chen, T.L.; Li, J.M.; Mi, J.Q.; Zhu, Y.M.; Liu, Y.F.; et al. A distinct glucose metabolism signature of acute myeloid leukemia with prognostic value. Blood 2014, 124, 1645–1654. [Google Scholar] [CrossRef]
- Tirado, H.A.; Balasundaram, N.; Laaouimir, L.; Erdem, A.; van Gastel, N. Metabolic crosstalk between stromal and malignant cells in the bone marrow niche. Bone Rep. 2023, 18, 101669. [Google Scholar] [CrossRef]
- Poulain, L.; Sujobert, P.; Zylbersztejn, F.; Barreau, S.; Stuani, L.; Lambert, M.; Palama, T.L.; Chesnais, V.; Birsen, R.; Vergez, F.; et al. High mTORC1 activity drives glycolysis addiction and sensitivity to G6PD inhibition in acute myeloid leukemia cells. Leukemia 2017, 31, 2326–2335. [Google Scholar] [CrossRef]
- Stuani, L.; Sabatier, M.; Saland, E.; Cognet, G.; Poupin, N.; Bosc, C.; Castelli, F.A.; Gales, L.; Turtoi, E.; Montersino, C.; et al. Mitochondrial metabolism supports resistance to IDH mutant inhibitors in acute myeloid leukemia. J. Exp. Med. 2021, 218, 20200924. [Google Scholar] [CrossRef]
- Ye, H.; Adane, B.; Khan, N.; Alexeev, E.; Nusbacher, N.; Minhajuddin, M.; Stevens, B.M.; Winters, A.C.; Lin, X.; Ashton, J.M.; et al. Subversion of Systemic Glucose Metabolism as a Mechanism to Support the Growth of Leukemia Cells. Cancer Cell 2018, 34, 659–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, C.L.; Stevens, B.M.; D’Alessandro, A.; Reisz, J.A.; Culp-Hill, R.; Nemkov, T.; Pei, S.; Khan, N.; Adane, B.; Ye, H.; et al. Inhibition of Amino Acid Metabolism Selectively Targets Human Leukemia Stem Cells. Cancer Cell 2018, 34, 724–740. [Google Scholar] [CrossRef] [Green Version]
- Panuzzo, C.; Jovanovski, A.; Pergolizzi, B.; Pironi, L.; Stanga, S.; Fava, C.; Cilloni, D. Mitochondria: A Galaxy in the Hematopoietic and Leukemic Stem Cell Universe. Int. J. Mol. Sci. 2020, 21, 3928. [Google Scholar] [CrossRef]
- Farge, T.; Saland, E.; de Toni, F.; Aroua, N.; Hosseini, M.; Perry, R.; Bosc, C.; Sugita, M.; Stuani, L.; Fraisse, M.; et al. Chemotherapy-Resistant Human Acute Myeloid Leukemia Cells Are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism. Cancer Discov. 2017, 7, 716–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bosc, C.; Broin, N.; Fanjul, M.; Saland, E.; Farge, T.; Courdy, C.; Batut, A.; Masoud, R.; Larrue, C.; Skuli, S.; et al. Autophagy regulates fatty acid availability for oxidative phosphorylation through mitochondria-endoplasmic reticulum contact sites. Nat. Commun. 2020, 11, 4056. [Google Scholar] [CrossRef]
- Samudio, I.; Harmancey, R.; Fiegl, M.; Kantarjian, H.; Konopleva, M.; Korchin, B.; Kaluarachchi, K.; Bornmann, W.; Duvvuri, S.; Taegtmeyer, H.; et al. Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J. Clin. Investig. 2010, 120, 142–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Wu, B.; Li, Y.; Chen, J.; Ye, Z.; Tian, X.; Wang, J.; Xu, X.; Pan, S.; Zheng, Y.; et al. Amino acid catabolism regulates hematopoietic stem cell proteostasis via a GCN2-eIF2α axis. Cell Stem Cell 2022, 29, 1119–1134. [Google Scholar] [CrossRef]
- Grenier, A.; Poulain, L.; Mondesir, J.; Jacquel, A.; Bosc, C.; Stuani, L.; Mouche, S.; Larrue, C.; Sahal, A.; Birsen, R.; et al. AMPK-PERK axis represses oxidative metabolism and enhances apoptotic priming of mitochondria in acute myeloid leukemia. Cell Rep. 2022, 38, 110197. [Google Scholar] [CrossRef] [PubMed]
- Willems, L.; Chapuis, N.; Puissant, A.; Maciel, T.T.; Green, A.S.; Jacque, N.; Vignon, C.; Park, S.; Guichard, S.; Herault, O.; et al. The dual mTORC1 and mTORC2 inhibitor AZD8055 has anti-tumor activity in acute myeloid leukemia. Leukemia 2012, 26, 1195–1202. [Google Scholar] [CrossRef] [Green Version]
- Willems, L.; Jacque, N.; Jacquel, A.; Neveux, N.; Maciel, T.T.; Lambert, M.; Schmitt, A.; Poulain, L.; Green, A.S.; Uzunov, M.; et al. Inhibiting glutamine uptake represents an attractive new strategy for treating acute myeloid leukemia. Blood 2013, 122, 3521–3532. [Google Scholar] [CrossRef] [Green Version]
- Mossmann, D.; Park, S.; Hall, M.N. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat. Rev. Cancer 2018, 18, 744–757. [Google Scholar] [CrossRef]
- Ju, H.Q.; Zhan, G.; Huang, A.; Sun, Y.; Wen, S.; Yang, J.; Lu, W.H.; Xu, R.H.; Li, J.; Li, Y.; et al. ITD mutation in FLT3 tyrosine kinase promotes Warburg effect and renders therapeutic sensitivity to glycolytic inhibition. Leukemia 2017, 31, 2143–2150. [Google Scholar] [CrossRef] [Green Version]
- Lopes-Coelho, F.; Nunes, C.; Gouveia-Fernandes, S.; Rosas, R.; Silva, F.; Gameiro, P.; Carvalho, T.; Gomes da Silva, M.; Cabeçadas, J.; Dias, S.; et al. Monocarboxylate transporter 1 (MCT1), a tool to stratify acute myeloid leukemia (AML) patients and a vehicle to kill cancer cells. Oncotarget 2017, 8, 82803–82823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saulle, E.; Spinello, I.; Quaranta, M.T.; Pasquini, L.; Pelosi, E.; Iorio, E.; Castelli, G.; Chirico, M.; Pisanu, M.E.; Ottone, T.; et al. Targeting Lactate Metabolism by Inhibiting MCT1 or MCT4 Impairs Leukemic Cell Proliferation, Induces Two Different Related Death-Pathways and Increases Chemotherapeutic Sensitivity of Acute Myeloid Leukemia Cells. Front. Oncol. 2021, 10, 621458. [Google Scholar] [CrossRef] [PubMed]
- Spinello, I.; Saulle, E.; Quaranta, M.T.; Pasquini, L.; Pelosi, E.; Castelli, G.; Ottone, T.; Voso, M.T.; Testa, U.; Labbaye, C. The small-molecule compound AC-73 targeting CD147 inhibits leukemic cell proliferation, induces autophagy and increases the chemotherapeutic sensitivity of acute myeloid leukemia cells. Haematologica 2019, 104, 973–985. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Tran, L.; Tang, J.; Nguyen, V.; Sewell, E.; Xiao, J.; Hino, C.; Wasnik, S.; Francis-Boyle, O.L.; Zhang, K.K.; et al. FBP1-Altered Carbohydrate Metabolism Reduces Leukemic Viability through Activating P53 and Modulating the Mitochondrial Quality Control System In Vitro. Int. J. Mol. Sci. 2022, 23, 11387. [Google Scholar] [CrossRef] [PubMed]
- Hosseini, M.; Rezvani, H.R.; Aroua, N.; Bosc, C.; Farge, T.; Saland, E.; Guyonnet-Dupérat, V.; Zaghdoudi, S.; Jarrou, L.; Larrue, C.; et al. Targeting Myeloperoxidase Disrupts Mitochondrial Redox Balance and Overcomes Cytarabine Resistance in Human Acute Myeloid Leukemia. Cancer Res. 2019, 79, 5191–5203. [Google Scholar] [CrossRef] [Green Version]
- Aroua, N.; Boet, E.; Ghisi, M.; Nicolau-Travers, M.L.; Saland, E.; Gwilliam, R.; de Toni, F.; Hosseini, M.; Mouchel, P.L.; Farge, T.; et al. Extracellular ATP and CD39 Activate cAMP-Mediated Mitochondrial Stress Response to Promote Cytarabine Resistance in Acute Myeloid Leukemia. Cancer Discov. 2020, 10, 1544–1565. [Google Scholar] [CrossRef]
- Shi, R.; Tang, Y.Q.; Miao, H. Metabolism in tumor microenvironment: Implications for cancer immunotherapy. MedComm 2020, 1, 47–68. [Google Scholar] [CrossRef]
- Forte, D.; García-Fernández, M.; Sánchez-Aguilera, A.; Stavropoulou, V.; Fielding, C.; Martín-Pérez, D.; López, J.A.; Costa, A.S.H.; Tronci, L.; Nikitopoulou, E.; et al. Bone Marrow Mesenchymal Stem Cells Support Acute Myeloid Leukemia Bioenergetics and Enhance Antioxidant Defense and Escape from Chemotherapy. Cell Metab. 2020, 32, 829–843. [Google Scholar] [CrossRef]
- Yazdani, Z.; Mousavi, Z.; Moradabadi, A.; Hassanshahi, G. Significance of CXCL12/CXCR4 Ligand/Receptor Axis in Various Aspects of Acute Myeloid Leukemia. Cancer Manag. Res. 2020, 12, 2155–2165. [Google Scholar] [CrossRef] [Green Version]
- Mulcahy Levy, J.M.; Thorburn, A. Autophagy in cancer: Moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ. 2020, 27, 843–857. [Google Scholar] [CrossRef]
- Horne, G.A.; Stobo, J.; Kelly, C.; Mukhopadhyay, A.; Latif, A.L.; Dixon-Hughes, J.; McMahon, L.; Cony-Makhoul, P.; Byrne, J.; Smith, G.; et al. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia 2020, 34, 1775–1786. [Google Scholar] [CrossRef]
- Chude, C.I.; Amaravadi, R.K. Targeting Autophagy in Cancer: Update on Clinical Trials and Novel Inhibitors. Int. J. Mol. Sci. 2017, 18, 1279. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.; Eom, J.I.; Jeung, H.K.; Jang, J.E.; Kim, J.S.; Cheong, J.W.; Kim, Y.S.; Min, Y.H. Induction of cytosine arabinoside-resistant human myeloid leukemia cell death through autophagy regulation by hydroxychloroquine. BioMed. Pharmacother. 2015, 73, 87–96. [Google Scholar] [CrossRef]
- Haghi, A.; Salemi, M.; Fakhimahmadi, A.; Mohammadi Kian, M.; Yousefi, H.; Rahmati, M.; Mohammadi, S.; Ghavamzadeh, A.; Moosavi, M.A.; Nikbakht, M. Effects of different autophagy inhibitors on sensitizing KG-1 and HL-60 leukemia cells to chemotherapy. IUBMB Life 2021, 73, 130–145. [Google Scholar] [CrossRef]
- Dykstra, K.M.; Fay, H.R.S.; Massey, A.C.; Yang, N.; Johnson, M.; Portwood, S.; Guzman, M.L.; Wang, E.S. Inhibiting autophagy targets human leukemic stem cells and hypoxic AML blasts by disrupting mitochondrial homeostasis. Blood Adv. 2021, 5, 2087–2100. [Google Scholar] [CrossRef]
- Matsuo, H.; Nakatani, K.; Harata, Y.; Higashitani, M.; Ito, Y.; Inagami, A.; Noura, M.; Nakahata, T.; Adachi, S. Efficacy of a combination therapy targeting CDK4/6 and autophagy in a mouse xenograft model of t(8;21) acute myeloid leukemia. Biochem. Biophys. Rep. 2021, 27, 101099. [Google Scholar] [CrossRef]
- Bago, R.; Malik, N.; Munson, M.J.; Prescott, A.R.; Davies, P.; Sommer, E.; Shpiro, N.; Ward, R.; Cross, D.; Ganley, I.G.; et al. Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase. Biochem. J. 2014, 463, 413–427. [Google Scholar] [CrossRef] [Green Version]
- Meunier, G.; Birsen, R.; Cazelles, C.; Belhadj, M.; Cantero-Aguilar, L.; Kosmider, O.; Fontenay, M.; Azar, N.; Mayeux, P.; Chapuis, N.; et al. Antileukemic activity of the VPS34-IN1 inhibitor in acute myeloid leukemia. Oncogenesis 2020, 9, 94. [Google Scholar] [CrossRef]
- Putyrski, M.; Vakhrusheva, O.; Bonn, F.; Guntur, S.; Vorobyov, A.; Brandts, C.; Dikic, I.; Ernest, A. Disrupting the LC3 Interaction Region (LIR) Binding of Selective Autophagy Receptors Sensitizes AML Cell Lines to Cytarabine. Front. Cell Dev. Biol. 2020, 8, 208. [Google Scholar] [CrossRef] [Green Version]
- Du, J.; Wang, T.; Li, Y.; Zhou, Y.; Wang, X.; Yu, X.; Ren, X.; An, Y.; Wu, Y.; Sun, W.; et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic. Biol. Med. 2019, 131, 356–369. [Google Scholar] [CrossRef]
- Zhao, L.; Zhan, H.; Jiang, X.; Li, Y.; Zeng, H. The role of cholesterol metabolism in leukemia. Blood Sci. 2019, 1, 44–49. [Google Scholar] [CrossRef] [PubMed]
- Mondet, J.; Chevalier, S.; Mossuz, P. Pathogenic Roles of S100A8 and S100A9 Proteins in Acute Myeloid and Lymphoid Leukemia: Clinical and Therapeutic Impacts. Molecules 2021, 26, 1323. [Google Scholar] [CrossRef] [PubMed]
- Niu, F.; Yu, Y.; Li, Z.; Ren, Y.; Li, Z.; Ye, Q.; Liu, P.; Ji, C.; Qian, L.; Xiong, Y. Arginase: An emerging and promising therapeutic target for cancer treatment. BioMed. Pharmacother. 2022, 149, 112840. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Wu, C.L.; Wang, X.; Ban, Q.; Quan, C.; Liu, M.; Dong, H.; Li, J.; Kim, G.Y.; Choi, Y.H.; et al. SP600125 enhances C-2-induced cell death by the switch from autophagy to apoptosis in bladder cancer cells. J. Exp. Clin. Cancer Res. 2019, 38, 448. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.C.; Lin, Y.C.; Liao, Y.H.; Liou, J.P.; Chen, C.H. MPT0G612, a Novel HDAC6 Inhibitor, Induces Apoptosis and Suppresses IFN-γ-Induced Programmed Death-Ligand 1 in Human Colorectal Carcinoma Cells. Cancers 2019, 11, 1617. [Google Scholar] [CrossRef] [Green Version]
- Jang, J.E.; Eom, J.I.; Jeung, H.K.; Cheong, J.W.; Lee, J.Y.; Kim, J.S.; Min, Y.H. Targeting AMPK-ULK1-mediated autophagy for combating BET inhibitor resistance in acute myeloid leukemia stem cells. Autophagy 2017, 13, 761–762. [Google Scholar] [CrossRef] [Green Version]
- Jang, J.E.; Eom, J.I.; Jeung, H.K.; Chung, H.; Kim, Y.R.; Kim, J.S.; Cheong, J.W.; Min, Y.H. PERK/NRF2 and autophagy form a resistance mechanism against G9a inhibition in leukemia stem cells. J. Exp. Clin. Cancer Res. 2020, 39, 66. [Google Scholar] [CrossRef] [Green Version]
- Mouchel, P.L.; Serhan, N.; Betous, R.; Farge, T.; Saland, E.; De Medina, P.; Hoffmann, J.S.; Sarry, J.E.; Poirot, M.; Silvente-Poirot, S.; et al. Dendrogenin A Enhances Anti-Leukemic Effect of Anthracycline in Acute Myeloid Leukemia. Cancers 2020, 12, 2933. [Google Scholar] [CrossRef]
- Qiu, L.; Zhou, G.; Cao, S. Targeted inhibition of ULK1 enhances daunorubicin sensitivity in acute myeloid leukemia. Life Sci. 2020, 243, 117234. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Saulle, E.; Spinello, I.; Quaranta, M.T.; Labbaye, C. Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting. Cells 2023, 12, 1553. https://doi.org/10.3390/cells12111553
Saulle E, Spinello I, Quaranta MT, Labbaye C. Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting. Cells. 2023; 12(11):1553. https://doi.org/10.3390/cells12111553
Chicago/Turabian StyleSaulle, Ernestina, Isabella Spinello, Maria Teresa Quaranta, and Catherine Labbaye. 2023. "Advances in Understanding the Links between Metabolism and Autophagy in Acute Myeloid Leukemia: From Biology to Therapeutic Targeting" Cells 12, no. 11: 1553. https://doi.org/10.3390/cells12111553