MYELOID NEOPLASIA Caspase-3 controls AML 1-ETO – driven leukemogenesis via autophagy modulation in a ULK 1-dependent manner

The t(8;21), which leads to the expression of the AML1-ETO (AE) fusion transcription factor, represents the most frequent chromosomal translocation in acute myeloid leukemia (AML), occurring in;4% to 12% of adult and 12% to 30% of pediatric patients. The leukemogenicity of AE has been evaluated in multiple mouse models. AE-expressing transgenic mice do not develop leukemia in the absence of other secondary events, suggesting that cooperating events are required. Some mouse models of AE-driven AML have been developed, such as expression of AE in Cdkn1a-null hematopoietic stem cells (HSCs) or expression of AML1-ETO9a (AE9a), an alternatively spliced variant of AML1-ETO, in wild-type (WT) HSCs, which leads to fully penetrant AML after a prolonged latency. Our recent studies showed that bothmousemodels could accurately predict cooperating events in human t(8;21) AML. Caspase-3, an executioner caspase, plays multiple roles in cell processes, such as apoptosis, embryonic and hematopoietic development, and homeostasis. Caspase-3 has been found to be essential for normal brain development in some genetic mouse strains; however, Caspase-3–deficient mice are viable and fertile in the C57BL/6 strain with no apparent defects in brain pathology. Caspase-3 has been shown to play important roles at multiple steps in embryonic stem cells and HSCs, affecting self-renewal and differentiation. In the hematopoietic system, loss of Caspase-3 leads to accelerated proliferation and impaired differentiation of bonemarrow cells. Caspase-3 is also involved in the negative regulation of B-cell proliferation following antigen stimulation and activated Caspase-3 participates in T-cell proliferation in response to T-cell stimulation. It has been shown that uncleavedCaspase-3 levels are higher in the peripheral blood cells of AML patients compared with hematologically normal individuals, which suggests that the caspase pathway is dysregulated in AML.We and others have shown that AE is a direct substrate of Caspase-3 and the cleavage sites are TMPD188 and LLLD368.Moreover, a truncatedAE protein (DAE), generated by cleavage of AE at Asp188, worked as a dominant-negative protein by interactingwithAEand interferingwith itsoncogenic functions. Together, these data suggest that AE may accumulate in a Caspase-3 compromised background and thereby accelerate leukemogenesis. In this study, we sought to determine the role of Caspase-3 in leukemogenesis in vivo, by expressing AE9a in Caspase-3 knockout mouse model. We found that loss of Caspase-3 impaired self-renewal and

[1]  V. Vacic,et al.  Integrative genetic analysis of mouse and human AML identifies cooperating disease alleles , 2016, The Journal of experimental medicine.

[2]  D. Ferguson,et al.  Autophagy limits proliferation and glycolytic metabolism in acute myeloid leukemia , 2015, Cell Death Discovery.

[3]  John M Asara,et al.  Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates. , 2015, Molecular cell.

[4]  C. Baldus,et al.  Outlook on PI3K/AKT/mTOR inhibition in acute leukemia , 2015, Molecular and Cellular Therapies.

[5]  E. Sausville,et al.  Autophagy modulation: a target for cancer treatment development , 2015, Cancer Chemotherapy and Pharmacology.

[6]  Jessica L. Larson,et al.  Caspase-3 Deficiency Results in Disrupted Synaptic Homeostasis and Impaired Attention Control , 2015, The Journal of Neuroscience.

[7]  Jiong Hu,et al.  Autophagy Is an Important Event for Low Dose Cytarabine Treatment in Acute Myeloid Leukemia U937 Cell Line , 2014 .

[8]  T. Martinović,et al.  Idarubicin induces mTOR-dependent cytotoxic autophagy in leukemic cells. , 2014, Experimental cell research.

[9]  L. Zitvogel,et al.  An autophagy-dependent anticancer immune response determines the efficacy of melanoma chemotherapy , 2014, Oncoimmunology.

[10]  J. DeVoss,et al.  A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3 , 2014, Nature.

[11]  M. Torgersen,et al.  Autophagy: friend or foe in the treatment of fusion protein-associated leukemias? , 2013, Autophagy.

[12]  N. Engedal,et al.  Targeting autophagy potentiates the apoptotic effect of histone deacetylase inhibitors in t(8;21) AML cells. , 2013, Blood.

[13]  David R McIlwain,et al.  Caspase functions in cell death and disease. , 2013, Cold Spring Harbor perspectives in biology.

[14]  N. Yuan,et al.  Chinese a Nti鄄 Cancer a Ssociation , 2022 .

[15]  M. Overholtzer,et al.  Interaction Between FIP200 and ATG16L1 Distinguishes ULK1 Complex-Dependent and -Independent Autophagy , 2012, Nature Structural &Molecular Biology.

[16]  S. Nimer,et al.  AML1-ETO driven acute leukemia: insights into pathogenesis and potential therapeutic approaches , 2012, Frontiers of Medicine.

[17]  Sangeeta Khare,et al.  Guidelines for the use and interpretation of assays formonitoring autophagy (3rd edition) , 2016 .

[18]  Robert Clarke,et al.  Guidelines for the use and interpretation of assays for monitoring autophagy , 2012 .

[19]  Chuanfeng Wu,et al.  Targeting of AML1-ETO in t(8;21) Leukemia by Oridonin Generates a Tumor Suppressor–Like Protein , 2012, Science Translational Medicine.

[20]  N. Mizushima,et al.  The role of Atg proteins in autophagosome formation. , 2011, Annual review of cell and developmental biology.

[21]  Shengbing Huang,et al.  The Role of Autophagy in Cancer: Therapeutic Implications , 2011, Molecular Cancer Therapeutics.

[22]  D. Fingar,et al.  ULK1 inhibits mTORC1 signaling, promotes multisite Raptor phosphorylation and hinders substrate binding , 2011, Autophagy.

[23]  Guoqiang Chen,et al.  Effector Caspases and Leukemia , 2011, International journal of cell biology.

[24]  D. Tang,et al.  Autophagy regulates myeloid cell differentiation by p62/SQSTM1-mediated degradation of PML-RARα oncoprotein , 2011, Autophagy.

[25]  Quan Chen,et al.  Beclin 1 cleavage by caspase-3 inactivates autophagy and promotes apoptosis , 2010, Protein & Cell.

[26]  N. Mizushima,et al.  The role of the Atg1/ULK1 complex in autophagy regulation. , 2010, Current opinion in cell biology.

[27]  G. Smyth,et al.  ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. , 2009, Journal of immunological methods.

[28]  J. Lane,et al.  Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis , 2009, Journal of Cell Science.

[29]  D. Teachey,et al.  Mammalian target of rapamycin inhibitors and their potential role in therapy in leukaemia and other haematological malignancies , 2009, British journal of haematology.

[30]  R A Knight,et al.  Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes , 2009, Cell Death and Differentiation.

[31]  P. Greaney,et al.  The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. , 2009, Blood.

[32]  Y. Akao,et al.  Intracellular glutathione regulates sesquiterpene lactone-induced conversion of autophagy to apoptosis in human leukemia HL60 cells. , 2009, Anticancer research.

[33]  Yuh‐Cheng Yang,et al.  Platonin induces autophagy-associated cell death in human leukemia cells , 2009, Autophagy.

[34]  M. Lübbert,et al.  Complementing mutations in core binding factor leukemias: from mouse models to clinical applications , 2008, Oncogene.

[35]  Ying Zhao,et al.  Vitamin D3 Induces Autophagy of Human Myeloid Leukemia Cells* , 2008, Journal of Biological Chemistry.

[36]  M. Abdul-Ghani,et al.  Rehabilitation of a contract killer: caspase-3 directs stem cell differentiation. , 2008, Cell stem cell.

[37]  M. Kyba,et al.  Caspase activity mediates the differentiation of embryonic stem cells. , 2008, Cell stem cell.

[38]  S. Karlsson,et al.  Hematopoietic stem cell responsiveness to exogenous signals is limited by caspase-3. , 2008, Cell stem cell.

[39]  N. Mizushima,et al.  Autophagy: process and function. , 2007, Genes & development.

[40]  Ming Yan,et al.  The p21Waf1 pathway is involved in blocking leukemogenesis by the t(8;21) fusion protein AML1-ETO. , 2007, Blood.

[41]  Ming Yan,et al.  Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent antitumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. , 2007, Blood.

[42]  YongSung Kim,et al.  ATG1, an autophagy regulator, inhibits cell growth by negatively regulating S6 kinase , 2007, EMBO reports.

[43]  R. Henschler,et al.  Arsenic but not all-trans retinoic acid overcomes the aberrant stem cell capacity of PML/RARalpha-positive leukemic stem cells. , 2007, Haematologica.

[44]  Huiping Sun,et al.  Eriocalyxin B induces apoptosis of t(8;21) leukemia cells through NF-κB and MAPK signaling pathways and triggers degradation of AML1-ETO oncoprotein in a caspase-3-dependent manner , 2007, Cell Death and Differentiation.

[45]  H. Sakagami,et al.  Induction of non-apoptotic cell death by morphinone in human promyelocytic leukemia HL-60 cells. , 2006, Anticancer research.

[46]  M. Minden,et al.  Constitutive phosphorylation of the S6 ribosomal protein via mTOR and ERK signaling in the peripheral blasts of acute leukemia patients. , 2006, Experimental hematology.

[47]  Ming Yan,et al.  A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis , 2006, Nature Medicine.

[48]  C. Récher,et al.  mTOR, a new therapeutic target in acute myeloid leukemia. , 2005, Cell cycle.

[49]  S. Nimer,et al.  Effects of the leukemia-associated AML1-ETO protein on hematopoietic stem and progenitor cells , 2004, Oncogene.

[50]  C. Zeiss,et al.  Caspase-3 in postnatal retinal development and degeneration. , 2004, Investigative ophthalmology & visual science.

[51]  T. Mak,et al.  Caspase-3 regulates cell cycle in B cells: a consequence of substrate specificity , 2003, Nature Immunology.

[52]  R. Flavell,et al.  Strain‐Dependent Neurodevelopmental Abnormalities in Caspase‐3‐Deficient Mice , 2002, Journal of neuropathology and experimental neurology.

[53]  J. Downing,et al.  Expression of a conditional AML1-ETO oncogene bypasses embryonic lethality and establishes a murine model of human t(8;21) acute myeloid leukemia. , 2002, Cancer cell.

[54]  I. Weissman,et al.  AML1-ETO expression is directly involved in the development of acute myeloid leukemia in the presence of additional mutations , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[55]  Xuening Wang,et al.  Carnosic acid and promotion of monocytic differentiation of HL60-G cells initiated by other agents. , 2001, Journal of the National Cancer Institute.

[56]  D. Tenen,et al.  Analysis of the role of AML1-ETO in leukemogenesis, using an inducible transgenic mouse model. , 2000, Blood.

[57]  R. Sékaly,et al.  Early Activation of Caspases during T Lymphocyte Stimulation Results in Selective Substrate Cleavage in Nonapoptotic Cells , 1999, The Journal of experimental medicine.

[58]  J. Tschopp,et al.  Caspase Activation Is Required for T Cell Proliferation , 1999, The Journal of experimental medicine.

[59]  A. Porter,et al.  Emerging roles of caspase-3 in apoptosis , 1999, Cell Death and Differentiation.

[60]  A. Matsuura,et al.  Apg1p, a novel protein kinase required for the autophagic process in Saccharomyces cerevisiae. , 1997, Gene.

[61]  F. Fassy,et al.  Evidence for CPP32 Activation in the Absence of Apoptosis during T Lymphocyte Stimulation* , 1997, The Journal of Biological Chemistry.

[62]  Keisuke Kuida,et al.  Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice , 1996, Nature.

[63]  C. Kenific,et al.  Cellular and metabolic functions for autophagy in cancer cells. , 2015, Trends in cell biology.

[64]  G. Kroemer,et al.  NF-kappaB inhibition sensitizes to starvation-induced cell death in high-risk myelodysplastic syndrome and acute myeloid leukemia. , 2007, Oncogene.

[65]  P. Thall,et al.  Caspase 2 and caspase 3 protein levels as predictors of survival in acute myelogenous leukemia. , 1998, Blood.