MLL白血病关键靶基因的筛选和机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
[背景与目的]:白血病干细胞(Leukemia stem Cells, LSC)和正常造血干细胞(Hematopoietic Stem Cells, HSC)间的竞争与失衡伴随着白血病病程发展的全程,是决定治疗转归的关键。最新研究提示PI3K/PTEN/AKT/mTOR通路可能是纠正两者间失衡的理想干预靶点。尽管目前己发现两代mTOR复合物抑制剂,但其在临床应用中的效果并不理想。本研究应用细胞印迹高通量筛选技术,寻找mTOR复合物2特异性抑制剂。并初步探讨化合物Z1选择性降解部分混合谱系白血病(Mixed Lineage Leukemia,MLL)细胞中Rictor蛋白的分子机制,为MLL白血病的基因靶向治疗提供一定的理论基础。然而Rictor在MLL白血病中究竟扮演着什么样的角色呢?于是我们一方面探讨Rictor分子在MLL白血病细胞集落形成能力、增殖和凋亡等方面所起的作用;一方面体内研究Rictor分子在MLL白血病发病和维持中所起的作用。
     [方法]:
     1.高通量筛选技术寻找mTOR复合物2特异性抑制剂:采用细胞印迹高通量筛选(Cytoblot High Throughput Assay)技术,从华北制药集团新药研发中心小分子单体化合物文库中筛选mTOR复合物2特异性抑制剂,并进一步运用Western免疫印记法,初步验证筛选得到的小分子化合物对各种白血病细胞系中的mTOR复合物关键组分蛋白的抑制作用。
     2.化合物Z1降解Rictor蛋白分子机制的初步探讨:以THP-1细胞作为Rictor降解机制研究的模型细胞,Western免疫印迹技术检测蛋白合成抑制剂放线菌酮(Cycloheximide, CHX)和Z1联合蛋白酶体抑制剂MG-132分别作用该细胞后Rictor蛋白表达水平的变化;荧光素酶-蛋白酶体活性分析技术(Luciferase-Proteasome-Glo Cell-Based Assay)检测Z1处理后细胞蛋白酶体活性变化;构建体外泛素化模型,检测Rictor蛋白泛素化修饰情况。
     3. Rictor分子对MLL白血病细胞作用的体外实验研究:首先运用Rictor-shRNA慢病毒反义封闭技术对MLL白血病细胞、非MLL白血病细胞及正常细胞中的Rictor分子加以反义封闭;然后利用该系统所带GFP经流式分选获得GFP-Rictor-RNAi细胞;经流式细胞术、细胞免疫荧光术及Western免疫印迹法鉴定封闭效应后,通过甲基纤维素集落形成实验检测Rictor反义封闭对各种细胞集落形成能力的差别,并同时运用流式细胞术检测相应细胞增殖和凋亡情况的差别。
     4. Rictor分子对MLL白血病发病和维持影响的初步研究:建立白血病人源化NOD/SCID免疫缺陷小鼠模型;动态监测小鼠外周血hCD45+细胞及hCD45/GFP双阳性细胞比例,对异种移植是否成功加以评估,同时也对Rictor-RNAi组与其他组间抑制率进行比较;在实验的终点,比较各组小鼠的整体观以及肝脏、脾脏等器官;流式细胞仪分析各组骨髓、肝脏和脾脏hCD45/GFP双阳性细胞比例,并比较其差别;免疫组织化学染色技术检测各组肝脏、脾脏、肾脏和肺hCD45阳性细胞表达情况;RT-PCR技术检测移植小鼠外周血单个核细胞MLL融合基因表达;绘制生存曲线,评估各组小鼠生存期差别。
     [结果]:
     1.Z1化合物对部分MLL白血病细胞系中的mTOR复合物2保守组分Rictor蛋白有选择性抑制作用,而其他白血病细胞系经Z1化合物作用后仅出现mTOR复合物2下游分子磷酸化水平(如p-AKT ser473)的下降。
     2.用Western免疫印迹技术检测到,经10nmol/L CHX处理后不同时间点的THP-1细胞中Rictor蛋白均无降解现象;MG-132可以逆转Z1引起的THP-1细胞中Rictor蛋白的降解;THP-1细胞中蛋白酶体活性被Z1所抑制;GST标签的Rictor片段蛋白可以在体外被经Z1作用后的THP-1细胞粗提液(S100)泛素化。
     3.经流式细胞术鉴定,分选后各组GFP阳性细胞均在90%以上,同时细胞免疫荧光术及Western免疫印迹法均证实Rictor蛋白封闭效应明显;MLL白血病细胞Rictor-RNAi组集落形成能力明显低于NC-RNAi组(P<0.001),而非MLL白血病细胞和正常细胞中两组间差别均没有统计学意义(P>0.05);同时,MLL白血病细胞Rictor-RNAi组增殖能力也明显低于NC-RNAi组(P<0.01),在MLL白血病细胞Rictor-RNAi组中MG-132失去逆转Z1引起的细胞凋亡的作用,这些现象在其他类型的细胞中均没有出现。
     4.动态监测THP-1异种移植模型小鼠外周血hCD45+细胞随着时间的延长而比例增加,说明建模成功;THP-1感染Rictor-RNAi组模型鼠外周血hCD45+细胞比例与THP-1未感染慢病毒组以及THP-1感染NC-GFP组模型鼠外周血hCD45+细胞比例在为期9周的动态检测期均无明显差别(P>0.05),但THP-1感染Rictor-RNAi组模型鼠外周血hCD45/GFP双阳性细胞比例从第7周开始就显著低于NC-GFP对照组(P<0.01);实验监测期的终点,可见Rictor-RNAi组模型鼠整体状况好于NC-GFP对照组,且肝脏瘤结节数目体积均小于对照组;Rictor-RNAi组模型鼠肝脏、脾脏和骨髓中hCD45/GFP双阳性细胞比例均显著低于NC-GFP对照组(P<0.01);Kaplan-Meier曲线生存分析显示Rictor-RNAi组模型鼠生存时间长于对照组(P=0.033)。
     [结论]
     1.运用细胞印迹高通量筛选技术,从小分子单体化合物文库初步筛选并得到一种能够选择性降解部分MLL白血病细胞系中mTOR复合物2保守组分Rictor蛋白的特异性小分子化合物,为研究MLL白血病的靶向治疗提供了新的分子靶点。
     2. Rictor作为一种高度保守蛋白在一般情况下是很稳定的,但Z1可引起THP-1细胞中Rictor蛋白通过泛素-蛋白酶体途径(ubiquitin-proteasome pathway, UPP)降解,这种降解作用很可能是通过Z1活化该途径的E3泛素连接酶而实现的,与蛋白酶体自身活性无关。
     3. Rictor分子在MLL白血病细胞集落形成能力、增殖和凋亡调控等方面起着重要的作用,提示它可能是MLL白血病靶向治疗中的一个关键靶基因。
     4. Rictor分子是MLL白血病发病和维持所必须的。
[Background and objective]:The competition and imbalance growing state between leukemia stem cells and normal hematopoietic stem cells is the crux of therapeutical approaches and outcome of leukemia as accompanying the whole process of generation and development of leukemia. The latest research reveals that PI3K/PTEN/AKT/mTOR signaling pathway may be a new target for correcting the imbalance between leukemia stem cells and normal hematopoietic stem cells. Two generations of mTOR complex inhibitor have been developed, however, the practical clincal applications have received unfavorable outputs. In present study, we use cytoblot high throughput assay to identify cell-specific inhibitor of mTOR complex 2. Then to investigate the degradation effect of protein Rictor in parts of MLL cell lines induced by lead compound Z1, and to supply some theories for gene targeted therapy to MLL. This study was designed to investigate the role of Rictor in the colony forming, proliferation and apoptosis-regulation of MLL leukemic cells in vitro. To study the effect of Rictor in the pathogenesis and maintain of leukemia in vivo.
     [Methods]:
     1. Searching for specific inhibitor of mTOR complex 2 by developing a high-throughput cytoblot assay:specific inhibitor of mTOR complex 2 was screened from small molecule monomeric compound library of NCPC new drug research center by using cytoblot high throughput assay. Western blot was then used to determine the inhibiting effect of compound against the key component protein in mTOR complex.
     2. An approach to the mechanism of lead compound Zl in degradation protein Rictor: THP-1 cell line was taked as the cell model, Western blot was employed to detect the expressions of protein rictor after the treatment of CHX and Z1-MG-132-combined treatment, respectively; Luciferase-Proteasome-Glo Cell-Based Assay was used to measure the poteasome activity changes of THP-1 after treated by Zl; ubiquitylation in vitro model was constructed, and the situation of ubiquitylation in protein rictor was also detected.
     3. Study on the role of Rictor to MLL Ieukemic cells in vitro:Rictor-RNA interference (RNAi) by transcription of short hairpin RNAs (shRNAs) from a lentiviral system was applied to silence Rictor gene expression in MLL leukemic cells, non-MLL leukemic cells and normal cells; and then GFP-Rictor-RNAi cells were isolated by Flow cytometry using the targeting-GFP in this lentivirus RNAi system; after established by flow cytometry, cellular immunofluorescence and Western blot, methyl cellulose colony forming assay was used to detect the differences among these Rictor-RNAi groups. Meanwhile, the proliferation dynamics of DDAO-SE labeled cells was detected by flow cytometry and analyzed with ModFit software, and flow cytometry was also used to observe apoptosis of each cell group.
     4. Preliminary Study on the influence of Rictor in the pathogenesis and maintain of MLL leukemia in vivo:Mouse model of hukemia was established in NOD/SCID mice. To evaluate the effect of xenograft by dynamic monitoring the percentages of hCD45 positive cells and hCD45/GFP double positive cells in mice peripheral blood, meanwhile, it could be used as a means for evaluating transplant rates of group Rictor-RNAi distinguishing from the other two control groups. At the end point of observation post transplantation, the aspects, the sizes of livers and spleens, also with the number of tumor nodes in liver and spleen were compared among each group. Flow cytometry was used to analyze the percentages of hCD45/GFP double positive cells in bone marrow, liver and spleen. The expression of the hCD45 in the livers, spleens, kidneys and lungs of the mice post transplantation was detected by immunocytochemistry. The rearrangements of MLL gene in mice peripheral blood was detected by RT-PCR. The survival curve of THP-1 transplantation mice was draw, and used to evaluate the differences of survival time among each group by Kaplan-Meier method.
     [Results]:
     1. The screen results indicated that compound Z1 had selectively block activation on Rictor protein which was a conserved component of mTOR complex 2, this phenomenon appeared only in parts of MLL leukemia cell lines. Meanwhile, only decreased level of phosphorylated mTOR complex 2 downstream molecules (such as p-AKT ser473) was appeared in other leukemia cell lines treated by compound Z1.
     2. No degradation effect of rictor in THP-1 was detected after the treatment of 10μmol/L cycloheximide in different time points by western blot; MG-132 could reverse the degradation effect of rictor in THP-1 cell line by Z1; the activity of poteasome in THP-1 was inhibted by Z1; GST targeting Rictor fragment could be ubiquitylated by THP-1 S-100 fraction which was treated by Z1.
     3. After certified by flow cytometry, the positive rate of each sorted cell group was over 90%, meanwhile, both of cellular immunofluorescence and Western blot confirmed that protein Rictor was blocked obviously; colony forming ability of group Rictor-RNAi in MLL leukemic cells was found to significantly lower than group NC-RNAi (P< 0.001) There was no significant difference in colony forming ability between Rictor-RNAi and NC-RNAi groups in non-MLL leukemic cells and normal cells (P>0.05). Furthermore, proliferation of group Rictor-RNAi in MLL leukemic cells was also found to significantly lower than group NC-RNAi (P< 0.01).The effects of Z1 in combination with proteasome inhibitor MG-132 reversing the induced apoptosis by Z1 disappeared in group Rictor-RNAi of MLL leukemic cells, not in non-MLL leukemic cells or normal cells.
     4. The results of dynamic monitoring showed that the percentages of hCD45 positive cells in THP-1 xenograft mice peripheral blood increased with the increase of storage time, and it illustrated the success of this transplantation model. The percentages of hCD45 positive cells in THP-1 infected Rictor-RNAi group was not evidently different from those in the THP-1 blank group or THP-1 infected NC-GFP group during the nine-week dynamic monitoring period (P>0.05). However, the percentages of hCD45 positive cells in THP-1 infected Rictor-RNAi group had became significantly lower than those in the THP-1 blank or NC-GFP group from the seventh week (P<0.01). At the end point of observation post transplantation, the aspect of Rictor-RNAi group mice was better than that of THP-1 blank or NC-GFP group. Meanwhile, the sizes of the livers in Rictor-RNAi group were smaller than those in other two groups, and the numbers of liver tumor nodes in Rictor-RNAi group were less than those in other two groups. The percentages of hCD45/GFP double positive cells in mice liver nodes, spleens and bone marrow of Rictor-RNAi group were significantly lower than those of NC-GFP group(P<0.01).The analysis by Kaplan-Meier method showed that survival time of Rictor-RNAi group was longer than those of control groups (P=0.033).
     [Conclusion]:
     1. Taking cytoblot high throughput assay, a specific small molecule monomeric compound was found out which could degrade a conserved component of mTOR complex 2—Rictor selectively, and it will provide new target molecules for targeted gene therapy of MLL leukemia.
     2. A highly conserved protein Rictor is stable in normal situation, and the protein rictor could be degradated by ubiquitin-proteasome pathway(UPP), the degradation is likely to be achieved through the activation of E3 ubiquitin ligase by Z1, and has no relationship with the activation of proteasome.
     3. Rictor could play a critical role in the colony forming, proliferation and apoptosis regulation of MLL leukemia. It revealed that Rictor might be a potential key target gene in target therapy for MLL leukemia.
     4. Rictor was necessary for the pathogenesis and maintain of MLL leukemia in vivo.
引文
[1]Mathe G, Amiel JL, Schwarzenberg L, et al. Bone marrow transplantation in man. Transplant Proc,1969,1(1):16-24.
    [2]Chim CS, Kwong YL, Liang R, et al. All-trans retinoic acid (ATRA) in the treatment of acute promyelocytic leukemia (APL). Hematol Oncol,1996,14(3): 147-154.
    [3]Capriotti T. Gleevec:zeroing in on cancer. Medsurg Nurs,2002,11(6):301-304.
    [4]Min YH, Eom JI, Cheong JW, et al. Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia:its significance as a prognostic variable. Leukemia,2003,17(5):995-997.
    [5]Park S, Chapuis N, Tamburini J, et al. Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica,2010,95(5):819-828.
    [6]Yilmaz OH, Valdez R, Theisen BK, et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature,2006,441(7092): 475-482.
    [7]Sparks CA, Guertin DA. Targeting mTOR:prospects for mTOR complex 2 inhibitors in cancer therapy. Oncogene,2010,29(26):3733-3744.
    [8]Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood,2009,113(24): 6061-6068.
    [9]Krivtsov AV, Twomey D, Feng Z, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature,2006,442(7104):818-822.
    [1]Hu X, Shen H, Tian C, et al. Kinetics of normal hematopoietic stem and progenitor cells in a Notchl-induced leukemia model. Blood,2009,114(18):3783-3792.
    [2]Holmes T, O'Brien TA, Knight R, et al. Glycogen synthase kinase-3beta inhibition preserves hematopoietic stem cell activity and inhibits leukemic cell growth. Stem Cells,2008,26(5):1288-1297.
    [3]Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science,2005,307(5712):1098-1101.
    [4]Wu YT, Ouyang W, Lazorchak AS, et al. mTOR Complex 2 Targets Akt for Proteasomal Degradation via Phosphorylation at the Hydrophobic Motif. J Biol Chem,2011.
    [5]Chun KH, Kosmeder JW,2nd, Sun S, et al. Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells. J Natl Cancer Inst,2003,95(4):291-302.
    [6]Park S, Chapuis N, Bardet V, et al. PI-103, a dual inhibitor of Class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML. Leukemia,2008,22(9):1698-1706.
    [7]Dick JE. Looking ahead in cancer stem cell research. Nat Biotechnol,2009,27(1): 44-46.
    [8]Colmone A, Amorim M, Pontier AL, et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science, 2008,322(5909):1861-1865.
    [9]Yilmaz OH, Morrison SJ. The PI-3kinase pathway in hematopoietic stem cells and leukemia-initiating cells:a mechanistic difference between normal and cancer stem cells. Blood Cells Mol Dis,2008,41(1):73-76.
    [10]Marx J. Molecular biology. Cancer's perpetual source? Science,2007,317(5841): 1029-1031.
    [11]Zhang J, Grindley JC, Yin T, et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature,2006,441(7092):518-522.
    [12]Bhaskar PT, Hay N. The two TORCs and Akt. Dev Cell,2007,12(4):487-502.
    [13]Sabatini DM. mTOR and cancer:insights into a complex relationship. Nat Rev Cancer,2006,6(9):729-734.
    [14]Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov,2006,5(8):671-688.
    [15]Tamburini J, Chapuis N, Bardet V, et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia:rationale for therapeutic inhibition of both pathways. Blood,2008,111(1):379-382.
    [16]Guertin DA, Sabatini DM. The pharmacology of mTOR inhibition. Sci Signal,2009, 2(67):pe24.
    [17]Janes MR, Limon JJ, So L, et al. Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor. Nat Med,2010,16(2):205-213.
    [18]Laplante M, Sabatini DM. mTOR signaling at a glance. Journal of cell science, 2009,122(Pt 20):3589-3594.
    [19]Bentzinger CF, Romanino K, Cloetta D, et al. Skeletal muscle-specific ablation of raptor, but not of rictor, causes metabolic changes and results in muscle dystrophy. Cell Metab,2008,8(5):411-424.
    [20]Sparks CA, Guertin DA. Targeting mTOR:prospects for mTOR complex 2 inhibitors in cancer therapy. Oncogene,2010,29(26):3733-3744.
    [21]Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood,2009,113(24): 6061-6068.
    [1]Kisselev AF, Callard A, Goldberg AL. Importance of the different proteolytic sites of the proteasome and the efficacy of inhibitors varies with the protein substrate. J Biol Chem,2006,281(13):8582-8590.
    [2]Abmayr SM, Yao T, Parmely T, et al. Preparation of nuclear and cytoplasmic extracts from mammalian cells. Curr Protoc Mol Biol,2006, Chapter 12:Unit 12 11.
    [3]Sato S, Tomomori-Sato C, Parmely TJ, et al. A set of consensus mammalian mediator subunits identified by multidimensional protein identification technology. Mol Cell,2004,14(5):685-691.
    [4]Reid J, Svejstrup JQ. DNA damage-induced Defl-RNA polymerase Ⅱ interaction and Defl requirement for polymerase ubiquitylation in vitro. J Biol Chem,2004, 279(29):29875-29878.
    [5]Davydov IV, Kenten JH, Safiran YJ, et al. In vitro screening for substrates of the N-end rule-dependent ubiquitylation. Methods Enzymol,2005,399:415-432.
    [6]Wang X, Trotman LC, Koppie T, et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell,2007,128(1):129-139.
    [7]Sini P, James D, Chresta C, et al. Simultaneous inhibition of mTORC1 and mTORC2 by mTOR kinase inhibitor AZD8055 induces autophagy and cell death in cancer cells. Autophagy,2010,6(4).
    [8]Yu K, Shi C, Toral-Barza L, et al. Beyond rapalog therapy:preclinical pharmacology and antitumor activity of WYE-125132, an ATP-competitive and specific inhibitor of mTORC1 and mTORC2. Cancer Res,2010,70(2):621-631.
    [9]Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood,2009,113(24): 6061-6068.
    [10]Goldberg AL. Protein degradation and protection against misfolded or damaged proteins. Nature,2003,426(6968):895-899.
    [11]Burger AM, Seth AK. The ubiquitin-mediated protein degradation pathway in cancer:therapeutic implications. Eur J Cancer,2004,40(15):2217-2229.
    [12]Bedford L, Lowe J, Dick LR, et al. Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov,2011,10(1): 29-46.
    [13]Schulman BA, Harper JW. Ubiquitin-like protein activation by E1 enzymes:the apex for downstream signalling pathways. Nat Rev Mol Cell Biol,2009,10(5): 319-331.
    [14]Ye Y, Rape M. Building ubiquitin chains:E2 enzymes at work. Nat Rev Mol Cell Biol,2009,10(11):755-764.
    [15]David Y, Ziv T, Admon A, et al. The E2 ubiquitin-conjugating enzymes direct polyubiquitination to preferred lysines. J Biol Chem,2010,285(12):8595-8604.
    [16]Aghajan M, Jonai N, Flick K, et al. Chemical genetics screen for enhancers of rapamycin identifies a specific inhibitor of an SCF family E3 ubiquitin ligase. Nat Biotechnol,2010,28(7):738-742.
    [17]Espeseth AS, Huang Q, Gates A, et al. A genome wide analysis of ubiquitin ligases in APP processing identifies a novel regulator of BACE1 mRNA levels. Mol Cell Neurosci,2006,33(3):227-235.
    [18]Niessner H, Beck D, Sinnberg T, et al. The farnesyl transferase inhibitor lonafarnib inhibits mTOR signaling and enforces sorafenib-induced apoptosis in melanoma cells. J Invest Dermatol,2011,131(2):468-479.
    [19]Koul N, Sharma V, Dixit D, et al. Bicyclic triterpenoid Iripallidal induces apoptosis and inhibits Akt/mTOR pathway in glioma cells. BMC Cancer,2010,10:328.
    [20]Gu L, Gao J, Li Q, et al. Rapamycin reverses NPM-ALK-induced glucocorticoid resistance in lymphoid tumor cells by inhibiting mTOR signaling pathway, enhancing Gl cell cycle arrest and apoptosis. Leukemia,2008,22(11):2091-2096.
    [1]Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood,2009,113(24): 6061-6068.
    [2]Chen W, Kumar AR, Hudson WA, et al. Malignant transformation initiated by Mll-AF9:gene dosage and critical target cells. Cancer Cell,2008,13(5):432-440.
    [3]Yokoyama A, Cleary ML. Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell,2008,14(1):36-46.
    [4]Ernst P, Mabon M, Davidson AJ, et al. An Mll-dependent Hox program drives hematopoietic progenitor expansion. Curr Biol,2004,14(22):2063-2069.
    [5]Hsieh JJ, Cheng EH, Korsmeyer SJ. Taspasel:a threonine aspartase required for cleavage of MLL and proper HOX gene expression. Cell,2003,115(3):293-303.
    [6]Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science,2005,307(5712):1098-1101.
    [7]Barrette S, Douglas JL, Seidel NE, et al. Lentivirus-based vectors transduce mouse hematopoietic stem cells with similar efficiency to moloney murine leukemia virus-based vectors. Blood,2000,96(10):3385-3391.
    [8]Pearce L, Morgan L, Lin TT, et al. Genetic modification of primary chronic lymphocytic leukemia cells with a lentivirus expressing CD38. Haematologica,2010, 95(3):514-517.
    [9]Yamamoto JK, Pedersen NC, Ho EW, et al. Feline immunodeficiency syndrome--a comparison between feline T-lymphotropic lentivirus and feline leukemia virus. Leukemia,1988,2(12 Suppl):204S-215S.
    [10]Baran J, Pituch-Noworolska A, Krzeszowiak A, et al. Detection of cancer cells in the blood by FACS sorting of CD45-cells. Int J Mol Med,1998,1(3):573-578.
    [11]Basu S, Campbell HM, Dittel BN, et al. Purification of specific cell population by fluorescence activated cell sorting (FACS). J Vis Exp,2010, (41).
    [12]Miyamoto A, Shibamoto Y, Sugie C, et al. Absence of radioadaptive responses in four cell-lines in vitro as determined by colony formation assay. Kurume Med J,2006, 53(1-2):1-5.
    [13]Castilho RM, Squarize CH, Chodosh LA, et al. mTOR mediates Wnt-induced epidermal stem cell exhaustion and aging. Cell Stem Cell,2009,5(3):279-289.
    [14]Kim BC, Ryu MS, Oh SP, et al. TIS21/(BTG2) negatively regulates estradiol-stimulated expansion of hematopoietic stem cells by derepressing Akt phosphorylation and inhibiting mTOR signal transduction. Stem Cells,2008,26(9): 2339-2348.
    [15]Park S, Chapuis N, Tamburini J, et al. Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica,2010,95(5):819-828.
    [16]Liu YL, Castleberry RP, Emanuel PD. PTEN deficiency is a common defect in juvenile myelomonocytic leukemia. Leuk Res,2009,33(5):671-677.
    [17]Chiarini F, Del Sole M, Mongiorgi S, et al. The novel Akt inhibitor, perifosine, induces caspase-dependent apoptosis and downregulates P-glycoprotein expression in multidrug-resistant human T-acute leukemia cells by a JNK-dependent mechanism. Leukemia,2008,22(6):1106-1116.
    [18]Martelli AM, Tazzari PL, Tabellini G, et al. A new selective AKT pharmacological inhibitor reduces resistance to chemotherapeutic drugs, TRAIL, all-trans-retinoic acid, and ionizing radiation of human leukemia cells. Leukemia,2003,17(9):1794-1805.
    [19]Nishioka C, Ikezoe T, Yang J, et al. Blockade of mTOR signaling potentiates the ability of histone deacetylase inhibitor to induce growth arrest and differentiation of acute myelogenous leukemia cells. Leukemia,2008,22(12):2159-2168.
    [20]Guertin DA, Stevens DM, Saitoh M, et al. mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell,2009, 15(2):148-159.
    [21]Masri J, Bernath A, Martin J, et al. mTORC2 activity is elevated in gliomas and promotes growth and cell motility via overexpression of rictor. Cancer Res,2007, 67(24):11712-11720.
    [1]Kennedy JA, Barabe F. Investigating human leukemogenesis:from cell lines to in vivo models of human leukemia. Leukemia,2008,22(11):2029-2040.
    [2]Guertin DA, Stevens DM, Saitoh M, et al. mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell,2009, 15(2):148-159.
    [3]McCormack E, Bruserud O, Gjertsen BT. Review:genetic models of acute myeloid leukaemia. Oncogene,2008,27(27):3765-3779.
    [4]Ittner LM, Gotz J. Pronuclear injection for the production of transgenic mice. Nat Protoc,2007,2(5):1206-1215.
    [5]Houdebine LM. Production of pharmaceutical proteins by transgenic animals. Comp Immunol Microbiol Infect Dis,2009,32(2):107-121.
    [6]McCarthy N. Leukaemia:MLL makes friends and influences. Nat Rev Cancer,2010, 10(8):529.
    [7]Burmeister T. MLL:exploring the methylome. Blood,2010,115(23):4627-4628.
    [8]Liedtke M, Cleary ML. Therapeutic targeting of MLL. Blood,2009,113(24): 6061-6068.
    [9]Liu H, Cheng EH, Hsieh JJ. MLL fusions:pathways to leukemia. Cancer Biol Ther, 2009,8(13):1204-1211.
    [10]Hirase C, Maeda Y, Yamaguchi T, et al. mTOR inhibition and adult T-cell leukemia. Leuk Lymphoma,2009,50(4):645-647.
    [11]Park S, Chapuis N, Tamburini J, et al. Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica,2010,95(5):819-828.
    [12]Recher C, Dos Santos C, Demur C, et al. mTOR, a new therapeutic target in acute myeloid leukemia. Cell Cycle,2005,4(11):1540-1549.
    [1]Stass S, Mirro J, Melvin S, et al. Lineage switch in acute leukemia. Blood,1984, 64(3):701-706.
    [2]Mirro J, Zipf TF, Pui CH, et al. Acute mixed lineage leukemia:clinicopathologic correlations and prognostic significance. Blood,1985,66(5):1115-1123.
    [3]Mirro J, Kitchingman GR, Williams DL, et al. Mixed lineage leukemia:the implications for hematopoietic differentiation. Blood,1986,68(2):597-599.
    [4]Van den Berghe H, David G, Broeckaert-Van Orshoven A, et al. A new chromosome anomaly in acute lymphoblastic leukemia (ALL). Hum Genet,1979,46(2): 173-180.
    [5]Ernst P, Wang J, Korsmeyer SJ. The role of MLL in hematopoiesis and leukemia. Curr Opin Hematol,2002,9(4):282-287.
    [6]Yeoh EJ, Ross ME, Shurtleff SA, et al. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell,2002,1(2):133-143.
    [7]Armstrong SA, Staunton JE, Silverman LB, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet, 2002,30(1):41-47.
    [8]Kosaka Y, Koh K, Kinukawa N, et al. Infant acute lymphoblastic leukemia with MLL gene rearrangements:outcome following intensive chemotherapy and hematopoietic stem cell transplantation. Blood,2004,104(12):3527-3534.
    [9]Chen CS, Sorensen PH, Domer PH, et al. Molecular rearrangements on chromosome 11q23 predominate in infant acute lymphoblastic leukemia and are associated with specific biologic variables and poor outcome. Blood,1993,81(9): 2386-2393.
    [10]Mrozek K, Heinonen K, Lawrence D, et al. Adult patients with de novo acute myeloid leukemia and t(9; 11)(p22; q23) have a superior outcome to patients with other translocations involving band 11q23:a cancer and leukemia group B study. Blood,1997,90(11):4532-4538.
    [11]Corral J, Lavenir I, Impey H, et al. An M11-AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice:a method to create fusion oncogenes. Cell,1996,85(6):853-861.
    [12]Chen W, Li Q, Hudson WA, et al. A murine M11-AF4 knock-in model results in lymphoid and myeloid deregulation and hematologic malignancy. Blood,2006, 108(2):669-677.
    [13]Basecke J, Podleschny M, Clemens R, et al. Lifelong persistence of AML associated MLL partial tandem duplications (MLL-PTD) in healthy adults. Leuk Res,2006, 30(9):1091-1096.
    [14]Wang J, Iwasaki H, Krivtsov A, et al. Conditional MLL-CBP targets GMP and models therapy-related myeloproliferative disease. EMBO J,2005,24(2):368-381.
    [15]Tamai H, Miyake K, Takatori M, et al. Activated K-Ras protein accelerates human MLL/AF4-induced leukemo-lymphomogenicity in a transgenic mouse model. Leukemia,2011.
    [16]Forster A, Pannell R, Drynan LF, et al. Engineering de novo reciprocal chromosomal translocations associated with Mll to replicate primary events of human cancer. Cancer Cell,2003,3(5):449-458.
    [17]Chen W, Kumar AR, Hudson WA, et al. Malignant transformation initiated by M11-AF9:gene dosage and critical target cells. Cancer Cell,2008,13(5):432-440.
    [18]Slany RK, Lavau C, Cleary ML. The oncogenic capacity of HRX-ENL requires the transcriptional transactivation activity of ENL and the DNA binding motifs of HRX. Mol Cell Biol,1998,18(1):122-129.
    [19]Somervaille TC, Cleary ML. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell,2006,10(4): 257-268.
    [20]Barabe F, Kennedy JA, Hope KJ, et al. Modeling the initiation and progression of human acute leukemia in mice. Science,2007,316(5824):600-604.
    [21]Meyer C, Kowarz E, Hofmann J, et al. New insights to the MLL recombinome of acute leukemias. Leukemia,2009,23(8):1490-1499.
    [22]Ayton PM, Cleary ML. Molecular mechanisms of leukemogenesis mediated by MLL fusion proteins. Oncogene,2001,20(40):5695-5707.
    [23]Strissel PL, Strick R, Rowley JD, et al. An in vivo topoisomerase II cleavage site and a DNase I hypersensitive site colocalize near exon 9 in the MLL breakpoint cluster region. Blood,1998,92(10):3793-3803.
    [24]So CW, Cleary ML. Common mechanism for oncogenic activation of MLL by forkhead family proteins. Blood,2003,101(2):633-639.
    [25]Ida K, Kitabayashi I, Taki T, et al. Adenoviral E1A-associated protein p300 is involved in acute myeloid leukemia with t(11;22)(q23;q13). Blood,1997,90(12): 4699-4704.
    [26]Rowley JD, Reshmi S, Sobulo O, et al. All patients with the T(11;16)(q23;p13.3) that involves MLL and CBP have treatment-related hematologic disorders. Blood, 1997,90(2):535-541.
    [27]Milne TA, Martin ME, Brock HW, et al. Leukemogenic MLL fusion proteins bind across a broad region of the Hox a9 locus, promoting transcription and multiple histone modifications. Cancer Res,2005,65(24):11367-11374.
    [28]Yu M, Honoki K, Andersen J, et al. MLL tandem duplication and multiple splicing in adult acute myeloid leukemia with normal karyotype. Leukemia,1996,10(5): 774-780.
    [29]Dorrance AM, Liu S, Yuan W, et al. Mll partial tandem duplication induces aberrant Hox expression in vivo via specific epigenetic alterations. J Clin Invest,2006, 116(10):2707-2716.
    [30]Ferrando AA, Armstrong SA, Neuberg DS, et al. Gene expression signatures in MLL-rearranged T-lineage and B-precursor acute leukemias:dominance of HOX dysregulation. Blood,2003,102(1):262-268.
    [31]Kumar AR, Li Q, Hudson WA, et al. A role for MEIS1 in MLL-fusion gene leukemia. Blood,2009,113(8):1756-1758.
    [32]Hess JL, Bittner CB, Zeisig DT, et al. c-Myb is an essential downstream target for homeobox-mediated transformation of hematopoietic cells. Blood,2006,108(1): 297-304.
    [33]Nakanishi H, Nakamura T, Canaani E, et al. ALL1 fusion proteins induce deregulation of EphA7 and ERK phosphorylation in human acute leukemias. Proc Natl Acad Sci U S A,2007,104(36):14442-14447.
    [34]Armstrong SA, Kung AL, Mabon ME, et al. Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification. Cancer Cell,2003,3(2):173-183.
    [35]Wang Z, Smith KS, Murphy M, et al. Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted therapy. Nature,2008,455(7217):1205-1209.
    [36]Yokoyama A, Somervaille TC, Smith KS, et al. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell,2005, 123(2):207-218.
    [37]Li Z, Lu J, Sun M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci U S A,2008,105(40): 15535-15540.
    [38]Cozzio A, Passegue E, Ayton PM, et al. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev, 2003,17(24):3029-3035.
    [39]Huntly BJ, Shigematsu H, Deguchi K, et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell,2004,6(6):587-596.
    [40]Deguchi K, Ayton PM, Carapeti M, et al. MOZ-TIF2-induced acute myeloid leukemia requires the MOZ nucleosome binding motif and TIF2-mediated recruitment of CBP. Cancer Cell,2003,3(3):259-271.
    [41]Griffin JD, Lowenberg B. Clonogenic cells in acute myeloblastic leukemia. Blood, 1986,68(6):1185-1195.
    [42]Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood,2002,99(2):507-512.
    [43]Yilmaz OH, Valdez R, Theisen BK, et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature,2006,441(7092): 475-482.
    [44]Jin L, Hope KJ, Zhai Q, et al. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med,2006,12(10):1167-1174.
    [45]Krause DS, Lazarides K, von Andrian UH, et al. Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nat Med,2006, 12(10):1175-1180.
    [46]Dobson CL, Warren AJ, Pannell R, et al. Tumorigenesis in mice with a fusion of the leukaemia oncogene Mll and the bacterial lacZ gene. EMBO J,2000,19(5): 843-851.
    [47]Felix CA, Megonigal MD, Chervinsky DS, et al. Association of germline p53 mutation with MLL segmental jumping translocation in treatment-related leukemia. Blood,1998,91(12):4451-4456.
    [48]Mahgoub N, Parker RI, Hosler MR, et al. RAS mutations in pediatric leukemias with MLL gene rearrangements. Genes Chromosomes Cancer,1998,21(3): 270-275.
    [49]Taketani T, Taki T, Sugita K, et al. FLT3 mutations in the activation loop of tyrosine kinase domain are frequently found in infant ALL with MLL rearrangements and pediatric ALL with hyperdiploidy. Blood,2004,103(3):1085-1088.
    [50]Eguchi M, Eguchi-Ishimae M, Knight D, et al. MLL chimeric protein activation renders cells vulnerable to chromosomal damage:an explanation for the very short latency of infant leukemia. Genes Chromosomes Cancer,2006,45(8):754-760.
    [51]Zuber J, Radtke I, Pardee TS, et al. Mouse models of human AML accurately predict chemotherapy response. Genes Dev,2009,23(7):877-889.