用户名: 密码: 验证码:
SHIP基因在白血病发病机制中作用的研究
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:白血病是原发于造血系统的恶性肿瘤。近年来,其发病率呈增加趋势。在35岁以下人群中,白血病的死亡率占恶性肿瘤第一位,严重危害中青年人的生命,所以值得特别重视。急性白血病的发生包括了复杂的基因突变、缺失、癌基因激活及抑癌基因失活等多种分子生物学水平的异常。尽管许多基因在白血病发病中的作用及其机制已初步明确,但所有的基因异常仍不能解释全部白血病的发生机制,因此,寻找新的白血病相关基因并研究其生物学功能一直是白血病研究领域的热点之一,不仅有助于阐明白血病发生发展的机制,而且可能找到一些治疗靶位点。
     SHIP基因是继PTEN之后发现的仅在造血细胞表达的一种新的肌醇磷酸酶,其主要功能是降解PIP3成为PI3,4P2而负调控PI3K/Akt路径。本课题组多年来一直围绕SHIP基因功能与白血病发生发展关系开展研究工作,取得了一系列有意义的研究成果。我们首次报道急性白血病患者存在SHIP基因突变,并证实SHIP基因低表达和表达缺失与人白血病细胞恶性增殖密切相关,通过干扰RNA封闭K562细胞的bcr/abl融合基因使SHIP蛋白重新表达并可以逆转细胞的异常增殖表型,近年来也陆续发现和报道了一些白血病细胞中新的SHIP基因突变。这些都提示SHIP基因具有调控白血病细胞增殖、凋亡的结构基础。但是,有关SHIP基因调控白血病细胞系K562的PI3K/Akt信号传导通路的分子机理及其在抑制和/或逆转K562细胞增殖能力中的地位,以及SHIP基因突变对PI3K/Akt的调控有何改变等问题,目前国内外均无报道。本研究的目的是:(1)探讨野生型SHIP基因对人白血病细胞系K562中PI3K/Akt信号通路相关蛋白表达水平的影响;(2)构建携带突变型SHIP基因的慢病毒载体并转染K562细胞系;(3)探讨SHIP基因突变后在K562细胞中的功能变化,为进一步研究SHIP基因在白血病发病机制中的作用提供实验依据。
     第一部分野生型SHIP基因对白血病细胞系K562增殖凋亡的实验研究
     目的探讨肌醇5′磷酸酶基因SHIP对人白血病细胞系K562增殖、凋亡及细胞周期的影响,并对其可能的作用机制进行初步探讨。
     方法将携带有野生型SHIP基因和空载体的慢病毒表达载体转染人白血病细胞系K562。MTT检测细胞生长情况;流式细胞仪检测细胞转染效率和细胞周期分布;显微镜及透射电镜下观察细胞形态及超微结构改变,TUNEL、Hoechst33342荧光染色、细胞集落形成等实验检测细胞增殖及凋亡情况。实时荧光定量PCR(FQ-PCR)检测转染前后SHIP mRNA表达变化, Western Blot检测各转染组SHIP、Akt、p-Akt308、p-Akt473、Bcl-2、Bax、Bad、Bcl-xL、CyclinD1、p27kip1、p21WAF1、NFκB蛋白水平变化。Caspase活性检测试剂盒检测Caspase-3、Caspase-9活性。NFκB活性检测试剂盒检测NFκB活性变化。
     结果
     1转染48 h后,流式细胞仪检测慢病毒载体转染K562细胞效率为(74.6±5.8)%。FQ-PCR和Western blot显示,转染SHIP基因后K562细胞中SHIP呈高效表达,表明转染成功。
     2转染野生型SHIP基因的K562细胞表现为增殖受抑,增殖指数减低,克隆形成能力降低,DNA合成减少和凋亡增多,与转染空载体组和未转染组相比有统计学差异(P<0.01)。
     3野生型SHIP基因过表达导致细胞周期G0/G1期至S期和G2/M期阻滞。转染第5天,K562/wtSHIP细胞的G0/G1期百分数(60.69±8.3)%明显高于不表达SHIP的K562/FIV细胞(37.48±6.78)%和空白对照组(35.5±4.05)%。
     4转染野生型SHIP基因的K562细胞Akt308和Akt473磷酸化水平(分别为:0.125±0.016,0.351±0.064)明显低于K562/FIV细胞(Akt308:0.323±0.025,Akt473:0.788±0.102)和未转染组(Akt308:0.324±0.023,Akt473:0.801±0.095)( P<0.01),后两组间Akt-308和Akt473磷酸化水平均无统计学差异(P>0.05)。
     5野生型SHIP基因抑制IL-3诱导的K562细胞增殖,发现外源性野生型SHIP蛋白表达组K562细胞p-Akt308和p-Akt473水平增加程度明显低于对照组呈下降趋势,而总Akt表达水平无明显变化;撤除IL-3刺激后,K562/wtSHIP组细胞Akt磷酸化水平快速下降,而转染空载体组细胞Akt磷酸化水平下降缓慢,持续时间较长。
     6转染野生型SHIP基因的K562细胞能明显增加p27蛋白(0.302±0.059)和p21蛋白(0.335±0.027)表达,而cyclin D1蛋白表达(0.142±0.019)明显降低,转染空载体组的K562细胞p27、p21和cyclin D1蛋白的表达(p27:0.159±0.023;p21:0.188±0.032;cyclin D1:0.221±0.031)与未转染组(p27:0.152±0.022;p21:0.182±0.034;cyclin D1:0.228±0.033)相比无明显改变。
     7野生型SHIP基因对凋亡相关蛋白表达的影响
     7.1转染SHIP基因的K562细胞出现caspase-9和caspase-3酶活性增加[caspase-9 : 32.82±5.80 nmol/(h·mg) ; caspase-3 :16.80±1.98nmol/(h·mg)],转染空载体组的K562细胞caspase-9和caspase-3活性[caspase-9: 8.75±2.66nmol/(h·mg);caspase-3:5.48±1.31nmol/(h·mg)]与未转染组[caspase-9 : 8.22±1.25nmol/(h·mg) ; caspase-3 :5.27±0.58nmol/(h·mg)]相比无显著改变;以上结果表明野生型SHIP基因过表达可激活K562细胞caspase-9和caspase-3。
     7.2转染野生型SHIP基因能明显下调K562细胞bcl-xL蛋白表达(K562/wtSHIP组:0.109±0.015;K562/FIV组:0.385±0.043;未转染组:0.392±0.049);显著上调促凋亡蛋白bad表达(K562/wtSHIP组:1.018±0.216;K562/FIV组:0.315±0.052;未转染组:0.304±0.037);K562/wtSHIP组bax蛋白表达( 0.348±0.037 )高于K562/FIV组(0.342±0.043)和未转染组(0.339±0.032),但三组间无统计学意义(P>0.05);各组bcl-2表达无显著性差异(P>0.05)。
     7.3转染野生型SHIP基因下调K562细胞NF-κB表达(K562/wtSHIP:0.483±0.027;K562/FIV组:0.924±0.081;未转染组:0.937±0.075);各组IκBа表达水平无显著性差异(P>0.05)。K562/wtSHIP组NF-κB的活性随时间延长而降低,K562/FIV组和未转染组细胞无显著性变化。
     结论过表达野生型SHIP基因明显抑制白血病细胞系K562增殖,促进K562细胞凋亡,其机制可能通过抑制Akt磷酸化,进一步影响其下游与多种凋亡相关分子家族蛋白如Bcl-2家族、Caspase家族表达,并通过调控细胞周期相关蛋白表达使细胞周期阻滞在G0/G1期。
     第二部分SHIP基因突变体的构建
     目的急性白血病患者存在SHIP基因突变,可能与白血病发病有关,构建SHIP基因突变体(muSHIPP28L),为研究突变基因的表达和功能与白血病的发病机制奠定基础。
     方法以重组表达质粒wtSHIP为模板,用Stratagen Mutant Kit试剂盒通过PCR定点突变技术对SHIP编码序列TTC进行定点突变→CTC,突变体经测序鉴定正确,连接到慢病毒载体Receiver-Lv31(ORF不带标签)。经重组慢病毒包装,病毒滴度测定,感染K562细胞,采用FQ-PCR和Western blot方法检测SHIP mRNA和蛋白表达。
     结果
     1构建的突变型SHIPP28L经DNA测序验证,其碱基序列与设计序列完全一致。
     2感染293T细胞48 h后在荧光显微镜下观察到GFP较强表达。制备的病毒滴度达4.9×106 pfu/ml。
     3 FCM分析表达GFP绿色荧光的阳性细胞百分率,计算活细胞转染效率。悬浮培养的人白血病细胞系K562细胞转染效率分别为K562/wtSHIP:74.6%,K562/FIV:82.5%,K562/muSHIP:76.3%。
     4 FQ-PCR和Western blot方法检测转染野生型和突变型SIHP基因的K562细胞中SHIP基因高表达,而K562/FIV组和未转染K562细胞中内源性SHIP基因表达极低,说明外源性野生型和突变型SHIP转染成功。
     结论运用基因定点突变基因重组技术成功构建了SHIP基因P28L突变体,为进一步探讨SHIP基因与白血病发病机制创造条件。
     第三部分SHIP基因突变体在白血病细胞系K562细胞中的表达及其功能的研究
     目的将构建的SHIP基因突变体稳定表达于人白血病细胞系K562中,观察SHIP基因突变后在K562细胞中的功能变化,为进一步研究SHIP基因在白血病发病机制中的作用提供实验依据。
     方法FQ-PCR和Western blot鉴定野生型和突变型SHIP基因在K562细胞中的表达;MTT比较K562/wtSHIP和K562/muSHIP两组细胞生长情况;流式细胞分析比较野生型和突变型SHIP基因对细胞周期的不同影响;Hoechst33342荧光染色比较细胞凋亡情况。Western Blot比较转染野生型和突变型SHIP基因后对K562细胞Akt、p-Akt308、p-Akt473、Bcl-2、Bax、Bad、Bcl-xL、CyclinD1、p27kip1、p21WAF1、NFκB蛋白表达的调控。Caspase活性检测试剂盒检测各组Caspase-3、Caspase-9活性。利用高压液相色谱分析比较各转染组PIP3和PI3,4P2水平。
     结果
     1野生型SHIP基因明显抑制K562细胞生长,而SHIP基因突变体丧失了这种功能。流式细胞仪分析结果表明K562/wtSHIP细胞主要表现为G0/G1期细胞增多、G2/M期细胞减少,PI降低。突变型SHIP基因对K562细胞周期无明显影响。
     2 Hoechest染色发现转染野生型SHIP基因的K562细胞早期凋亡率[(38.4±3.08)%]明显高于转染SHIP基因突变子(P28L)[(8.4±1.11)%]和空载体组[(8.06±0.95)%]。SHIP基因突变子不能促进细胞凋亡。
     3 SHIP基因突变体不能下调K562细胞p-Akt308和p-Akt473的磷酸化水平(p-Akt308:0.334±0.031;p-Akt473:0.826±0.163)。
     4转染野生型SHIP基因的K562细胞周期蛋白p21和p27蛋白水平显著增高,cyclin D1水平明显降低,但转染SHIP基因突变体的K562细胞中细胞周期蛋白表达无上述改变。
     5 K562/muSHIP组细胞caspase-3和caspase-9酶活性显著低于K562/wtSHIP组(P<0.05),与K562/FIV组和未转染组相比无统计学差异(P>0.05)。
     6转染野生型SHIP基因组K562细胞bcl-xL表达降低,而SHIP基因突变体对K562细胞bcl-xL表达(0.367±0.028)无影响;K562/muSHIP基因组和未转染组、K562/FIV组间无显著性差异(P>0.05)。K562/muSHIP组细胞bad蛋白表达水平(0.322±0.063)较K562/wtSHIP组低,与K562/FIV组和未转染组相比无显著性差异(P>0.05)。K562/muSHIP组细胞bcl-2表达水平(0.155±0.035)较K562/wtSHIP组、K562/FIV组和未转染组间无统计学意义(P>0.05)。Bax在K562/wtSHIP组细胞表达水平略高于K562/muSHIP组(0.329±0.025)但无统计学差异(P>0.05)。
     7应用高压液相色谱分析结果显示:转染wtSHIP基因组K562细胞中PI(3,4,5P3水平下降,显著低于转染muSHIP组、转染空载体组和未转染组PIP3的水平(P<0.01),后三组间PIP3水平无显著性差异(P>0.05)。K562/wtSHIP组K562细胞PI(3,4)P2水平明显高于K562/muSHIP组、K562/FIV组和未转染组K562细胞PIP2水平(P<0.01),后三组细胞间PI3,4P2水平相比无统计学意义(P>0.05)。
     结论
     1 SHIP基因突变体SHIPP28L导致其增殖抑制及促凋亡功能丧失。
     2与野生型SHIP基因相比,SHIPP28L突变体导致其对K562细胞caspase-3、caspase-9和NF-κB活性的调节作用丢失。
     3 SHIP基因调控PI3K/Akt信号通路蛋白表达可能是其抑制人白血病细胞系K562增殖的重要分子机理之一,SHIP基因对K562细胞增殖的负调控作用及促凋亡作用有赖于其基因结构和功能正常。
Background: Leukemia is the most common malignant clone’s disease of hematopoietic stem cells. The incidence rate of leukemia has the tendency of step-up recent years. Leukemia is the consequences of a multistep process resulting from a series of genetic changes and incidence, and its process is related with activation of oncogene or inactivation of antioncogene. Altohugh many genes’function and machanisms in leukemia has been explicit, all novel gene can not explain all pathogenesis of all leukemia. Thereby, it has become one of the hotspots to investigate leukemia by identifying new correlative oncogene or tumor suppressor genes and studying its biologic function,and it is important for elucidate the pathogenesy of leukemia.
     SH2-containing inositol 5’-phosphatase (SHIP) is a recently identified lipid phosphatase after phosphatase and tensin homologue deleted on chromosome 10(PTEN) was observed, which hydrolyzes the 5 prime phosphate of the inositol ring from inositol 3,4,5-P3. It is mainly expressed in hematopoietic cells. Its phosphatase activity toward PIP3 has been linked to suppress several signaling pathways, including Akt, leading to the inhibition of hematopoietic cell proliferation and survival. Our previous researching works have proved that low expression and deletion of SHIP gene was closely correlated with the abnormal proliferation of patients with leukemia; lower Akt activation was detected in K562 cell lines, which was caused by the re-expresion SHIP. transfection of wild type SHIP into the K562 cells can regulate the expression of proliferative relative genes and reverse the proliferative phenotype of K562. The molecular mechanism that SHIP gene suppress leukemia may be involved in some important signal pathways. Up to now, However, there are no any reports about the exact mechanisms of SHIP gene on the regulation of PI3K/Akt signal transduction pathway, the effects of SHIP gene on suppression and/or reversion of abnormal proliferation in human leukemia cell line K562 in the world. There are also no any reports about what changes of the PI3K/Akt signal transduction regulation will happened if SHIP has site mutation in K562 cell lines. The aim of this study are as follows: (1)To detect the influence of cell proliferation and apoptosis of K562 cells transfected with wild-type SHIP gene, and to investigate the molecular mechanism of apoptosis of K562 cells transfected with wtSHIP gene; (2) To construct SHIP gene’s mutant (P28L) found in AML patients. (3) To investigate the functional changes of SHIP gene’s mutant by over-expression it into human leukemia cell line K562,and to make a preliminary discussion on the mechanism of SHIP gene abnormality in leukemia.
     Part one: Effects of wild-type SHIP on proliferation, apoptosis and cell cycle of K562
     Objective To investigate in vitro the effects of SH2-containing inositol 5’-phosphatase gene-SHIP on proliferation, apoptosis, cell cycle of human leukemia cell line K562 and to explore its possible machinesim preliminarily.
     Method Based on the results of our previous studies, with different human leumemia cell lines K562 as study samples, including K562, K562/wtSHIP, K562/FIV; the growth of K562 was observed by MTT assay; the transfection efficiency and cell cycle distribution were assessed by flow cytometry (FCM). Morphological characteristics of light microscope,cell colony fomation,TUNEL and Hoechest 33342 fluorescent staining method were also used to test proliferation and apoptosis of K562 cells. The expression level and difference of SHIP mRNA was detected by real-time fluorescent relative-quantification reverse transcriptional PCR (FQ-PCR); the the expression level and difference of SHIP, Akt, p-Akt473, p-Akt308 and the member of apoptosis family Bcl-2, Bcl-xL, Bax, Bad, cell cycle related molecular CyclinD1, p21WAF1 and p27Kip1 were detected by Western blot; Caspase-3, Caspase-9 protein activity were detected by caspase activity kits.
     Results
     1 The transfection efficiency was about 74.6±5.8% in K562 cell line after transfected with lentivirus on 48 h detected by flow cytometry. The recombinant lentivirus pReceiver-LV-SHIP has been successfully transfected into K562 cell line.
     2 The decreased ability of proliferation and DNA synthesis, cell colony fomation ability and enhanced apoptosis rate were observed in K562 cells transfected with wild-type SHIP, and the same changes had not been observed in K562 cells transfected with empty vector and untransfected K562(P<0.01).
     3 Cell cycle distribution showed the ratio of G0/G1 phase of K562 cells transfected with wild-type SHIP gene increased from (34.15±8.30)% to (60.69±8.30)%, the ratio of G2/M+S phase decreased form(65.85±9.36)% to (38.85±7.09)%, the cell cycle was arrested at G0/G1 phase.
     4 After transfection wtSHIP at the third days, western blot results showed the Akt expression levels had no change but p-Akt308 and p-Akt473 expression level (0.125±0.016 and 0.351±0.064 respectively) decreased compared with K562/FIV group ( Akt308 : 0.323±0.025 , Akt473 :0.788±0.102) and untransfected group(Akt308:0.324±0.023,Akt473:0.801±0.095)( P<0.01).
     5 wtSHIP gene can inhibit the proliferation of K562
     6 After K562 cells transfected wtSHIP, the cell cycle related molecular expression levels showed that the protein expression levels of p27、p21 (0.302±0.059、0.335±0.027 respectively) increased compared with K562 cells transfected with empty vector (p27 kip1:0.159±0.023;p21WAF1:0.188±0.032) and untransfected group (p27Kip1:0.152±0.022;p21WAF1:0.182±0.034) (P<0.05), but the protein expression of CyclinD1 (0.142±0.019)decreased (P<0.05).
     7 The effects of wtSHIP on apoptosis related proteins in human leukemia cell line K562
     7.1 After transfection wtSHIP at the third days, the caspase-9 and capsase-3 activity[caspase-9:32.82±5.80nmol/(h·mg); caspase-3: 16.80±1.98nmol/(h·mg)] up-regulated compared with K562/FIV group [caspase-9: 8.75±2.66nmol/(h·mg); caspase-3: 5.48±1.31nmol/(h·mg)] and untransfected K562 cells caspase-9: 8.22±1.25nmol/(h·mg);caspase-3: 5.27±0.58nmol/(h·mg)] (P<0.05).
     7.2 The bcl-xL protein expression level in K562/wtSHIP group was lower than K562/FIV group and untransfected K562 (K562/wtSHIP: 0.109±0.015; K562/FIV: 0.385±0.043; untransfected group: 0.392±0.049)(P<0.05). The protein expression levels of Bad in K562/wtSHIP was significantly higher than that of K562/FIV and untransfected K562, but no significant difference of protein expression level of Bad was observed among K562/FIV and untransfected group(K562/wtSHIP: 1.018±0.216; K562/FIV: 0.315±0.052; untransfected group: 0.304±0.037). No significant difference of protein expression levels of bax and bcl-2 was observed among K562/wtSHIP、K562/FIV and untransfected K562.
     7.3 In untransfected group, FIV-GFP group, and wtSHIP-GFP group the protein expression of NF-κB (p65) was significantly down-regulated than that in K562/FIV and untransfected group(K562/wtSHIP:0.483±0.027;K562/FIV组:0.924±0.081;untransfected group:0.937±0.075)(P<0.05); and no significant difference of IκBаprotein level was observed among K562/wtSHIP、K562/FIV and untransfected K562.
     Conclusions
     1 This part of study indicated that over expression wild-type SHIP gene could remarkably suppress proliferation of K562 cells, and induce K562 cell apoptosis.
     2 The effects of wild type SHIP gene on K562 probably via inhibiting the phosphorylation of Akt, and regulating its downstream target protein such as apoptosis related family expression such as Bcl-2 family, Caspase family and the cell cycle related melbourne expression.
     Part two: Contruction of a lentiviral vector carrying human SHIP mutated gene and its expression in K562 cell line
     Objective To construct lentiviral vector carrying human SHIP gene and investigate its expression in K562 cell line.
     Method Full-length cDNAs of wild type SHIP gene (wt-SHIP) was amplified from the human chondrocytes by RT-PCR, and site-directed mutagenesis was used to obtain full-length cDNAs of the mutated SHIP gene (mut28P/L). The products was cloned into p-Receiver-LV31vector and their sequences was analysed, then subcloned into lentivirus vector pReceiver-LV31, the inserted fragments were analysed and reconfirmed by restriction endonuclease analysis and sequencing. SHIP-lentivirus gene containing green fluorescent protein gene (SHIP-GFP) or the empty vector (FIV-GFP) was transfected into K562 cells. The green fluorescence protein expression of lentivirus plasmids were observed using laser scanning microscopy after 48 hours of transfection. The mRNA expression levels of wtSHIP, muSHIP gene were detected by FQ-PCR. The protein expression levels of wtSHIP and muSHIP were detected by western blot. .
     Results
     1 By restriction endonuclease analysis and sequencing, the sequences of SHIPP28L was consistent with that in literature, and the opening frames were matched with the vector sequence.
     2 Green fluorescent can be seen in transfected 293T after transfection at 48 h. The virus titer was 4.9×106 pfu/ml.
     3 Green fluorescence signal was distributed throughout K562 cells transfected with wtSHIP and muSHIP vector.The transfection efficiency was K562/wtSHIP-74.6%、K562/FIV-82.5%、K562/muSHIP-76.3%, respectively after transfected with lentivirus on the second day.
     4 SHIP mRNA and protein were at the highly expression levels in K562/wtSHIP and K562/muSHIP, but in K562/FIV and untransfected group SHIP mRNA and protein level was extremely low, which shown that is transfected into K562 successfully.
     Conclusions
     1 Lentivirus expression vector of SHIP gene’s mutant of P28L was successfully constructed by using site-directed mutagenesis of SHIP cDNA.
     2 It was proved that the sequence of the SHIPP28L mutant was correct DNA sequencing.
     3 Green fluorescent、FQ-PCR and Western blot can be seen in transfected K562 with muSHIP-GFP and wtSHIP-GFP, which shown that it was transfected into K562 successfully.
     Part three:The study of mutation of SHIP in molecular pathogenesis of leukemia
     Objective There are two categories of mutations or gene rearrangements which play important role in molecular mechanisms of leukemogenesis. One class of mutations or gene rearrangements involving mostly tyrosine kinase gene such as BCR/ABL of chronic myelogeous leukemia (CML) confer a proliferative and/or survival advantage to hematopoietic progenitors, and second class of mutations involving mostly transcription factor associated with hematopoietic cell differentiation such as PML/RAR mutation serve primarily to impair hematopoietic differentiation and subsequent apoptosis of cells. Though FLT3-ITD mutation and PML/RAR in acute promyelocytic leukemia (APL) patients as well as c-KIT mutation and AML1/ETO in AML-M2b patients have been reported, the molecular defect involving two categories of mutation are largely unidentified. In this part we focused on elucidate the role of SHIP mutation in leukemogenesis, its association with biological characters of leukemia and its potential in developing new target therpy.
     Method The recombined lentivirus plasmids muSHIPP28L was transfected stably into human leukemia cells K562. The cell proliferation, cell life cycle and cell apoptosis of K562/muSHIP cells were determined by MTT, fluorescent staining and flow cytometry. Differentially expressed genes were reconfirmed by Western blot.
     Results
     1 The enhanced ability of proliferation and DNA synthesis, and decreased apoptosis rate was observed in K562 cells transfected with muSHIPP28L, and the same changes had been observed in K562/FIV and untransfected K562 cells. Increased G0/G1 cells was observed in K562/wtSHIP cells by flow cytometry result, but no such a difference was observed in K562/muSHIP.
     2 The early apoptosis rate in K562/muSHIP(P28L) group[(8.4±1.11)%] was significantly lower than that in K562/wtSHIP group[(38.4±3.08)%] by Hoechest stainging, no significant difference of the early apoptosis rate was observed between K562/muSHIP and K562/FIV[(8.06±0.95)%]. The SHIP mutant could not induce K562 cell apoptosis.
     3 SHIPP28L mutant can’t down-regulate phosphorylation of Akt308 and 473(p-Akt308:0.334±0.031;p-Akt473:0.826±0.163).
     4 The cell cycle related molecular expression levels of p21WAF1 and p27KIP1 in K562 cells transfected with wtSHIP were significantly up-regulated, but the protein expression of cyclin D1 down-regulated; no changes indicated above in K562/muSHIP.
     5 The activity of caspase-3 and capase-9 in K562/muSHIP group were significantly lower than those in K562/wtSHIP group(P<0.05), no significant difference was seen between K562/FIV group and untranfected group(P>0.05).
     6 The protein expression of bcl-xL decreased in K562/wtSHIP group, but no changes indicated above in K562/muSHIP(0.367±0.028). The expression of bad protein in K562/muSHIP group (0.322±0.063) was lower than that in K562/wtSHIP group , no changes indicate among K562/muSHIP、K562/FIV group and untransfected group(P>0.05). There was no significant difference of bcl-2 expression was observed among K562/muSHIP group、K562/wtSHIP group、K562/FIV group and untransfected group ( P > 0.05 ) . The expression of bax protein in K562/wtSHIP group is a little higher than that in K562/muSHIP group(0.329±0.025), but no significant difference between them(P>0.05).
     7 High pressure liquid chromatography result shown that the PI3,4,5-P3 level was decreased in K562/wtSHIP,no changes above indicate above in K562/muSHIP group、K562/FIV group and untransfected group. The PI3,4P2 level in K562/wtSHIP group was significantly higher than that in K562/muSHIP group、K562/FIV group and untransfected group(P<0.01), but no significant difference of PIP3 and PI3,4P2 were seen among the last three group(P>0.05).
     Conclusions
     1 Site-directed mutation of SHIP gene did not influence on the expression level of gene protein related to the PI3K/Akt signal pathway in the human leukemia cell line K562, however, also decrease ability of apoptosis of K562.
     2 There are significant difference of caspase-3、caspase-9 and NFκB activity between K562 cells transfected by wild-type SHIP and SHIPP28L mutant.
     3 The effects of SHIP gene on the regulation the protein expression in the typical PI3K/Akt signal pathway might be an important molecular mechanism that SHIP gene suppress the proliferation and induce the apoptosis of K562, and it depend on the structure and function of normal.
引文
1 Vanhaesebroeck B, Waterfield MD. Signaling by distinct classes of phosphoinositide 3-kinases. Exp Cell Res, 1999, 253:239~254
    2 Geier SJ, Algate PA, Carlberg K, et al. The human SHIP gene is differentially expressed in cell lineages of the bone marrow and blood. Blood, 1997, 89:1876~1885
    3 Helgason CD, Antonchuk J, Bodner C, et al. Homeostasis and regeneration of the hematopoietic stem cell pool are altered in SHIP-deficient mice. Blood, 2003, 102:3541~3547
    4 Kisseleva MV, Cao L, Majerus PW. Phosphoinositide-specific inositol phlyphosph ate 5-phosphataseⅣinhibits Akt/protein kinase B phosphorylation and leads to apoptotic cell death. J Biol Chem, 2002, 277:6266~6272
    5 Liu QR, Sasaki T, Kozieradzki I, et al. SHIP is negative regulator of growth factor receptor-mediated PKB/Akt activation and myeloid cell survival. Genes Dev, 1999, 13:786~791
    6 Luo JM, Yoshida H, Komura S, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17:1~8
    7 Luo JM (罗建民), Liu ZL, Hao HL, et al. Mutation analysis of SHIP gene in acute leukemia. Chin J Hematol (中华血液学杂志), 2004, 25:385~388
    8 Ren JH (任金海),Luo JM,Yang JC, et al. The expression of SHIP in leukemia cells and its effect on Akt phosphorylation. Chin J Hematol (中华血液学杂志), 2007, 28:844~845
    9 Liu XJ (刘小军), Luo JM, Yang L, et al. The expression changes of SHIP in the chronic mylogenous leukemia and its possible reason. MedicalJournal of Chinese People’s Liberation Army (解放军医学杂志), 2007, 33:701~703
    10 Xu Q, Simpson SE, Scialla TJ, et al. Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood, 2003, 102:972~980
    11 Kubota Y, Ohnishi H, Kitanaka A, et al. Constitutive activation of PI3K is involved in the spontaneous proliferation of primary acute myeloid leukemia cells:direct evidence of PI3K activation. Leukemia, 2004, 18:1438~1440
    12 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, 17:995~997
    13 Zhang SJ (张苏江),Li JY,Shi JY,et al. Study of mutation and single nucleotide polymorphism of PDGFβand SHIP gene in acute myeloid leukemia. Chin JHematol(中华血液学杂志), 2006, 27:383~385
    14 Chang F, Lee JT, Navolanic PM, et al. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy. Leukemia, 2003, 17:590~603
    15 Xu Q, Simpson SE, Scialla TJ, et al. Survival of acute myeloid leukemia cells requries PI3 kinase activition. Blood, 2003, 102: 972~980
    16 Emilio Hirsch, Carlotta Costa, Elisa Ciraolo. Phosphoinositide 3-kinases as a common platform for multi-hormone signaling. Jourmal of Ensocrinology, 2007, 194:243~256
    17 Blagosklonny MV, Pardee AB. The restriction point of the cell cycle. Cell Cycle, 2000, 1:103~110
    18 Grandage VL, Gale RE, Linch DC,et al. PI3-kinase/Akt is constitutively active in primary acute myeloid leukaemia cells and regulates survival and chemoresistance via NF-kappaB, Mapkinase and p53 pathways. Leukemia, 2005, 19:586~594
    19 Gloire G, Charlier E, Rahmouni S, et al. Restoration of SHIP-1 activity in human leukemic cells modifies NF-κB activation pathway and cellular survival upon oxidative stress. Oncogene, 2006, 25:5485~5494
    20 Poter AG, Janicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ, 1999, 6: 99~104
    21 Persad S, Attwell S, Gray V, et al. Rrgulation of protein kinase B/Akt-serine 473 phosphorylation by intrgrin-linked kinase: critical roles kinase activity and amino acids arginine 211 and serine 343. J Biol Chem, 2001, 276:27462~27469
    22 Rapoport AP, Simons-Evelyn M, Chen T, et al. Flavopiridol induces apoptosis and caspase-3 activation of a newly characterized Burkitt's lymphoma cell line containing mutant p53 genes. Blood Cells Mol Dis, 2001, 27:610~624
    23 Shimizu S, Ide T, Yanagida T. Electrophysiological study of a novel large pore formed by Bax and the voltage dependent anion channel that is permeable to cytochrome c. J Biol Chem, 2000, 275: 12321~12325
    24 Siskind LJ, Feinstein L, Yu T,et al. Anti-apoptotic Bcl-2 family proteins disassemble ceramide channels. J Biol Chem, 2008, 58:2172~2179
    25 Perkins C, Kin CN, Fang G, et al. Arsenic induces apoptosis of multidrug-resistant human myeloid leukemia cells that express Bcr-Abl or overexpress MDR, MRP, Bcl-2, or Bcl-x(L). Blood, 2000, 95:1014~1022
    1 Liu QR, Sasaki T, Kozieradzki I, et al. SHIP is negative regulator of growth factor receptor-mediated PKB/Akt activation and myeloid cell survival. Genes Dev, 1999, 13: 786~791
    2 Kisseleva MV, Cao L, Majerus PW. Phosphoinositide-specific inositol phlyphosph ate 5-phosphataseⅣinhibits Akt/protein kinase B phosphorylation and leads to apoptotic cell death. J Biol Chem, 2002, 277:6266~6272
    3 Luo JM, Yoshida H, Komura S, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17:1~8
    4 Luo JM (罗建民), Liu ZL, Hao HL, et al. Mutation analysis of SHIP gene in acute leukemia. Chin J Hematol (中华血液学杂志), 2004, 25:385~388
    5贾晓辉,罗建民,等白血病患者SHIP基因表达.临床荟萃, 2008, 23: 4~8
    6 Ren JH (任金海),Luo JM,Yang JC, et al. The expression of SHIP in leukemia cells and its effect on Akt phosphorylation. Chin J Hematol (中华血液学杂志), 2007, 28:844~845
    7 Liu XJ (刘小军), Luo JM, Yang L, et al. The expression changes of SHIP in the chronic mylogenous leukemia and its possible reason. Medical Journal of Chinese People’s Liberation Army (解放军医学杂志), 2007, 33:701~703
    8任金海,罗建民,杜行严,等伊马替尼对K562细胞内SHIP基因表达的影响及促凋亡作用.肿瘤, 2007, 27:853~856
    9 Kono M, Miyaza KIG, Nakamura H, et al. Site-directed mutagenesis in hemoglobin: at tempts to control the oxygen affinity with cooperativety preserved. Protein, 1998, 11:199~204
    10 Colosimo A, Xu Z, Novelli G, et al. Simple version of megaprimer PCR for site-directed mutagenesis. Biotechniques, 1999, 26:870~873
    11 Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science, 1996, 272:26~267
    12 Naldini L, Blomer U, Gage FH, et al. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Nat1 Acad Sci USA, 1996, 92:11382~11388
    13 Zufferey R, Naby D, Mandel RJ, et al. Mutiply attenuated lentiviral vector achieved efficient gene delivery in vivo. Nat Biotechnol, 1997, 15:871~875
    14 Miyoshi H, Takshashi M, Gage FH, et al. Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector. Proc Natl Acad Sci USA, 1997, 94:10319~10323
    15 Goldman MJ,Lee PS, Yang JS, et al. Lentiviral vectors foe gene therapy of cystic fibrosis. Hum Gene Ther, 1997, 8:2261~2268
    16 Dull T, Zufferey R, Kelly M, et al. A third-generation lentiviral vectors with a conditional packaging system. J Virol, 1998, 72:8463~8471
    17 Buchschacher GL Jr, Wong-Sstaal F. Development of lentiviral vectors for gene therapy for human diseases. Blood, 2000, 95:2499
    18 Burns JC, Friedmann T, Driever W, et al. Vesiculai stomatitis virus G glycoprotein pseudotyped retoviral vectors:concentration to a very high titer and efficient gene transfer into mammalian and nonmammalian cells. Proc Natl Acad Sci USA, 1993, 90:8033~8037
    19 Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003, 4:346~358
    20 Rubinson DA, Dillon CP, Kwiatkowski AV, et al. A lentivirus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference. Nat Genet, 2003, 33:401~406
    21 Liau SS, Ashley SW, Whang EE. Lentivirus-mediated RNA interference of HMGA1 prmotes chemosensitivity to gemcitabine in pancreaticadenocarcinoma. J Gastrointest Surg, 2006, 10:1254~1263
    22 Aggerholm A, Gronbaek K, Guldberg P, et al. Mutational analysis of the tumor suppressor gene MMAC1/PTEN in malignant myeloid disorders. Eur J Haematol, 2000, 65:109~113
    23 Yang J, Liu J, Zheng J, et al. A reappraisal by quantitative flow cytometry analysis of PTEN expression in acute leukemia. Leukemia, 2007, 21:2072~2074
    24 Mochizuki H, Schwartz JP, Tanaka K, et al. Higher-titer human immunodeficiency virus type 1-based vector systems for gene delivery into nondividing cells. J Virol, 1998, 72:8873~8883
    25 Kafri T, H van Prang, Ouyang L, et al. A packaging cell line for lentiviral vectors. J Virol, 1999, 73:576~584
    1 Warner JK, Wang JC, Takenaka K, et al. Direct evidence for cooperating genetic events in the leukemic transformation of normal human hematopoietic cells. Leukemia, 2005, 19:1794~1805
    2 Tonks A, Tonks AJ, Pearn L, et al. Expression of AML1-ETO in human myelomonocytic cells selectively inhibits granulocytic differentiation and promotes their self-renewal. Leukemia, 2004, 18:1238~1245
    3 Nanri T, Matsuno N, Kawakita T, et al. Mutations in the receptor tyrosine kinase pathway are associated with clinical outcome in patients with acute myeloblastic leukemia harboring t(8;21)(q22;q22). Leukemia, 2005, 19:1361~1366
    4 Schwable J, Choudhary C, Thiede C, et al. RGS2 is an important target gene of Flt3-ITD mutations in AML and functions in myeloid differentiation and leukemic transformation. Blood, 2005, 105:2107~2114
    5 Helgason CD, Antonchuk J, Bodner C, et al. Homeostasis and regeneration of the hematopoietic stem cell pool are altered in SHIP-deficient mice. Blood, 2003, 102:3541~3547
    6 J-M Luo, H Yoshida, S Komura, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17:1~8
    7罗建民,刘泽林,郝洪岭,等SHIP基因在急性白血病中的突变分析.中华血液学杂志, 2004, 25:385~388
    8张苏江,李建勇,施静艺,等急性髓系白血病患者PDGFβ和SHIP基因突变及其单核苷酸多态性研究.中华血液学杂志, 2006, 27:383~385
    9 Yu WM, Daino H, Chen J, et al. Effects of a leukemia-associated gain-of-function mutation of SHP-2 phosphatase on interleukin-3 signaling. J Biol Chem, 2006, 281:5426~5434
    10 Wang S, Yu WM, Zhang W, et al. Noonan syndrome/leukemia-associated gain-of-function mutations in SHP-2 phosphatase (PTPN11) enhance cell migration and angiogenesis. J Biol Chem, 2009, 284:913~920
    11 Helgason CD, Antonchuk J, Bodner C, et al. Homeostasis and regeneration of the hematopoietic stem cell pool are altered in SHIP-deficient mice. Blood, 2003, 102:3541~3547
    12贾晓辉,罗建民,韩颖,等白血病患者SHIP和其他相关基因表达及其意义.临床荟萃, 2008, 1:4~8
    13 Christopher P, Cheryl D, Helgason R, et al. The inositol 5’-phosphatase SHIP-1 and the src kinase Lyn negatively regulate macrophage colony-sitmulating factor-induced Akt activity. J Biol Chem, 2003, 278: 38628~38636
    14 Martin S, Shalini V, Yuri B, et al. SHIP1, an SH2 domain containing phoyinositol-5-phosphatase, regulates migration through two critical tyrosine residues and forms a novel signaling complex with DOK2 and CRKL. J Biom Chem, 2001, 26:2451~2458
    15 Rizzo MG, Giombini E, Diverio D, et al. Analysis of p73 expression pattern in acute myeloid leukemias: lack of DeltaN-p73 expression is a frequent feature of acute promyelocytic leukemia. Leukemia, 2004, 18:1804~1809
    16 Sujobert P, Bardet V, Cornillet-Lefebvre P, et al. Essential role for the p110δisoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. Blood, 2005, 106:1063~1066
    17 Kelly LM, Gilliland DG. Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet, 2002, 3:179~198
    18 Kubota Y, Ohnishi H, Kitanaka A, et al. Constitutive activation of PI3K is involved in the spontaneous proliferation of primary acute myeloid leukemia cells: direct evidence of PI3K activation. Leukemia, 2004, 18:1438~1440
    19 Sujobert P, Bardet V, Cornillet-Lefebvre P, et al. Essential role for the p110δisoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. Blood, 2005, 106:1063~1066
    20 Sampath D, Cortes J, Estrov Z, et al. Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood, 2006, 107:2517~2524
    21 Kisseleva MV, Cao L, Majerus PW, et al. Phosphoinositide-specific inositol polyphosphate 5-phosphataseⅣinhibits Akt/protein kinase B phosphorylation and leads to apoptotic cell death. J Biol Chem, 2002, 277: 6266~272
    22 Christopher P, Cheryl D, Helgason R, et al. The inositol 5’-phosphatase SHIP-1 and the src kinase Lyn negatively regulate macrophage colony-sitmulating factor-induced Akt activity. J Biol Chem, 2003, 278: 38628~8636
    1 Aman MJ, Lamkin H, Okada T, et al. The inositol hosphatase SHIP inhibits Akt/PKB activation in B cells. J Biol Chem, 1998, 273: 33922~33928
    2 Odai H, Sasaki A, Iwamatsu K, et al. Purification and molecular cloning of SH2- and SH3-containing inositol polyphosphate-5-phosphatase, which is involved in the signaling pathway of granulocyte-macrophage colony stimulating factor, erythropoietin, and Bcr-Abl. Blood, 1997, 89: 2745~2756
    3 Damen JE, Liu L, Cutler RL, et al. Erythropoietin stimulation with Grb2 and a 145-kDa protein induced to associate with Shc by multiple cytokines is an inositol tetraphatase. Proc Natl Acad Sci, 1993, 93:1689~1693
    4 Chacko GW, Tridandapani S, Damen JE, et al. Negative signaling in B lymphocytes induces tyrosine phosphorylation of the 145-kDa inositol polyphosphate 5-phosphatase, SHIP. J Immunol, 1996, 157:2234~2238
    5 Lamkin TD, Walk SF, Liu L, et al. Shc interaction with Src homology 2 domain containing isositol phosphatase (SHIP) in vivo requires the Shc-phosphotyrosine binding domain and two specific phosphotyrosines on SHIP. J Biol Chem, 1997, 272:10396~10401
    6 Lucas DM, Rohrschneider LR. A novel spliced form of SH2-containing inositol phosphatase is expressed during myeloid development. Blood, 1999, 93:1922~1933
    7 Damen JE, Liu L, Ware MD, et al. Multiple forms of the SH2-containing inositol phosphatase, SHIP, are generated by C-terminal truncation. Blood, 1998, 92:1199~1205
    8 Mancini A, Koch A, Wilms R, et al. The SH2- containing inosital 5- phosphatase (SHIP)-1 is implicated in the control of cell– cell junction and induces dissociation and dispersion of MDCK cells. Oncogene, 2002, 21:1477~1482
    9 Schaffhausen B. SH2 domain structure and function. Biochim Biophys, 1995, 1242:61-75
    10 Lioubin MN, Algate PA, Tsai S, et al. p150SHIP, a signal transduction molecule with inositol polyphosphate-5-phosphatase activity. Genes & Dev, 1996, 10: 1084~1095
    11 Geier SJ, Algate PA, Carlberg K, et al. The human SHIP gene is differentially expressed in cell lineages of the bone marrow and blood. Blood, 1997, 89:1876~1885
    12 Ware MD, Rosten P, Damen JE, et al. Cloning and characterization of human SHIP, the 145-Kd inositol 5-phosphatase that associates with SHC after cytokine stimulation. Blood, 1996, 88:2833~2840
    13 Sly LM, Rauh MJ, Kalesnikoff J, et al. SHIP, SHIP2, and PTEN activities are regulated in vivo by modulation of their protein levels: SHIP is up-regulated in macrophages and mast cells by lipopolysaccharide. Exp Hematol, 2003, 31:1170~1181
    14 Liu Q, Sasaki T, Kozieradzki I, et al. SHIP is a negative regulator of growth factor receptor-mediated PKB/Akt activation and myeloid cellsurvival. Genes Dev, 1999, 13:786~791
    15 Christopher P. Susheela Tridandapani Cheryl D, Helgason R, et al. The Inositol 5-Phosphatase SHIP-1 and the Src Kinase Lyn Negatively Regulate Macrophage Colony-stimulating Factor-induced Akt Activity. The Journal Of Biological Chemistry, 2003, 278:38628~38636
    16 Sasaki T, Suzuki A, Sasaki J, et al. Phosphoinosintide 3-kinases inimmunity: lesson from knockout mice. J Bio Chem, 2002, 131:495~501
    17 Jianmin L, Yoshida H, Komura S, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17, 1~8
    18 Liu Q, Shalaby F, Jones J, et al. The SH2-containing inositol polyphosphate 5-phosphatase, SHIP, is expressed during hematopoiesis and spermatogenesis. Blood, 1998, 91: 2753~2759
    19 Bruyns C, Pesesse X, Moreau C, et al. The two SH2-domain-containing inositol 5-phosphatases SHIP1 and SHIP2 are coexpressed in human T lymphocytes. Biol Chem, 1999, 380:969-974
    20 Pesesse X, Deleu S, Smedt D, et al. Identification of a second SH2-domain-containing protein closely related to the phosphatidylinositol polyphosphate 5-phosphatase SHIP. Biochem Biophys Res Comm, 1997, 239:697~700
    21 Wisniewski D, Strife A, Swendeman S, et al. A novel SH2-containing phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase(SHIP2) is constitutively tyrosine phosphorylated and associated with src homologous and collagen gene (SHC) in chronic myelogenous leukemia progenitor cells. Blood, 1999, 93:2707~2720
    22 Hanada M, Feng J, Hemmings BA. Structure, regulation and function of PKB/Akt--a major therapeutic target. Biochim Biophys Acta, 2004, 1697:3~16
    23 Kruosaki T. Genetic analysis of B cell antigen receptor signaling. Anna Rev Immunol, 1999, 17:555-592
    24 Maresco DL, Osborne JM, Cooney D, et al. The SH2-containing 5’-inositol phosphatase (SHIP) is tyrosine phosphorylated after Fc gammareceptor clustering in monocytes. J Immunol, 1999, 162:458~6465
    25 Sattler M, Salgia R, Shrikhande G, et al. The phosphatidylinositol polyphosphate 5-phosphatase SHIP and the protein tyrosine phosphatase SHP2 form a complex in hematopoietic cells which can be regulated by BCR/ABL and growth factors. Oncogene, 1997, 15:2379~2384
    26 J-M Luo, H Yoshida, S Komura, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17, 1~8
    27 Tridandapani S, Pradhan M, LaDine JR, et al. Proein interactions of Src homology 2 (SH2) domain-containing inositol phosphatase (SHIP) : Association with Shc displaces SHIP from FcγRⅡb in B cells. J Immunnol, 1999, 162:1408~1414
    28 Tridandapani S, Chacko GW, Van Brocklyn JR, et al. Negative signaling in B cells causes reduced Ras activity by reducing Shc-Grb2 interactions. J Immunol, 1997, 158:11125~1132
    29 Javad Aman M, Thomas D, Okada L, et al. The Inositol Phosphatase SHIP Inhibits Akt/PKB Activation in B Cells. J Biol Chem, 1998, 273:33922~33928
    30 Sawada T, Hamano N, Satoh H, et al. Mutation analysis of PTEN/MMAC1 gene in Japanese patients with Cowden disease. Japn J Cancer Res, 2000, 91:700~705
    31 Podsypanina K, Ellenson LH, Nemes A, et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systerm. Proc Natl Acad Sci, 1999, 96:1563~1568
    32 Brauweiler A. Differential regulation of B cell development, activation, and death by the 5’inositol phosphatase SHIP. J Exp Med, 2000, 191:1545~1554
    33 Sato K, Ochi A. Superclustering of B cell receptor and FcγRⅡB1 activates Src homology 2-containing protein tyrosine phosphatase-1. J Immunol, 1998, 161:2716~2722
    34 Yamanashi Y. Role of the rasGAP-associtated docking protein p62(dok) in negative regulation of B cell receptor-mediated signaling. Genes Dev, 2000, 14:11~16
    35 Horn S, Meyer J, Heukeshoven J, et al. The inositol 5-phosphatase SHIP is expressed as 145 and 135 kDa proteins in blood and bone marrow cells in vivo, whereas carboxyltruncated forms of SHIP are generated by proteolytic cleavage in vitro. Leukemia, 2001, 15:112~120
    36 Kalesnikoff J, Baur N, Leitges M, et al. J Immunol, 2002, 168:4737~4746
    37 Sly LM, Rauh MJ, Kalesnikoff J, et al. LPS-induced upregulation of SHIP is essential for endotoxin tolerance. Immunity, 2004, 21:227~239
    38 Mantovani A, Sozzani S, Locati M, et al. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononular phagocytes. Trends Immunol, 2002, 23:549~555
    39 Rauh MJ, Ho C, Pereira A, et al. Krystal: SHIP represses the generation of alternatively activated macrophages. Immunity, 2005, 23:361~374
    40 Myers MP, Stolarov JP, Eng C, et al. PTEN, the tumor suppressor from human chromosome 10q23, is a dual-specificity phosphatase. Proc Natl Acad Sci, 1997, 94: 9052~9057.
    41 Sattler M , Verma S , Byrne CH , et al. BCR/ ABL directly inhibits expression of SHIP , an SH-2containing polyinositol-5-phosphatase involved in the regulation of hematopoiesis. Mol Cell Biol ,1999 ,19 :7473~7480
    1 Smith M, Barnett M, Bassan R, et al. Adult acute myeloid leukemia. Crit Rev Oncol Hematol, 2004, 50:197~222
    2 Warner JK, Wang JC, Takenaka K, et al. Direct evidence for cooperating genetic events in the leukemic transformation of normal human hematopoietic cells. Leukemia, 2005, 19:1794~1805
    3 Sambani C, La Starza R, et al. Partial duplication of the MLL oncogene in patients with aggressive acute myeloid leukemia. Haematologica, 2004, 89:403~407
    4 Tonks A, Tonks AJ, Pearn L, et al. Expression of AML1-ETO in human myelomonocytic cells selectively inhibits granulocytic differentiation and promotes their self-renewal. Leukemia, 2004, 18:1238~1245
    5 Nanri T, Matsuno N, Kawakita T, et al. Mutations in the receptor tyrosine kinase pathway are associated with clinical outcome in patients with acute myeloblastic leukemia harboring t(8;21)(q22;q22). Leukemia, 2005, 19:1361~1366
    6 Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acuted myeloid leukemia: a meta-analysis. Leukemia, 2005, 19:1345~1349
    7 Cammenga J. Gatekeeper pathways and cellular background in the pathogenesis and therapy of AML. Leukemia, 2005, 19:1719~1728
    8 Hanada M, Feng J, Hemmings BA. Structure regulation and function of PKB/Akt-a major therapeutic target. Biochim Biophys Acta, 2004, 1697:3~16
    9 Osaki M, Oshimura M, Ito H. PI3K-Akt pathway: its functions andalterations in human cancer. Apoptosis, 2004, 9:667~676
    10 Kim D, Dan HC, Park S, et al. Akt/PKB signaling mechamisms in cancer and chemoresistance. Front Biosci, 2005, 10:975~987
    11 Kubota Y, Ohnishi H, Kitanaka A, et al. Constitutive activation of PI3K is involved in the spontaneous proliferation of primary acute myeloid leukemia cells: direct evidence of PI3K activation. Leukemia, 2004, 18:1438~1440
    12 Cho H, Mu J, Kim JK, et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2(PKBβ). Science, 2001, 292:1728~1731
    13 Chen WS Xu PZ, Gottlob K, et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes Dev, 2001, 15:2203~2208
    14 Ye K. PIKE/muclear PI 3-kinase signaling in preventing programmed cell death. J Cell Biochem, 2005, 96:463~472
    15 Du K, Tsichlis PN. Regulation of the Akt kinase by interacting proteins. Oncogene, 2005, 24:7401~7409
    16 Plo I, Bettaieb A, Payrastre B, et al. The phosphoinositide 3-kinase/Akt pathway is activated by daunorubicin in human acute myeloid leukemia cell lines. FEBS Lett, 1999, 452:150~154
    17 Sansal I, Stellers WR. The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol, 2004, 22:2954~2963
    18 Borgatti P, Martelli AM, Tabellini G, et al. Threonine 308 phosphorylated form of Akt translocates to the nucleus of PC12 cells under nerve growth factor sitmulation and associates with the nuclear matrix protein nucleolin. J Cell Physiol, 2003, 196:79~88
    19 Brandts CH, Sargin B, Rode M, et al. Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res, 2005, 65:9643~9650
    20 Min YH, Eom JI, Cheong JW, et al. Constitutive phosphorylation ofAkt/PKB protein in acute myeloid leukemia: its significance as a prognostic variable. Leukemia, 2003, 17:995~997
    21 Steelman LS, Pohnert SC, Shelton JG, et al. JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia, 2004, 18:189~218
    22 Schwable J, Choudhary C, Thiede C, et al. RGS2 is an important target gene of Flt3-ITD mutations in AML and functions in myeloid differentiation and leukemic transformation. Blood, 2005, 105:2107~2114
    23 Choudhary C, Schwable J, Brandts C, et al. AML-associated Flt3 kinase domain mutations show signal transduction differences compared with Flt3 ITD mutations. Blood, 2005, 106:265~273
    24 Frohing S, Scholl C, Gilliland DG, et al. Genetic of myeloid malignancies: pathogenetic and clinical implications. J Clin Oncol, 2005, 23:6285~6295
    25 Larizza L, Magnani I, Beghini A. The Kasumi-1 cell line: a t(8,21)-kit mutant model for acute myeloid leukemia. Leuk Lymphoma, 2005, 46:247~255
    26 Sawyer C, Sturge J, Bennett DC, et al. Regulation of breast cancer cell chemotaxis by the phosphoinositide 3-kinase p110δ. Cancer Res, 2003, 63:1667~1675
    27 Cheong JW, Eom JI, Maeng HY, et al. Phosphatase and tensin homologue phosphorylation in the C-terminal regulatory domain is frequently observed in acute myeloid leukaemia and associated with poor clinical outcome. Br J Heamatol, 2003, 122:454~456
    28 Grandage VL, Gale RE, Linch DC, et al. PI3-kinase/Akt is constitutively active in primary acute myeloid leukemia cells and regulates survival and chemoresistance via NF-kappaB, Mapkinase and p53 pathways. Leukemia, 2005, 19:586~594
    29 Liu TC, Lin PM, Chang JG, et al. Mutation analysis of PTEN/MMAC1 in acute myeloid leukemia. Am J Hematol, 2000, 63:170~175
    30 Aggerholm A, Gronbaek K, Guldberg P, et al. Mutational analysis of the tumor suppressor gene MMAC1/PTEN in malignant myeloid disorders.Eur J Haematol, 2000, 65:109~113
    31 Luo JM, Yoshida H, Komura S, et al. Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia. Leukemia, 2003, 17:1~8
    32 List AF, Glinsmann-Gibson B, Stadheim C, et al. Vascular endothelial growth factor receptor-1 and receptor-2 initiate a phosphatidylinositide
    3-kinase-dependent clonogenic response in acute myeloid leukemia cells. Exp Hematol, 2004, 32:526~535
    33 Thompson JE, Thompson CB. Putting the rap on Akt. J Clin Oncol, 2004, 22:4271~4226
    34 Rizzo MG, Giombini E, Diverio D, et al. Analysis of p73 expression pattern in acute myeloid leukemias: lack of DeltaN-p73 expression is a frequent feature of acute promyelocytic leukemia. Leukemia, 2004, 18:1804~1809
    35 Dai Y, Rahmani M, Pei XY, et al. Farnesyltransferase inhibitors interact synergistically with the Chk1 inhibitor UCN-01 to induce apoptosis in human leukemia cells through interruption of both Akt and MEK/ERK pathways and activation of SEK1/JNK. Blood, 2005, 105:1706~1716
    36 Wattel E, Preudhomme C, Hecquet B, et al. p53 mutations are associated with resistance to chemotherapy and short survival in hematologic malignancies. Blood, 1994, 84:3148~3157
    37 Brandts CH, Sargin B, Rode M, et al. Consittutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res, 2005, 65:9643~9650
    38 Cheong JW, Eom JI, Maeng HY, et al. Consittutive phosphorylation of FKHR transcription factor as a prognostic variable in acute myeloid leukemia. Leuk Res, 2003, 27:1159~1162
    39 Min YH, Cheong JW, Kim JY, et al. Cytoplasmic mislocalization of p27Kip1 protein is associated with comstitutive phoshorylation of Akt or protein kinase B and poor prognosis in acute myelogenous leukemia.Cancer Res, 2004, 64:5225~5231
    40 Min YH, Cheong JW, Kim JY, et al. Cytoplasmic mislocalization of p27Kip1 protein is associated with comstitutive phoshorylation of Akt or protein kinase B and poor prognosis in acute myelogenous leukemia. Cancer Res, 2004, 64:5225~5231
    41 Browne GJ, Proud CG. Regulation of peptide-chain elongation in mammalian cells. Eur J Biochem, 2002, 269:5360~5368
    42 Prunier C, Hocevar BA, Howe PH. Wnt signaling: physiology and pathology. Growth Factors, 2004, 22:141~150
    43 Tickenbrock L, Schwable J, Wiedehage M, et al. Flt3 tandem duplication mutations cooperate with Wnt signaling in leukemic signal transduction. Blood, 2005, 105:3699~3706
    44 O’Gorman DM, McKenna SL, McGabon AJ, et al. Sensitisation of HL—60 human leukemic cells to cytotoxic drug-induced apoptosis by inhibition of PI3-kinase survival signals. Leukemia, 2000, 14:602~611
    45 Ramos AM, Fernandez C, Amran D, et al. Pharmacologic inhibitors of PI3K/Akt potentiate the apoptotic action of the antileukemic drug arsenic trioxide via glutathione depletion and increased peroxide accumulation in myeloid leukemia cells. Blood, 2005, 105:4013~4020
    46 Kaufmann SH, Steensma DP. On the TRAIL of a new therpy for leukemia. Leukemia, 2005, 19:2195~2202
    47 Bortul R, Tazzari PL, Cappellini A, et al. Constitutively active Akt1 protects HL60 leukemia cells from TRAIL-induced apoptosis through a mechanism involving NF-κB activation and cFLIP(L) up-regulation. Leukemia, 2003, 17:379~389
    48 Carter BZ, Kornblau SM, Tsao T, et al. Caspase-independent cell death in AML: caspase inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis. Blood, 2003,102:4179~4186
    49 Raffaella A, Simona R, Rosanna P, et al. Rapamycin stimulates apoptosis of childhood acute lymphoblastic leukemia cells. Blood, 2005,106:1400~1406
    50 Semba S, Itoh N, Ito M, et al. The in vitro and in vivo effects of 2-(4-morpholinyl)-8-phenyl-chromone(LY294002), a specific inhibitor of phosphatidylinositol 3’-kinase, in human colon cancer cells. Clin Cancer Res, 2002, 8:1957~1963
    51 Schultz RM, Merriman RL, Andis SL, et al. In vitro and in vivo antitumor activity of the phosphatidylinositol-3-kinase inhibitor, wortmannin. Aniticcancer Res, 1995, 15:1135~1139
    52 Sujobert P, Bardet V, Cornillet-Lefebvre P, et al. Essential role for the p110δisoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. Blood, 2005, 106:1063~1066
    53 Sampath D, Cortes J, Estrov Z, et al. Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood, 2006, 107:2517~2524
    54 Yu W, Rao Q, Wang M, et al. The Hsp90 inhibitor 17-allylamide-17-demethoxygeldanamycin induces apoptosis and differentiation of Kasumi-1 harboring the Asn822Lys KIT mutation and down-regulated KIT protein level. Leuk Res, 2006, 30:575~582
    55 Dutcher JP. Mammalian targets of rapamycin inhibition. Clin Cancer Res, 2004, 10:63825~63875
    56 Recher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood, 2005, 105:2527~2534
    57 Yu C, Rahmani M, Dai Y, et al. The lethal effects of pharmacological cyclin-dependent kinase inhibitors in human leukemia cells proceed through a phosphatidylinositol 3-kinase/Akt-dependent process. Cancer Res, 2003, 63:1822~1833
    58 Xu RH, Pelicano H, Zhang H, et al. Synergistic effect of targeting mTOR by rapamycin and depleting ATP by inhibition of glycolysis in lymphoma and leukemia cells. Leukemia, 2005, 19:2153~2158
    59 Shi Y, Yan H, Frost P, et al. Mammalian target of rapamycin inhibitors activate the Akt kinase in multiple myeloma cells by up-regulating theinsulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther, 2005, 4:1533~1540
    60 Takada Y, Andreeff M, Aggarwal BB. Indole-3-carbinol suppresses NF-κB and IkappaBalpha kinase activation, causing inhibition of expression of NF-κB-regulated antiapoptotic in myeloid and leukemia cells. Blood, 2005, 106:641~649

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700