骨髓增生异常综合征骨髓造血细胞分化异常及其相关机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的
     对骨髓增生异常综合征(Myelodysplastic Syndrome, MDS)患者骨髓造血细胞的分化异常进行检测,并对其分化异常的部分相关机制进行研究;同时进一步寻找MDS恶性克隆细胞,并探讨其过度增殖、分化异常及凋亡逃逸等恶性特征。
     方法
     研究对象为天津医科大学总医院自2008年7月至2010年10月新诊断的MDS患者62例、MDS转化的急性髓系白血病(MDS-AML)8例、急性髓系白血病(AML)7例、病例对照组16例及正常对照32名。第一部分检测MDS患者骨髓造血细胞分化异常情况,采用流式细胞术(FCM)检测MDS患者骨髓粒系、单核系及红系分化抗原;干细胞分群;单个核细胞细胞周期分布,并分析分化异常情况与染色体恶性克隆负荷的相关性。第二部分研究导致MDS患者骨髓造血细胞分化异常的相关机制,采用FCM检测MDS患者干/祖细胞表面干细胞因子受体(SCF-R)、红细胞生成素受体(EpoR)、粒细胞集落刺激因子受体(G-CSFR)及血小板生成素受体(TpoR)的表达情况,并分析其临床意义;干/祖细胞转录因子GATA-1、GATA-2的表达情况;RT-PCR的方法检测GATA-1基因在MDS患者骨髓中的表达情况。第三部分寻找MDS恶性克隆细胞,检测MDS患者骨髓干细胞中白细胞介素-3受体α(IL-3Ra)表达情况,并探讨IL-3Ra阳性细胞的异常增殖及分化特征。
     结果
     第一部分粒系细胞分化抗原表达的模式和比例:选择CD13/CD11b CD13/CD16及CD11b/CD16组合来分析粒细胞分化抗原表达模式,对照组和病例对照组骨髓粒系细胞组合模式分别为“对钩”、“镰刀,或“反7”状,MDS患者骨髓粒系细胞发育分化中的抗原表达失去正常序贯表达模式,呈现抗原的连续性表达中断等不同程度的改变。高危组CDllb/CD11b+比值(0.32±0.29)明显高于低危组(0.16±0.23)、病例对照组(0.05±0.03)及正常对照组(0.07±0.05,P<0.01);高危组CD16/CD16+比值(1.32±0.77)明显高于病例对照组(0.33±0.32)和正常对照组(0.39±0.31,P<0.05);高危组骨髓粒细胞CD13的平均荧光强度(MFI)明显高于正常对照组(P<0.01),侧向角散射光信号(SSC)的MFI明显低于病例对照组和正常对照组(P<0.01)。高危组CD11b-HLA-DR+(4.08%±2.97%)、CD11b-HLA-DR'(14.90%±15.44%)、CD16-HLA-DR- (38.98%±21.59%)、CD11b+CD16-(32.57%±16.52%)及CD13+CD16-(43.97%±20.31%)细胞占粒细胞比例明显高于低危组、病例对照组及对照组(P<0.05),其他组间比较差异无统计学意义。单核系细胞分化抗原表达的模式和比例:选择CD13/CD16和CD11b/HLA-DR组合来分析单核细胞分化抗原表达模式,MDS患者骨髓单核系细胞发育分化中的抗原表达模式相对于对照组的序贯组合模式出现不同程度的改变。高危组CD11b-HLA-DR+(12.38%±8.97%), CD11lb+HLA-DR-(38.33%±18.43%)细胞占单核系细胞比例明显高于病例对照组和正常对照组(P<0.05)。高危组单核细胞CD14(56.74%±17.73%)表达率明显低于正常对照组,CD64的表达率各组之间比较,差异无统计学意义,CD56(32.45%±20.07%)、CD34(13.54%±13.73%)及CD7(15.31%±12.76%)表达率明显高于病例对照组及正常对照组。红系细胞分化抗原表达的模式和比例:应用CD71和血型糖蛋白A(glycophorin A,GlyA)的组合来分析红系细胞的分化,对照组和病例对照组两种抗原的组合模式均为双阳性表达,部分MDS患者可见CD71和GlyA表达不同步现象。低危组和高危组CD71+、GlyA+细胞占CD45-细胞的比例均显著低于病例对照组和正常对照组(P<0.05),CD71+GlyA+双阳性细胞分别占CD45-、CD71+细胞的比例均显著低于病例对照组和正常对照组(P<0.05), CD71+GlyA+双阳性细胞占GlyA+细胞的比显著低于正常对照组(P<0.05)。MDS患者粒系、单核系及红系抗原表达的比例和模式的异常数目与IPSS(国际预后积分系统)积分(r=0.662, P=0.000)、WPSS (WHO分型预后积分系统)积分(r=0.602,P=0.000)及恶性克隆负荷(r=0.477,P=0.001)均呈正相关。高危和MDS-AML组骨髓CD34+细胞比例(2.29%±2.17%、18.69±17.47%)明显高于对照组(0.36%±0.49%,P<0.05)。低危、高危及MDS-AML组CD34+CD38+细胞相对比例(86.09%±7.79%、81.68%±11.82%、82.88%±2.60%)显著低于对照组(92.21%±3.85%,P<0.05),而CD34+CD38-细胞比例(13.91%±7.79%、18.32%±11.82%、17.13%±2.60%)显著高于对照组(7.79%±3.85%,P<0.05)。MDS组CD34+CD38-细胞比例与IPSS (r=0.493, P=0.001)、WPSS积分(r=0.586,P=0.000)均呈正相关。CD34+CD38-细胞比例≤12.0%的MDS患者治疗有效率高于CD34+CD38-细胞比例>12.0%的患者,差异无统计学意义。低危、高危及MDS-AML组单个核细胞(BMMNC)中Go/G1期细胞比例(94.52%±4.32%,96.07%±3.88%,94.65%±4.55%)明显高于对照组(88.94%±7.30%,P<0.01),而3组患者S期(4.63%±3.34%,3.45%±3.80%,5.12%±4.55%)和G2/M期(0.84%±1.52%,0.48%±0.74%,0.22%±0.34%)细胞比例明显低于对照组(9.06%±6.50%,2.00%±2.93%,P<0.05),3组S+G2/M期细胞比例明显高于对照组(P<0.01)。
     第二部分MDS患者骨髓造血细胞造血因子受体及部分转录因子的检测,对照组骨髓CD34+CD38+细胞亚群EpoR表达率(18.68%±18.34%)显著低于CD34+CD38细胞亚群(63.61%±19.98%,P<0.01),两亚群之间SCF-R.G-CSFR及TpoR表达率差异无统计学意义。在CD34+CD38+细胞亚群中,3组间SCF-R和TpoR表达率差异无统计学意义,而低危组和高危组EpoR的表达率(8.30%±6.55%、7.82%±7.98%)明显低于对照组(18.68%±18.34%,P<0.05),G-CSFR的表达率(29.78%±19.14%、28.66%±21.14%)明显低于对照组(44.37%±23.43%,P<0.05);在CD34+CD38-细胞亚群中,3组间SCF-R和G-CSFR表达率差异无统计学意义,低危组和高危组EpoR的表达率(42.19%±21.87%、25.67%±15.64%)明显低于对照组(63.61%±19.98%,P<0.01),TpoR的表达率(5.42%±4.72%、4.05%±3.95%)明显低于对照组(10.13%±8.31%,P<0.05)。MDS患者骨髓CD34+CD38+和CD34+CD38-细胞亚群表面受体表达率低的患者其外周血血红蛋白水平、中性粒细胞及血小板计数减低的发生率明显高于受体表达率不低的患者(均P<0.05)。在CD34+CD38+细胞亚群中,低危组和高危组GATA-1的表达率高于对照组,差异无统计学意义,高危组GATA-2的表达率(12.47%±13.00%)明显高于对照组(5.64%±7.32%,P<0.05),余组间比较差异无统计学意义;在CD34+CD38-细胞亚群中,高危组GATA-1的表达率(28.16%±13.71%)明显高于对照组(12.76%±14.02%,P<0.05),低危组和高危组GATA-2的表达率(17.12%±10.61%、29.38%±7.71%)明显高于对照组(10.07%±9.26%,P<0.05)。GATA-1基因在高危组骨髓单个核细胞(BMMNC)中的表达量为(0.504±0.156),显著高于对照组(0.323±0.086,P<0.01),余组间比较差异无统计学意义。
     第三部分MDS骨髓干细胞中白细胞介素-3受体α(IL-3Rα)表达情况及IL-3Rα阳性细胞增殖及分化等特征的检测。低危组、高危组及AML组CD34+CD38细胞中CD123+细胞比例(42.48%±25.88%、51.62%±29.24%、56.19%±32.20%)明显高于对照组(9.06%±10.04%, P<0.01)。CD 123+CD34+CD38-/CD34+CD38表达率与恶性克隆负荷呈显著正相关(r=0.419,P=0.003),与MDS骨髓粒系、单核系及红系细胞分化模式和比例的异常数目呈显著正相关(r=0.462,P=0.001)。低危组、高危组及AML组CD34+CD38 CD 123+细胞中GATA.1.GATA.2及CD71表达率均明显高于其在CD34+CD38+CD123+细胞中的表达率(P<0.01);3组CD34+CD38-CD123+细胞中EpoR表达率高于其在CD34+CD38-CD123细胞中的表达率,但差异无统计学意义;3组CD34+CD38-CD123+细胞中CDllb、CD114及Annexin V表达率均明显低于其在CD34+CD38-CD123-细胞中表达率(P<0.05)。
     结论
     (1)MDS患者骨髓粒系、单核系及红系细胞抗原不同步表达,表现为阶段特异性和系别特异性的异常,诸如成熟阶段细胞异常表达幼稚细胞抗原,SSC值降低即胞质低颗粒化\脱颗粒现象、连续性表达中断、抗原部分表达降低\缺失,髓系细胞异常表达淋巴系抗原等,并且由低危向高危进展的过程中存在分化异常的积累并与IPSS、WPSS积分及恶性克隆负荷成显著正相关。这提示分化抗原检测可能有助于MDS患者的早期诊断和预后判断。
     (2)MDS患者原代骨髓CD34+细胞亚群分化异常,CD34+CD38-/CD34+比例增高,并和IPSS、WPSS积分呈正相关性;骨髓单个核细胞存在G1期阻滞现象,提示MDS患者造血细胞增殖分化异常,CD34+细胞亚群和细胞周期测定有助于MDS患者的辅助诊断以及疗效和预后判断。
     (3)MDS患者原代骨髓CD34+细胞亚群膜表面部分造血细胞因子受体表达减低,CD34+细胞亚群转录因子GATA-1和GATA-2表达增高,这可能与MDS患者分化异常导致无效造血有关,且有望用于辅助诊断MDS。
     (4)MDS患者原代骨髓中CD34+CD38-CD123+细胞增高,并与恶性克隆负荷及骨髓造血.细胞分化异常项数目呈显著正相关,这些细胞表现为过度增殖、分化异常、部分造血因子受体的表达缺陷及凋亡逃逸等恶性特征,提示该团细胞可能是MDS恶性克隆细胞及最终转化为AML的恶性根源。
Objective
     This study was to detect the differentiation abnormalities of bone marrow hematopoietic cells in myelodysplastic syndrome (MDS), its related mechanisms, MDS malignant clone cells and their malignant characteristics of excessive proliferation, abnormal differentiation and apoptosis escape.
     Methods
     Sixty-two cases of MDS patients,8 acute myeloid leukemias preceded by MDS (MDS-AML),7 acute myeloid leukemias and 16 case-controls, as well as 32 normal controls were enrolled in this study. PartⅠThe differentiation antigens on the membrane of bone marrow granulocytes, monocytes and erythroblasts, the subset of stem cell and bone marrow cell cycle were measured by flow cytometry. PartⅡThe expressions of stem cell factor receptor(SCF-R), erythropoietin receptor(EpoR), granulocyte colony-stimulating factor receptor (G-CSFR) and thrombopoietin receptor (TpoR) on hematopoietic stem/progenior cells and the expressions of GATA-1 and GATA-2 in nuclear of stem/progenitor cells were measured by flow cytometry. The expression of GATA-1 mRNA in the bone marrow cells of cases with MDS and normal controls were measured by RT-PCR. PartⅢThe expression of interleukin-3 receptorα(CD123) on the bone marrow stem cells of cases with MDS, AML and normal controls were measured by flow cytometry. Further to investigate the characteristics of proliferation and differentiation of CD123+ cells.
     Results
     PartⅠThe granulocytic differentiation was analyzed with the combinations of CD13/CD11b, CD13/CD16 and CD11b/CD16. The "right hook", "sickle" and "retroflex 7" shape expressions were found in normal and case controls while there were various changes in MDS groups. The ratios of CD11b-/CD11b+(0.32±0.29) and CD16-/CD16+(1.32±0.77) were significantly higher in high-risk MDS group than those in case-control group(0.05±0.03 and 0.33±0.32 respectively) and control group (0.07±0.05 and 0.39±0.31 respectively) (P<0.05). The MFI (mean fluorescence index) of SSC (side scatter) in the granulocyte gate of high-risk MDS group was significantly lower while MFI of CD13 was significantly higher than the case-control and control groups. The mean percentages of CD11b-HLA-DR+(4.08%±2.97%), CD11b-HLA-DR-(14.90%±15.44%), CD16-HLA-DR-(38.98%±21.59%), CDllb+CD16-(32.57%±16.52%) and CD13+CD16-(43.97%±20.31%) granulocytes of high-risk MDS group were significantly higher than those of low-risk, case-control and control groups (P<0.05). The monocytic differentiation was analyzed with the combinations of CD13/CD16 and CDllb/HLA-DR. There were various changes in MDS group comparison with case-control and control groups. The mean percentages of CD11b"HLA-DR+(12.38%±8.97%), CD11b+HLA-DR-(38.33%±18.43%) monocytes of high-risk MDS group were significantly higher than those of case-control and control groups (P<0.05). The mean percentage of CD14+ (56.74%±17.73%) on monocytes of high-risk MDS group was significantly lower than those of control group(P<0.05). The mean percentages of CD56+(32.45%±20.07%), CD34+(13.54%±13.73%) and CD7+(15.31%±12.76%) of high-risk MDS group were significantly higher than those of case-control and control groups (P<0.05). The erythroid cell lineage differentiation was analyzed with CD71/glycophorin A combination. Double positive expression was found in all case controls and controls, but asynchronous expression of CD71/glycophorin A was found in some MDS cases. The mean percentages of CD71+and GlyA+cells in CD45" cell population were significantly lower in low-risk and high-risk MDS groups. The mean percentages of CD71+GlyA+double positive cells in CD45", CD71+and GlyA+cell population were significantly lower in low-risk and high-risk MDS groups. The abnormal numbers of the antigen expression of MDS cases per case correlated directly with their IPSS (international prognostic scoring system) (r=0.662, P=0.000), WPSS (WHO adapted prognostic scoring system) (r=0.602, P=0.000) scores and malignant clone burden(r=0.477, P=0.001). The mean percentages of CD34+cells in bone marrow karyocyte of high risk(2.29%±2.17%) and MDS-AML groups(18.69%±17.47%) were significantly higher than that of control group (0.36%±0.49%, P<0.05). The mean percentages of CD34+CD38+cells were significantly lower in low risk, high risk and MDS-AML groups(86.09%±7.79%, 81.68%±11.82%and 82.88%±2.60% respectively) than that in control group (92.21%±3.85%, P<0.05), thus the percentages of CD34+CD38" cells were significantly higher in either MDS (low-risk and high-risk) or MDS-AML groups (13.91%±7.79%,18.32%±11.82%or 17.13%±2.60% respectively) than that in control group (7.79%±3.85%, P<0.05). The percentages of CD34+CD38'cells of MDS cases correlated directly with their IPSS (r=0.493, P=0.001) and WPSS (γ=0.586,P=0.000) scores. MDS patients with low percentages of CD34+CD38-(≤12.0%) cells presented higher therapeutic efficacy than those with high percentages of CD34+CD38'(> 12.0%) cells (P> 0.05), but not reveal significant differences. The percentages of bone marrow mononuclea cells(BMMNCs) in Go/Gi phase of in low-risk, high-risk and MDS-AML groups (94.52%±4.32%, 96.07%±3.88%and 94.65%±4.55%respectively) were significantly higher than that of control group(88.94%±7.30%, P<0.01), thus the percentages of BMMNCs in S and G2/M phase were significantly lower in either MDS (low-risk and high-risk) or MDS-AML groups than that in control group (P<0.05).
     Part II In control group, the mean percentage of antigen expression of EpoR was significantly lower in CD34+CD38+cells (18.68%±18.34%) than that in CD34+CD38- cells (63.61%±19.98%, P<0.01).The expressions of SCF-R,G-CSFR and TpoR were not significantly different between the two cell populations. The expression of EpoR on CD34+CD38+cells of low-risk and high-risk MDS groups(8.30%±6.55%,7.82%±7.98%) were significantly lower than that of control group(18.68%±18.34%, P<0.05). The expression of G-CSFR on CD34+CD38+cells of low-risk and high-risk MDS groups(29.78%±19.14%,28.66%±21.14%)were significantly lower than that of control group(44.37%±23.43%, P<0.05). The amount of EpoR on CD34+CD38- cells of low-risk and high-risk MDS groups(42.19%±21.87%,25.67%±15.64%) were significantly lower than that of control group(63.61%±19.98%, P<0.01), The expression of TpoR on CD34+CD38' cells of low-risk and high-risk MDS groups(5.42%±4.72%,4.05%±3.95%) were significantly lower than that of control group(10.13%±8.31%, P<0.05). The incidence of cytopenia with low expression rates of hemopoietic cytokine receptors on CD34+ cells were higher than that of MDS with high expression rates of hemopoietic cytokine receptors. The expression of GATA-1 on CD34+CD38+cells of MDS groups were higher than that of control group, but not reveal significant differences.The expression of GATA-2 on CD34+CD38+cells of high-risk MDS group (12.47%±13.00%) was significantly higher than that of control group(5.64%±7.32%, P<0.05). The expression of GATA-1 on CD34+CD38- cells of high-risk MDS group (28.16%±13.71%) was significantly higher than that of control group(12.76%±14.02%, P<0.05). The expression of GATA-2 on CD34+CD38" cells of low-risk and high-risk MDS groups(17.12%±10.61%,29.38%±7.71%) were significantly higher than that of control group(10.07%±9.26%, P<0.05). The expression of GATA-1 mRNA in BMMNC of MDS patients (0.504±0.156) was significantly higher than that of normal controls (0.323±0.086, P<0.01).
     PartⅢThe ratios of CD34+CD38"CD123+/CD34+CD38- in the bone marrow cells of low-risk, high-risk MDS and AML groups (42.48%±25.88%,51.62%±29.24%, 56.19%±32.20%) were significantly higher than that of normal control (9.06%±10.04%, P<0.01). The ratio of CD34+CD38"CD123+/CD34+CD38'in MDS groups was significantly positively correlated with the malignant clone burden(r=0.419,P=0.003) and the abnormal numbers of the differentiation antigen expression (r=0.462, P=0.001). The expressions of GATA-1, GATA-2 and CD71 in CD34+CD38-CD123+cells were significantly higher than those in CD34+CD38" CD123-cells of MDS and AML groups(P<0.01). The expression of EpoR on CD34+CD38-CD123+cells was higher than that on CD34+CD38-CD123- cells, but not reveal significant differences. The expressions of CD11b, CD114 and Annexin V on CD34+CD38-CD123+cells were significantly lower than those on CD34+CD38-CD123-cells of MDS and AML groups(P<0.05).
     Conclusions
     (1) There are abnormal expressions of differentiation antigens on bone marrow myeloied cells of MDS patients. And the severity is correlated with the IPSS, WPSS scores and malignant clone burden. The abnormal differentiation of myeloid cells is probably involved in the pathogenesis of MDS. So the examination of these antigenic expressions with flow cytometry might be helpful for diagnosis and prognosis of MDS.
     (2) There was abnormalitie of differentiation of CD34+bone marrow cells and high proportion of G0/G1 cells which indicated a G1 phase arrest in MDS. That indicated there were abnormal characteristics of differentiation and proliferation in hematopoietic cells of MDS. So the examination of CD34+bone marrow cells and cell cycle might be helpful for MDS diagnosis and assessment of prognosis and therapeutic effect.
     (3) There were abnormalities of some membrane hemopoietic cytokine receptors and transcription factors of CD34+bone marrow cells in MDS, which were associated with MDS cytopenia and might be useful for MDS diagnosis.
     (4) The percentage of CD34+CD38-CD123+cells in the bone marrow of MDS patients was increased. These cells manifest the malignant characteristics of excessive proliferation, abnormal differentiation and apoptosis escape. CD34+CD38" CD123+cells were probably the malignant clone cells in MDS.
引文
[1]Valent P, Horny HP, Bennett JM, et al. Definitions and standards in the diagnosis and treatment of the myelodysplastic syndromes:Consensus statements and report from a working conference. Leuk Res,2007,31(6):727-736.
    [2]Afable MG 2nd, Wlodarski M, Makishima H, et al. SNP array-based karyotyping: differences and similarities between aplastic anemia and hypocellular myelodysplastic syndromes. Blood,2011 Apr 28 [Epub ahead of print].
    [3]Theilgaard-Monch K, Boultwood J, Ferrari S, et al. Gene expression profiling in MDS and AML:potential and future avenues. Leukemia.2011 Mar 29. [Epub ahead of print]
    [4]Greenberg PL, Attar E, Bennett JM, et al. Myelodysplastic syndromes. J Natl Compr Cane Netw.2011,9(1):30-56.
    [5]Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the myelodysplastic syndromes. Br J Haematol,1982,51:189-199.
    [6]Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood,1997,89:2079-2088.
    [7]Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood,2002,100:2292-2302.
    [8]曹燕然,邵宗鸿,施均,等.骨髓增生异常综合征多指标综合诊断的前瞻性研究.中国实用内科杂志,2006,8:1145-1147.
    [9]Ogata K, Kishikawa Y, Satoh C, et al. Diagnostic application of flow cytometric characteristics of CD34+cells in low-grade myelodysplastic syndromes. Blood, 2006,108:1037-1044.
    [10]Cherian S, Moore J, Bantly A, et al. Peripheral blood MDS score:a new flow cytometric tool for the diagnosis of myelodysplastic syndromes. Cytometry B Clin Cytom,2005,64:9-17.
    [11]Loken MR, van de Loosdrecht A, Ogata K, et al. Flow cytometry in myelodysplastic syndromes:report from a working conference. Leukemia Research,2008,32:5-17.
    [12]Lorand-Metze I, Ribeiro E, Lima CS, et al. Detection of hematopoietic maturation abnormalities by flow cytometry in myelodysplastic syndromes and its utility for the differential diagnosis with non-clonal disorders. Leukemia Research,2007,31:147-155.
    [13]Stachurski D, Smith BR, Pozdnyakova O, et al. Flow cytometric analysis of myelomonocytic cells by a pattern recognition approach is sensitive and specific in diagnosing myelodysplastic syndrome and related marrow diseases:emphasis on a global evaluation and recognition of diagnostic pitfalls. Leukemia Research, 2008,32:215-224.
    [14]Hussein K, Theophile K, Busche G, et al. Aberrant microRNA expression pattern in myelodysplastic bone marrow cells. Leuk Res.2010,34(9):1169-74.
    [15]Yalcintepe L, Frankel AE, Hogge DE, et al. Expression of interleukin-3 receptor subunits on defined subpopulations of acute myeloid leukemia blasts predicts the cytotoxicity of diphtheria toxin interleukin-3 fusion protein against malignant progenitors that engraft in immunodeficient mice. Blood,2006, 108(10):3530-3537.
    [16]Djokic M, Bjorklund E, Blennow E, et al. Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica, 2009,94:1016-1019.
    [17]张之南,沈悌.血液病诊断及疗效标准.3版.北京:科学技术出版社,2007.
    [18]van de Loosdrecht AA, Westers TM, Westra AH, et al. Identification of distinct prognostic subgroups in low- and intermediate-1 risk myelodysplastic syndromes by flow cytometry. Blood,2008,111:1067-1077.
    [19]van de Loosdrecht AA, Alhan C, Bene MC, et al. Standardization of flow cytometry in myelodysplastic syndromes:report from the first European LeukemiaNet working conference on flow cytometry in myelodysplastic syndromes. Hematologica,2009,94:1124-1134.
    [20]Satoh C, Dan K, Yamashita T, et al. Flow cytometric parameters with little interexaminer variability for diagnosing low-grade myelodysplastic syndromes. Leuk Res,2008,32 (5):699-707.
    [21]Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood,2009,114:937-51.
    [22]Baur AS, Meuge-MorawC, Schmidt PM, et al. CD34/QBEND10 immunostaining in bone marrowbiopsies:an additional parameter for the diagnosis and classification of myelodysplastic syndromes. Eur J Haematol,2000,64:71-9.
    [23]Ogata K, Della Porta MG, Malcovati L, Picone C, Yokose N, Matsuda A,et al. Diagnostic utility of flow cytometry in low-grade myelodysplastic syndromes:a prospective validation study. Haematologica,2009,94:1066-74.
    [24]Della Porta MG, Malcovati L, Invernizzi R, et al. Flow cytometry evaluation of erythroid dysplasia in patients with myelodysplastic syndrome. Leukemia,2006, 20:549-555.
    [25]McClintock-Treep SA, Briggs RC, Shults KE, et al. Quantitative assessment of myeloid nuclear differentiation antigen distinguishes myelodysplastic syndrome from normal bone marrow. Am J Clin Pathol,2011,135(3):380-5.
    [26]Florian S, Sonneck K, Hauswirth AW, et al. Detection of molecular targets on the surface of CD34+/CD38-stem cells in various myeloid malignancies. Leuk Lymphoma,2006,47:207-222.
    [27]Monreal MB, Pardo ML, Pavlovsky MA, et al. Increased immature hematopoietic progenitor cells CD34+/CD38dim in myelodysplasia. Cytometry B Clin Cytom,2006,70:63-70.
    [28]Goardon N, Nikolousis E, Sternberg A, et al. Reduced CD38 expression on CD34+cells as a diagnostic test in myelodysplastic syndromes. Haematologica, 2009,94:1160-1163.
    [29]施均,邵宗鸿,刘鸿,等.骨髓增生异常综合征细胞周期调控基因表达谱的研究.中华血液学杂志,2005,26:10-14.
    [30]Thomas X, Le QH, Danaila C, et al. Bone marrow biopsy in adult acute lymphoblastic leukemia:morphological characteristics and contribution to the study of prognostic factors. Leuk Res,2002,26:909-918.
    [31]Malcovati L, Germing U, Kuendgen A, et al. Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. J Clin Oncol,2007,25:3503-3510.
    [32]Navarro I, Ruiz MA, Cabello A, et al. Classification and scoring systems in myelodysplastic syndromes:a retrospective analysis of 311 patients. Leuk Res, 2006,30:971-977.
    [33]Cherian S, Moore J, Bantly A, et al. Peripheral blood MDS score:a new flow cytometric tool for the diagnosis of myelodysplastic syndromes. Cytometry B Clin Cytom,2005,64(1):9-17.
    [34]Germing U, Strupp C, Kuendger A, et al. Prospective validation of the WHO proposals for the classification of myelodysplastic syndromes. Haematologica, 2006,91:1596-1604.
    [35]Wells DA, Benesch M, Loken MR, et al. Myeloid and monocytic dyspoiesis as determined by flow cytometric scoring in myelodysplastic syndrome correlates with the IPSS and with outcome after hematopoietic stem cell transplantation. Blood,2003,102:394-403.
    [36]Wagner W, Ansorge A, Wirkner U, et al. Molecular evidence for stem cell function of the slow-dividing fraction among human hematopoietic progenitor cells by genome-wide analysis. Blood,2004,104:675-686.
    [37]Florian S, Sonneck K, Hauswirth AW, et al. Detection of molecular targets on the surface of CD34+/CD38-stem cells in various myeloid malignancies. Leuk Lymphoma,2006,47:207-222.
    [38]Kent. D, Copley. M, Benz. C, et al. Regulation of hematopoietic stem cells by the steel factor/KIT signaling pathway. Clin. Cancer Res,2008,14:1926-1930.
    [39]Scholl. C, Gilliland. DG, Frohling. S. Deregulation of signaling pathways in acute myeloid leukemia. Semin. Oncol,2008,35:336-345.
    [40]Shao Z, Zhang H, Chen G, et al. Expression and function of c-kit receptor in bone marrow mononuclear cells of patients with myelodysplastic syndromes. Chin MedJ,2001,114:481-485.
    [41]Inthal A, Krapf G, Beck D, et al. Role of the erythropoietin receptor in ETV6/RUNX1-positive acute lymphoblastic leukemia. Clin Cancer Res,2008, 14:7196-7204.
    [42]T6vari J, Pirker R, Timar J, et al. Erythropoietin in cancer:an update. Curr Mol Med,2008,8:481-491.
    [43]Graf M, Hecht K, Reif S, et al. Expression and prognostic value of hemopoieti cytokine receptors in acute myeloid leukemia (AML):implications for future therapeutical strategies. Eur J Haematol,2004,72:89-106.
    [44]Takeshita A, Shinjo K, Naito K, et al. Erythropioetin receptor in myelodysplastic syndrome and leukemia. Leuk Lymphoma,2002,43:261-264.
    [45]Graf M, Hecht K, Reif S, et al. Expression and prognostic value of hemopoieti cytokine receptors in acute myeloid leukemia (AML):implications for future therapeutical strategies. Eur J Haematol,2004,72:89-106.
    [46]Yue LZ, Fu R, Wang HQ, et al. Expression of CD 123 and CD 114 on the bone marrow cells of patients with myelodysplastic syndrome. Chin Med J (Engl), 2010,123:2034-7.
    [47]Sultana TA, Harada H, Ito K, et al. Expression and functional analysis of G-CSFR on CD34 cells in patients with MDS and MDS-AML. Br J Haematol, 2003,121:63-75.
    [48]Kimura A, Sultana TA. Granulocyte colony-stimulating factor receptors on CD34++cells in patients with myelodysplastic syndrome (MDS) and MDS-acute myeloid leukemia. Leuk Lymphoma,2004,45:1995-2000.
    [49]Ninos JM, Jefferies LC, Cogle CR, et al. The thrombopoietin receptor, c-Mpl, is a selective surface marker for human hematopoietic stem cells. J Transl Med,2006, 16:1-18.
    [50]Edvardsson L, Dykes J, Olofsson T. Isolation and characterization of human myeloid progenitor populations-TpoR as discriminator between common myeloid and megakaryocyte/erylhroid progenitors. Exp Hematol,2006, 34:599-609.
    [51]Wetzler M, Baer MR, Bernstein SH, et al. Expression of c-mpl mRNA, the receptor for thrombopoietin, in acute myeloid leukemia blasts identifies a group of patients with poor response to intensive chemotherapy. J Clin Oncol,1997,15: 2262-2268.
    [52]Grass. JA, ME. Boyer, S. Pal, et al. GATA-1-dependent transcriptional repression of GATA-2 via disruption of positive autoregulation and domain-wide chromatin remodeling. Proc. Natl. Acad. Sci,2003,100:8811-8816.
    [53]Yuan-Yeh Kuo, Zee-Fen Chang, GATA-1 and Gfi-1B interplay to regulate Bcl-xL transcription. Molecular and cellular biology,2007,7:4261-4272.
    [54]Trainor CD, Mas C, Archambault P, et al. GATA-1 associates with and inhibits p53. Blood,2009,114(1):165-73.
    [55]Rylski. M, JJ. Welch, YY. Chen, et al. GATA-1-mediated proliferation arrest during erythroid maturation. Mol. Cell. Biol,2003,23:5031-5042.
    [56]Maratheftis CI, Bolaraki PE, Voulgarelis M. GATA-1 transcription factor is up-regulated in bone marrow hematopoietic progenitor CD34(+) and erythroid CD71(+) cells in myelodysplastic syndromes. Am J Hematol,2007, 82(10):887-92.
    [57]Duff C, Smith-Miles K, Lopes L, et al. Mathematical modelling of stem cell differentiation:the PU.1-GATA-1 interaction. J Math Biol,2011 Apr 2. [Epub ahead of print].
    [58]Bouilloux F, Juban G, Cohet N, et al. EKLF restricts megakaryocytic differentiation at the benefit of erythrocytic differentiation. Blood,2008, 112:576-584.
    [59]Hong W, Nakazawa M, Chen YY, et al. FOG-1 recruits the NuRD repressor complex to mediate transcriptional repression by GATA-1. EMBO J,2005,24: 2367-2378.
    [60]Cantor A. B, Iwasaki H, Arinobu Y, et al. Antagonism of FOG-1 and GATA factors in fate choice for the mast cell lineage. J. Exp,2008,205:611-624.
    [61]Shimizu R, Kuroha T, Ohneda O, et al. Leukemogenesis caused by incapacitated GATA-1 function. Mol Cell Biol,2004,24(24):10814-25.
    [62]邵宗鸿.骨髓增生异常综合征恶性克隆的早期识别与根治.中华内科杂志,2008,47:441-443.
    [63]Testa U, Riccioni R, Diverio D, et al. Interleukin-3 receptor in acute leukemia. Leukemia,2004,18:219-226.
    [64]Jordan CT, Upchurch D, Szilvassy SJ, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia,2000,14:1777-1784.
    [65]Djokic M, Bjorklund E, Blennow E, et al. Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica, 2009,94:1016-1019.
    [66]Munoz L, Nomdedeu JF, Lopez O, et al. Interleukin-3 receptor a chain (CD 123) is widely expressed in hematologic malignancies. Haematologica,2001, 86:1261-1269.
    [67]Riccioni R, Rossini A, Calabro L, et al. Immunophenotypic features of acute myeloid leukemias overexpressing the interleukin 3 receptor alpha chain. Leuk Lymphoma,2004,45:1511-1517.
    [68]Hassanein NM, Alcancia F, Perkinson KR, et al. Distinct expression patterns of CD 123 and CD34 on normal bone marrow B-cell precursors ("hematogones") and B lymphoblastic leukemia blasts. Am J Clin Pathol,2009,132:573-580.
    [69]Yalcintepe L, Frankel AE, Hogge DE Expression of interleukin-3 receptor subunits on defined subpopulations of acute myeloid leukemia blasts predicts the cytotoxicity of diphtheria toxin interleukin-3 fusion protein against malignant progenitors that engraft in immunodeficient mice.Blood, 2006;108(10):3530-3537.
    [70]Djokic M, Bjorklund E, Blennow E, et al. Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica, 2009,94:1016-1019.
    [71]Feuring-Buske M, Frankel AE, Alexander RL, et al. A diphtheria toxin-interleukin 3 fusion protein is cytotoxic to primitive acute myeloid leukemia progenitors but spares normal progenitors. Cancer Res,2002, 62:1730-1736.
    [72]Du X, Ho M, Pastan I. New immunotoxins targeting CD123, a stem cell antigen on acute myeloid leukemia cells. J Immunother,2007,30:607-613.
    [73]Miyazato A, Ueno S, Ohmine K, et al. Identification of myelodysplastic syndrome-specific genes by DNA microarray analysis with purified hematopoietic stem cell fraction. Blood,2001,98:422-427.
    [74]Crispino JD. GATA1 in normal and malignant hematopoiesis. Semin Cell Dev Biol,2005,16:137-147.
    [75]Arinobu Y, Mizuno S, Chong Y, et al. Reciprocal Activation of GATA-1 and PU.1 Marks Initial Specification of Hematopoietic Stem Cells into Myeloerythroid and Myelolymphoid Lineages. Cell Stem Cell,2007,12:416-427.
    [76]Cantor AB, Iwasaki H, Arinobu Y, et al. Antagonism of FOG-1 and GATA factors in fate choice for the mast cell lineage. J Exp Med,2008,205:611-624.
    [77]Ayala RM, Martinez-Lopez J, Albizua E, et al. Clinical significance of Gata-1, Gata-2, EKLF, and c-MPL expression in acute myeloid leukemia.Am J Hematol, 2009,84:79-86.
    [78]Sada E, Abe Y, Ohba R, et al.Vitamin K2 modulates differentiation and apoptosis of both myeloid and erythroid lineages. Eur J Haematol,2010,85:538-548.
    [79]Testa U, Riccioni R, Militi S, et al. Elevated expression of IL-3R alpha in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. Blood,2002,100:2980-2988.
    [80]De Waele M, Renmans W, Vander Gucht K, et al. Growth factor receptor profile of CD34+cells in AML and B-lineage ALL and in their normal bone marrow counterparts. Eur J Haematol,2001,66:178-187.
    [81]Van Rhenen A, Moshaver B, Kelder A, et al. Aberrant marker expression patterns on the CD34+CD38" stem cell compartment in acute myeloid leukemia allows to distinguish the malignant from the normal stem cell compartment both at diagnosis and in remission. Leukemia,2007,21:1700-1707.
    [82]Park S, Grabar S, Kelaidi C, et al. Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF:the GFM experience. Blood,2008,111:574-582.
    [83].Tadersten M, Malcovati L, Dybedal I, et al. Erythropoietin and granulocyte-colony stimulating factor treatment associated with improved survival in myelodysplastic syndrome. J Clin Oncol,2008,26:3607-3613.
    [84]Jadersten M, Malcovati L, Dybedal L, et al. Treatment with EPO and GCSF improves survival in MDS patients with low transfusion need. Blood [ASH meeting abstracts],2006,108(11):158a.
    [85]Park S, Grabar S, Kelaidi C, et al. Has treatment with EPO+/-G-CSF an impact on progression to AML and survival on low/int-1-risk MDS? A comparison between French-EPO patients and the IMRAW database. Leuk Res,2007, 31(Suppl 1):S113.
    [86]Golshayan AR, Jin T, Maciejewski J, et al. Efficacy of growth factors compared to other therapies for low-risk myelodysplastic syndromes. Br J Haematol,2007, 137:125-132.
    [87]Terpos E, Mougiou A, Kouraklis A, et al. Prolonged administration of erythropoietin increases erythroid response rate in myelodysplastic syndromes:a phase II trial in 281 patients. Br J Haematol,2002,118:174-180.
    [88]Benites BD, Traina F, Duarte Ada S, et al. Increased expression of APAF-1 in low-risk myelodysplastic syndrome:a possible role in the pathophysiology of myelodysplasia. Eur J Haematol,2010,84:525-530.
    [89]Morita Y, Kanamaru A, Miyazaki Y, et al. Comparative analysis of remission induction therapy for high-risk MDS and AML progressed from MDS in the MDS 200 study of Japan Adult Leukemia Study Group. Int J Hematol,2010, 91(l):97-103.
    [90]Sperr WR, Hauswirth AW, Florian S, et al. Human leukaemic stem cells:a novel target of therapy. Clin Cancer Res. Eur J Clin Invest,2004,34:31-40.
    1. van de Loosdrecht AA, Westers TM, Westra AH, et al. Identification of distinct prognostic subgroups in low- and intermediate-1 risk myelodysplastic syndromes by flow cytometry. Blood,2008,111:1067-1077.
    2. Ogata K, Kishikawa Y, Satoh C, et al. Diagnostic application of flow cytometric characteristics of CD34+ cells in low-grade myelodysplastic syndromes. Blood, 2006,108:1037-1044.
    3. Goardon N, Nikolousis E, Sternberg A, et al. Reduced CD38 expression on CD34+ cells as a diagnostic test in myelodysplastic syndromes. Haematologica, 2009,94:1160-1163.
    4. Djokic M, Bjorklund E, Blennow E, et al. Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica, 2009,94(7):1016-9.
    5. Florian S, Sonneck K, Hauswirth AW,et al. Detection of molecular targets on the surface of CD34+/CD38-stem cells in various myeloid malignancies. Leuk Lymphoma,2006,47(2):207-22.
    6. Loken MR, van de Loosdrecht A, Ogata K, et al. Flow cytometry in myelodysplastic syndromes:report from a working conference. Leukemia Research,2008,32:5-17.
    7. Lorand-Metze I, Ribeiro E, Lima CS, et al. Detection of hematopoietic maturation abnormalities by flow cytometry in myelodysplastic syndromes and its utility for the differential diagnosis with non-clonal disorders. Leukemia Research,2007, 31:147-155.
    8. Stachurski D, Smith BR, Pozdnyakova O, et al. Flow cytometric analysis of myelomonocytic cells by a pattern recognition approach is sensitive and specific in diagnosing myelodysplastic syndrome and related marrow diseases:emphasis on a global evaluation and recognition of diagnostic pitfalls. Leukemia Research, 2008,32:215-224.
    9. van de Loosdrecht AA, Alhan C, Bene MC, et al. Standardization of flow cytometry in myelodysplastic syndromes:report from the first European LeukemiaNet working conference on flow cytometry in myelodysplastic syndromes. Hematologica,2009,94:1124-1134.
    10. Greenberg PL, Attar E, Bennett JM, et al. Myelodysplastic syndromes. J Natl Compr CancNetw.2011,9(1):30-56.
    11. Della Porta MG, Malcovati L, Invernizzi R, et al. Flow cytometry evaluation of erythroid dysplasia in patients with myelodysplastic syndrome. Leukemia,2006, 20:549-555.
    12. Satoh C, Dan K, Yamashita T, et al. Flow cytometric parameters with little interexaminer variability for diagnosing low-grade myelodysplastic syndromes. Leuk Res,2008,32 (5):699-707.
    13. Maftoun-Banankhah S, Maleki A, Karandikar NJ, et al, Multi-parameter flow cytometric analysis reveals low percentage of bone marrow hematogones in myelodysp lastic syndromes. Am J C lin Pathol,2008,129:300-308.
    14.施均,邵宗鸿,刘鸿,等.骨髓增生异常综合征细胞周期调控基因表达谱的研究.中华血液学杂志,2005,26(1):10-14.
    15. Shimazaki K, Ohshima K, Suzumiya J, et al. Evaluation of apoptosis as a prognostic factor in myelodysplastic syndromes. Br J Haematol 2000, 110:584-590.
    16. Parker JE, Mufti GJ, Rasool F, et al. The role of apoptosis, proliferation, and the Bcl-2 related proteins in the myelodysplastic syndromes and acute myeloid leukemia secondary to MDS. Blood,2000,96:3932-3938.
    17. Pecci A, Travaglino E, Kiersy C, et al. Apoptosis in relation to CD34 antigen expression in normal and myelodysplastic bone marrow. Acta Haematol,2003, 109:29-34.
    18. Kent. D, Copley. M, Benz. C, et al. Regulation of hematopoietic stem cells by the steel factor/KIT signaling pathway. Clin Cancer Res,2008,14:1926-1930.
    19. Scholl. C, Gilliland. DG, Frohling. S, et al. Deregulation of signaling pathways in acute myeloid leukemia. Semin. Oncol,2008,35:336-345.
    20. Cortelezzi A, Cattaneo C, Cristiani S, et al. Low plasma stem cell factor levels correlate with in vitro leukemic growth in myelodysplastic syndromes. Leuk Res, 1999,23:271-275.
    21.Inthal A, Krapf G, Beck D, et al. Role of the erythropoietin receptor in ETV6/RUNX1-positive acute lymphoblastic leukemia. Clin Cancer Res.2008,14: 7196-7204.
    22. Takeshita A, Shinjo K, Naito K, et al. Erythropioetin receptor in myelodysplastic syndrome and leukemia. Leuk Lymphoma,2002,43:261-264.
    23. Graf M, Hecht K, Reif S, et al. Expression and prognostic value of hemopoieti cytokine receptors in acute myeloid leukemia (AML):implications for future therapeutical strategies. Eur J Haematol,2004,72:89-106.
    24. Ninos JM, Jefferies LC, Cogle CR, et al. The thrombopoietin receptor, c-Mpl, is a selective surface marker for human hematopoietic stem cells. J Transl Med,2006, 16:1-18.
    25. Edvardsson L, Dykes J, Olofsson T, et al. Isolation and characterization of human myeloid progenitor populations-TpoR as discriminator between common myeloid and megakaryocyte/erythroid progenitors. Exp Hematol,2006,34:599-609.
    26. Wetzler M, Baer MR, Bernstein SH, et al. Expression of c-mpl mRNA, the receptor for thrombopoietin, in acute myeloid leukemia blasts identifies a group of patients with poor response to intensive chemotherapy. J Clin Oncol,1997,15: 2262-2268.
    27. Grass. JA, ME. Boyer, S. Pal, et al. GATA-1-dependent transcriptional repression of GATA-2 via disruption of positive autoregulation and domain-wide chromatin remodeling.Proc. Natl. Acad. Sci. USA 2003,100:8811-8816.
    28. Yuan-Yeh Kuo, Zee-Fen Chang, GATA-1 and Gfi-1B interplay to regulate Bcl-xL transcription. Molecular and cellular biology,2007,7:4261-4272.
    29. Trainor CD, Mas C, Archambault P, et al. GATA-1 associates with and inhibits p53. Blood,2009,114(1):165-73.
    30. Rylski. M, JJ. Welch, YY. Chen,et al. GATA-1-mediated proliferation arrest during erythroid maturation. Mol. Cell. Biol,2003,23:5031-5042.
    31. Maratheftis CI, Bolaraki PE, Voulgarelis M. GATA-1 transcription factor is up-regulated in bone marrow hematopoietic progenitor CD34(+) and erythroid CD71(+) cells in myelodysplastic syndromes. Am J Hematol.2007, 82(10):887-92.
    32. Lowry. JA, Mackay. JP. GATA-1:one protein, many partners. Int. J. Biochem. Cell Biol.2006,38:6-11.
    33. Bouilloux F, Juban G, Cohet N, et al. EKLF restricts megakaryocytic differentiation at the benefit of erythrocytic differentiation. Blood,2008, 112:576-584.
    34. Hong W, Nakazawa M, Chen YY, et al. FOG-1 recruits the NuRD repressor complex to mediate transcriptional repression by GATA-1. EMBO J,2005,24: 2367-2378.
    35. Cantor A. B, Iwasaki H, Arinobu Y, et al. Antagonism of FOG-1 and GATA factors in fate choice for the mast cell lineage. J. Exp,2008,205:611-624.
    36. Nerlov CE, Querfurth H, Kulessa, et al. GATA-1 interacts with the myeloid PU.1 transcription factor and represses PU.1-dependent transcription. Blood,2000, 95:2543-2551.
    37. Zhang P, X Zhang, A Iwama, et al. PU.1 inhibits GATA-1 function and erythroid differentiation by blocking GATA-1 DNA binding. Blood,2000,96:2641-2648.
    38. 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(2):189-218.
    39. Richmond T. D, Chohan M, and Barber DL. Turning cells red:signal transduction mediated by erythropoietin. Trends Cell Biol,2005,15:146-155.
    40. Kotecha N, Flores NJ, Irish JM, et al, Single-cell profiling identifies aberrant STAT5 activation in myeloid malignancies with specific clinical and biologic correlates. Cancer Cell,2008,14(4):335-43.
    41. Wei Zhao, Claire Kitidis, Mark D Fleming, et al. Erythropoietin stimulates phosphorylation and activation of GATA-1 via the PI3-kinase-AKT signaling pathway, blood,2005,6:2516.
    42. Scheele JS, Ripple D, Lubbert M, et al. The role of ras and other low molecular weight guanine nucleotide (GTP)-grinding proteins during hematopoeitic cell differentiation. Cell Mol Life Sci,2000,57:1950-1963.
    43. Kalina U, Hofmann WK, Koschmieder S, et al. Alteration of c-mpl-mediated signal transduction in CD34(+) cells from patients with myelodysplastic syndromes. Exp Hematol,2000,28:1158-1163.
    44. Hopfer OJ, Komor M, Koehler IS, et a 1. GATA and BCL-xl downregulation in erythropoiesis during in vitro lineage specific differentiation of MDS hematopoietic progenitor cells is not induced by activated notch pathway. ASH Annual Meeting Abstracts,2007,110:4118.
    45. Jing Ai, Lawrence J. Druhan, Melissa G. Hunter, et al. LRG-accelerated differentiation defines unique G-CSFR signaling pathways downstream of PU.1 and C/EBPε that modulate neutrophil activation. Journal of Leukocyte Biology, 2008,83:1277-1285.
    46. Miyazato A, Ueno S, Ohmine K, et al. Identification of myelodysplastic syndrome-specific genes by DNA microarray analysis with purified hematopoietic stem cell fraction. Blood,2001,98(2):422-427.
    47. Schmidt JV, Matteson PG, Jones BK, et al. The Dlkl and Gtl2 genes arelinked and reciprocally imprinted. Genes Dev,2000,14:1997-2002.
    48. Raghunandan R, Ruiz-Hidalgo M, Jia Y, et al. Dlkl influences differentiation and function of B lymphocytes. Stem Cells Dev,2008,17(3):495-507.
    49. Qian CF, Yan W, Zhang JX, et al. Notchl induces enhanced expression of Delta-like-1 in the U251MG glioma cell line. Int J Mol Med,2009,24(4):445-51.
    50. Qi X, Chen Z, Liu D, et al. Expression of Dlkl gene in myelodysplastic syndrome determined by microarray and its effects on leukemia cells. Int J Mol Med,2008, 22(1):61-8.
    51. Sakajiri S, O'kelly J, Yin D, et al. Dlkl in normal and abnormal hematopoiesis. Leukemia,2005,19(8):1404-10.
    52. Moore KA, Pytowski B, Witte L, et al. Hematopoietic activity of a stromal cell transmembrane protein containing epidermal growth factor-like repeat motifs. Proc Natl Acad Sci U S A,1997,94:4011-4016.
    53. Hofmann W-K, de Vos S, Komor M, et al. Characterization of gene expression of CD34+cells from normal and myelodysplastic bone marrow. Blood,2002, 100:3553-3560.
    54. Ma L, Delforge M, Verhoef GEG, et al. The immunophenotype and function of dendritic cells in myelodysplastic syndromes. Blood,2002,100:166.
    55. Bellamy WT, Richter L, Sirjani D, et al. Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes. Blood,2001, 97:1427-1434.

© 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