当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2005年第8期 > 正文
编号:11357805
Transcriptional Profiling of Human Hematopoiesis During In Vitro Lineage-Specific Differentiation
http://www.100md.com 《干细胞学杂志》
     a Department of Hematology and Oncology, University Hospital, Frankfurt, Germany;

    b Department of Hematology, Oncology and Transfusion Medicine, University Hospital Benjamin Franklin, Berlin, Germany;

    c Division of Hematology/Oncology, UCLA School of Medicine, Los Angeles, California, USA

    Key Words. Gene expression ? Hematopoietic development ? Hematopoietic stem cell ? Microarray ? Transcription

    Correspondence: Wolf-K. Hofmann, M.D., Department of Hematology, Oncology and Transfusion Medicine, University Hospital Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany. Telephone: 49-30-8445-3421; Fax: 49-30-8445-4468; e-mail: W.K.Hofmann@charite.de

    ABSTRACT

    Normal hematopoiesis is maintained by pluripotent, long-term repopulating stem cells that generate progenitors capable of differentiating into all three hematopoietic lineages. Only a few pluripotent hematopoietic stem cells possesses the ability to actively self-renew at any given time . Hematopoietic differentiation is strictly regulated by several exogenous factors . Lineage-specific growth factors, such as erythropoietin (EPO), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), and thrombopoietin (TPO), induce proliferation and differentiation into functionally active peripheral blood (PB) cells by activating multiple genes in an orchestrated way . Other growth factors, such as stem cell factor (SCF), the ligand of the Flt3/Flt2 receptor tyrosine kinase (FL), interleukin (IL)-1, IL-3, and IL-6, support growth and survival of primitive progenitor cells .

    Disruptions of these intricate sequences of transcriptional activation and suppression of multiple genes can cause hematological diseases, such as leukemias, myelodysplastic syndromes (MDS), or myeloproliferative syndromes (MPS). Elucidating the pattern and sequence of gene expression during normal hematopoietic cell development may help to unravel the disease-specific mechanisms in hematopoietic malignancies.

    The first report describing sequential analysis of gene expression during hematopoietic differentiation in the mouse system has recently been published . Transcriptional profiling of murine multipotent hematopoietic progenitor cells (factor-dependent cell-Paterson -mix cell line) during multilineage differentiation revealed several known and as-yet unknown genes that are differentially expressed in erythroid and neutrophil differentiation as well as genes involved in self-renewal and differentiation .

    We have established an in vitro model that enables the detailed analysis of human hematopoietic differentiation originating from unstimulated, steady-state bone marrow CD34+ cells. Using this system, we have analyzed gene expression patterns of the three hematopoietic lineages at defined time points throughout lineage-specific differentiation. Because we used primary human CD34+ cells, the experiments were done in triplicates for each of the time points and each of the conditions to minimize individual changes in expression levels of approximately 22,500 genes that were analyzed.

    Our data provide several new insights into understanding differentiation and proliferation of human stem and progenitor cells. In addition, the data should be particularly suitable for comparative analysis of gene expression in hematopoietic malignancies, to characterize pathomechanisms of diseases, and, finally, to find genetically defined therapeutic targets.

    MATERIALS AND METHODS

    In Vitro Differentiation of CD34+ Cells

    CD34+ cells of healthy individuals were plated on different culture conditions for lineage-specific differentiation to generate erythropoietic, granulopoietic, and megakaryopoietic cells (Fig. 1). Lineage-specific cells were characterized by morphology and by immunophenotype as described. Cells from each of the donors were cultured separately, and RNA was prepared from each of the conditions for microarray hybridization.

    Figure 1. Morphology of CD34+ cells and cells generated by in vitro differentiation. Typical morphology is detectable in erythropoiesis (day 4, proerythroblasts; day 7, erythroblasts; day 11, normoblasts), granulopoiesis (incremental organization of nucleus resulting in polymorph nuclear cells), and megakaryopoiesis (increasing polyploidy). Abbreviations: EPO, erythropoietin; FL, ligand of the Flt3/Flt2 receptor tyrosine kinase; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte macrophage colony-stimulating factor; IL-3, interleukin-3; SCF, stem cell factor; TPO, thrombopoietin.

    On day 4 of culture, cell numbers increased markedly under conditions for erythropoiesis and granulopoiesis (>threefold). Because of the endomitotic development of megakaryocytic cells, the proliferation of these cells was slow compared with erythropoietic and granulopoietic cells. Table 1 shows a summary of cell numbers and the purities of the lineage-specific cells after positive selection used for gene expression analysis.

    Table 1. Summary of cell numbers, purities of the in vitro–differentiated cells, and the "present" calls (P) of the hybridized microarrays

    Oligonucleotide Microarrays

    Gene expression in cells sampled at days 0, 4, 7, and 11 of hematopoietic differentiation was analyzed by oligonucleotide microarrays (HG-U133A).

    As an indicator of high-quality hybridization results, we determined the value of expressed genes (P) of every array analyzed by MAS 5.0. The mean value of present calls (P-calls) was 10,074 out of 22,500 (44%; Table 1) and therefore above 25% in all of our experiments, as required by MIAME (minimum information about a microarray experiment) for a high-quality microarray experiment .

    Horizontal Analysis of Gene Expression During Lineage-Specific Differentiation

    Characterization of Genes Known to Be Lineage-Specific ? As a confirmation of the lineage-specific in vitro differentiation, we identified continuously upregulated genes that are already known to be associated with specific programs of hematopoietic differentiation. These genes were not expressed in CD34+ cells. Figure 2 shows the expression of specific genes for the erythrocytic (e.g., ANK, D9S57E, CD35), granulocytic (e.g., IRC1, FCGR2B), and megakaryocytic (e.g., SERPINE1, THBD, PDGF1, and CD42C) lineage in the course of differentiation.

    Figure 2. Confirmation of the in vitro differentiation of CD34+ cells by microarray analysis. During lineage-specific differentiation, the expression of well-known marker genes for each hematopoietic lineage analyzed increased continuously. Expression of erythrocytic (ANK, erythrocytic Ankyrin 1; Z39IG, immunoglobulin superfamily protein; CD35, complement component receptor 1, including Knops blood group system; D9S57E, tropomodulin), granulocytic (IRC1, leukocyte membrane antigen; FCGR2B, Fc fragment of immunoglobulin G, low-affinity IIb, receptor for CD32) and mega-karyocytic marker genes (SERPINE1, serine proteinase inhibitor, clade E ; PHS1, prostaglandin-endoperoxide synthase 1; THBD, thrombomodulin; MSR1, macrophage scavenger receptor 1; PDGF1, platelet-derived growth factor alpha; FCGR2A, receptor for CD32; CD42C, glycoprotein Ib ) is shown.

    Genes Strongly Associated with Specific Hematopoietic Lineages ? To investigate genes that are strongly associated with lineage-specific differentiation, we generated lists containing genes with continuously increasing or decreasing expression in each hematopoietic lineage. Continuously regulated genes of the hematopoietic lineages were hierarchically clustered (Figs. 3A–3C).

    Figure 3. Cluster analysis of genes associated with (A) erythropoiesis, (B) granulopoiesis, and (C) megakaryopoiesis. One cluster of continuously upregulated and another cluster of continuously down-regulated genes for each of the three hematopoietic lineages are shown. Color code: blue, low expression; red, high expression. The intensity of the color reflects the reliability of the expression data.

    Only a few genes increased or decreased continuously during erythropoietic (21 up, 58 down), granulopoietic (21 up, 30 down), and megakaryocytic differentiation (91 up, 37 down). Nine genes are regulated in both erythropoietic and granulopoietic cells (e.g., CD86 antigen, immunoglobulin superfamily protein, solute carrier family 7), three genes are regulated in erythropoiesis and megakaryopoiesis (FLJ20748, CD86, EST: 217678_AT), and three genes are regulated during granulopoietic and megakaryopoietic differentiation (potassium large-conductance calcium-activated channel, CD86, MGC5528). Only one gene is continuously regulated in all three differentiation programs (CD86), suggesting that it may be involved in commitment or differentiation of the hematopoietic stem cell. Table 2 lists genes with continuously increasing expression in erythropoiesis. Table 3 shows continuously decreasing genes in erythropoiesis. The data set for all lineages (continuously upregulated and downregulated genes) is given in the supplemental material (supplemental online Tables 1–6). In addition, the complete expression data of all samples will be available for downloading at http://knm1.ibe.med.uni-muenchen.de/kn_home/WKH/index.htm.

    Table 2. Genes continuously regulated during erythropoietic differentiation with continuously increasing expression

    Table 3. Genes continuously regulated during erythropoietic differentiation with continuously decreasing expression

    Table 4. Genes differentially expressed in erythropoiesis

    Table 5. Genes differentially expressed in granulopoiesis

    Table 6. Genes differentially expressed in megakaryopoiesis

    Vertical Analysis of Differentially Expressed Genes

    A vertical analysis was performed to identify genes that are differentially expressed in a specific hematopoietic lineage at a defined time point. The analysis of genes expressed in cells on specific lineage-directed differentiation conditions that show a greater than threefold change at any defined time point compared with cells in the remaining conditions (e.g., erythropoiesis versus granulopoiesis and megakaryopoiesis) resulted in lists of differentially expressed genes for each of the hematopoietic lineages. Tables 4–6 show highly differentially expressed genes for each lineage (A, erythropoiesis; B, granulopoiesis; C, megakaryopoiesis). The specific time point of expression is indicated. We could not detect genes common to all three hematopoietic lineages, supporting the lineage-specificity of the identified genes.

    Class Membership Prediction

    The maximal number of genes to predict the class membership was found to be 53 using five neighbors. We defined this set of genes to be able to classify a particular sample according to its lineage. Figure 4 represents the results of hierarchical clustering with Spearman’s confidence correlation of 25 samples of in vitro–differentiated CD34+ cells. Three subclusters indicate the affiliation to one of the three hematological lineages. One misclassification (sample E_d04_2) occurred. In Table 7, the predictive genes are listed according to the appearance in the cluster.

    Figure 4. Identification of genes expressed in differentiating CD34+ bone marrow cells, which are significantly correlated with each of the three analyzed hematopoietic lineages. Results represent the analysis by hierarchical clustering with Spearman’s confidence correlation of 25 samples of in vitro–differentiated CD34+ bone marrow cells. Fifty-three genes were selected to predict the class membership of each of the samples. The vertical list contains each of the samples. The horizontal list displays the 53 genes. Three clusters corresponding to the three hematological lineages were found, as indicated by the black bars. One misclassification occurred (sample ID: E_d04_2). For granulopoietic and megakaryopoietic differentiation, three and two samples are missing in the analysis, respectively. Color code: blue, low expression; red, high expression. The intensity of the color reflects the reliability of the expression data.

    Table 7. Genes that are predictive for the lineages of human in vitro–differentiating hematopoietic cells (see Fig. 4)

    Confirmation of Expression of Continuously Regulated Genes During In Vitro Hematopoietic Differentiation by Real-Time PCR

    To confirm the expression data from the oligonucleotide micro-array studies, we analyzed the expression of a selection of six significantly regulated genes (two in each lineage) in 25 samples by real-time PCR. The gene for PTGS2 was analyzed in erythropoietic and granulopoietic samples. All experiments were done in triplicate. The variance between the triplicates was less than 5%. The results were normalized to the expression of GPI in each of the samples. Figure 5 summarizes the expression data measured by real-time PCR for the selected genes. We could confirm our microarray results for both the continuously upregulated and downregulated genes not only for the single time points but also for the course of gene expression during hematopoietic differentiation.

    Figure 5. Validation of gene expression pattern of continuously regulated genes during lineage-specific differentiation by real-time polymerase chain reaction (PCR). The expression of a selection of six genes in 25 samples was analyzed by real-time PCR. Two genes per hematopoietic lineage were analyzed in samples of the appropriate lineage (the gene for PTGS2 was analyzed in erythropoietic and granulopoietic samples). We found with one exception (IRC1 should be upregulated in G) that for both the continuously upregulated and downregulated genes, the real-time PCR results are similar to the results detected by microarray analysis. X-axis, days; Y-axis, GPI-normalized expression levels as detected by real-time PCR.

    Gene Ontology Analysis

    Categorization of differentially expressed genes in all three hematopoietic lineages according to the molecular function yielded seven functional groups (genes with binding, catalytic, signal transducer, transcription regulator, structural molecule, enzyme regulator, and transporter activity; Fig. 6).

    Figure 6. Classification of differentially expressed genes of normal hematopoiesis according to gene ontology. Genes were annotated according to FatiGO Data Mining with Gene Ontology. Annotations are available for a subset of genes (~68%) represented on the HG-U133A microarray. The functional classification of a specific gene may be redundant, resulting from the assignment of one gene to more than one category.

    Genes with binding (E, 36%; versus G, 37%; versus M, 38%), catalytic (E, 26%; versus G, 19%; versus M, 19%), and signal transducer activity (E, 22%; versus G, 29%; versus M, 23%) were comparable between the three hematopoietic lineages. The percentage of genes performing transcription regulator activity or structure molecule activity (E, 2%; versus G, 7%; versus M, 3%; E, 6%; versus G, 2%; versus M, 1%) was slightly elevated in the granulopoietic or erythropoietic lineage, whereas genes functioning as enzyme regulators (E, 4%; versus G, 3%; versus M, 6%) and transporters (E, 4%; versus G, 3%; versus M, 10%) were overrepresented in the megakaryopoietic lineage. Further, it should be noted that the functional classification of a specific gene may be redundant, resulting from the assignment of one gene to more than one category.

    DISCUSSION

    This work was supported by the Deutsche Forschungsgemeinschaft (HO 2207/3-1), the BMBF Competence Network Leukemias, and the German Genome Research Network.

    REFERENCES

    Chan JY, Watt SM. Adhesion receptors on haematopoietic progenitor cells. Br J Haematol 2001;112:541–557.

    Lotem J, Sachs L. Cytokine control of developmental programs in normal hematopoiesis and leukemia. Oncogene 2002;21:3284–3294.

    Zhu J, Emerson SG. Hematopoietic cytokines, transcription factors and lineage commitment. Oncogene 2002;21:3295–3313.

    Ogawa M. Differentiation and proliferation of hematopoietic stem cells. Blood 1993;81:2844–2853.

    Broudy VC. Stem cell factor and hematopoiesis. Blood 1997;90:1345–1364.

    Haylock DN, Horsfall MJ, Dowse TL et al. Increased recruitment of hematopoietic progenitor cells underlies the ex vivo expansion potential of FLT3 ligand. Blood 1997;90:2260–2272.

    Bruno L, Hoffmann R, McBlane F et al. Molecular signatures of self-renewal, differentiation, and lineage choice in multipotential hemopoietic progenitor cells in vitro. Mol Cell Biol 2004;24:741–756.

    Hofmann WK, 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.

    Al Shahrour F, Diaz-Uriarte R, Dopazo J. FatiGO: a web tool for finding significant associations of Gene Ontology terms with groups of genes. Bioinformatics 2004;20:578–580.

    Peters UR, Tschan MP, Kreuzer KA et al. Distinct expression patterns of the p53-homologue p73 in malignant and normal hematopoiesis assessed by a novel real-time reverse transcription-polymerase chain reaction assay and protein analysis. Cancer Res 1999;59:4233–4236.

    Bustin SA. Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays. J Mol Endocrinol 2000;25:169–193.

    Brazma A, Hingamp P, Quackenbush J et al. Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet 2001;29:365–371.

    Guzman ML, Upchurch D, Grimes B et al. Expression of tumor-suppressor genes interferon regulatory factor 1 and death-associated protein kinase in primitive acute myelogenous leukemia cells. Blood 2001;97:2177–2179.

    Hofmann WK, de Vos S, Elashoff D et al. Relation between resistance of Philadelphia-chromosome-positive acute lymphoblastic leukaemia to the tyrosine kinase inhibitor STI571 and gene-expression profiles: a gene-expression study. Lancet 2002;359:481–486.

    Zeng W, Chen G, Kajigaya S et al. Gene expression profiling in CD34 cells to identify differences between aplastic anemia patients and healthy volunteers. Blood 2004;103:325–332.

    Bond HM, Bonelli P, Mesuraca M et al. Identification by differential display of transcripts regulated during hematopoietic differentiation. STEM CELLS 1998;16:136–143.

    Zhou G, Chen J, Lee S et al. The pattern of gene expression in human CD34+ stem/progenitor cells. Proc Natl Acad Sci U S A 2001;98:13966–13971.

    Steidl U, Kronenwett R, Rohr UP et al. Gene expression profiling identifies significant differences between the molecular phenotypes of bone marrow-derived and circulating human CD34+ hematopoietic stem cells. Blood 2002;99:2037–2044.

    Shojaei F, Gallacher L, Bhatia M. Differential gene expression of human stem progenitor cells derived from early stages of in utero human hematopoiesis. Blood 2004;103:2530–2540.

    Terskikh AV, Miyamoto T, Chang C et al. Gene expression analysis of purified hematopoietic stem cells and committed progenitors. Blood 2003;102:94–101.

    Park IK, He Y, Lin F et al. Differential gene expression profiling of adult murine hematopoietic stem cells. Blood 2002;99:488–498.

    Gubin AN, Njoroge JM, Bouffard GG et al. Gene expression in proliferating human erythroid cells. Genomics 1999;59:168–177.

    Bonafoux B, Lejeune M, Piquemal D et al. Analysis of remnant reticulocyte mRNA reveals new genes and antisense transcripts expressed in the human erythroid lineage. Haematologica 2004;89:1434–1438.

    Scicchitano MS, McFarland DC, Tierney LA et al. In vitro expansion of human cord blood CD36+ erythroid progenitors: temporal changes in gene and protein expression. Exp Hematol 2003;31:760–769.

    Kolbus A, Blazquez-Domingo M, Carotta S et al. Cooperative signaling between cytokine receptors and the glucocorticoid receptor in the expansion of erythroid progenitors: molecular analysis by expression profiling. Blood 2003;102:3136–3146.

    Kim JA, Jung YJ, Seoh JY et al. Gene expression profile of megakaryocytes from human cord blood CD34+ cells ex vivo expanded by thrombopoietin. STEM CELLS 2002;20:402–416.

    Gnatenko DV, Dunn JJ, McCorkle SR et al. Transcript profiling of human platelets using microarray and serial analysis of gene expression. Blood 2003;101:2285–2293.

    Tenedini E, Fagioli ME, Vianelli N et al. Gene expression profiling of normal and malignant CD34-derived megakaryocytic cells. Blood 2004;104:3126–3135.

    Itoh K, Okubo K, Utiyama H et al. Expression profile of active genes in granulocytes. Blood 1998;92:1432–1441.

    Bertrand G, Coste J, Segarra C et al. Use of serial analysis of gene expression (SAGE) technology reveals new granulocytic markers. J Immunol Methods 2004;292:43–58.

    Newburger PE, Subrahmanyam YV, Weissman SM. Global analysis of neutrophil gene expression. Curr Opin Hematol 2000;7:16–20.

    Kobayashi SD, Voyich JM, Buhl CL et al. Global changes in gene expression by human polymorphonuclear leukocytes during receptor-mediated phagocytosis: cell fate is regulated at the level of gene expression. Proc Natl Acad Sci U S A 2002;99:6901–6906.

    Fessler MB, Malcolm KC, Duncan MW et al. A genomic and proteomic analysis of activation of the human neutrophil by lipopolysaccharide and its mediation by p38 mitogen-activated protein kinase. J Biol Chem 2002;277:31291–31302.

    Tsukahara Y, Lian Z, Zhang X et al. Gene expression in human neutrophils during activation and priming by bacterial lipopolysaccharide. J Cell Biochem 2003;89:848–861.

    Song JH, Kim JM, Kim SH et al. Comparison of the gene expression profiles of monocytic versus granulocytic lineages of HL-60 leukemia cell differentiation by DNA microarray analysis. Life Sci 2003;73:1705–1719.

    Rubinfeld B, Munemitsu S, Clark R et al. Molecular cloning of a GTPase activating protein specific for the Krev-1 protein p21rap1. Cell 1991;65:1033–1042.

    Shang X, Zhou YT, Low BC. Concerted regulation of cell dynamics by BNIP-2 and Cdc42GAP homology/Sec14p-like, proline-rich, and GTPase-activating protein domains of a novel Rho GTPase-activating protein, BPGAP1. J Biol Chem 2003;278:45903–45914.

    Teye K, Tsuneoka M, Arima N et al. Increased expression of a Myc target gene Mina53 in human colon cancer. Am J Pathol 2004;164:205–216.

    Lutz W, Leon J, Eilers M. Contributions of Myc to tumorigenesis. Biochim Biophys Acta 2002;1602:61–71.

    Krummrei U, Baulieu EE, Chambraud B. The FKBP-associated protein FAP48 is an antiproliferative molecule and a player in T cell activation that increases IL2 synthesis. Proc Natl Acad Sci U S A 2003;100:2444–2449.

    Communi D, Gonzalez NS, Detheux M et al. Identification of a novel human ADP receptor coupled to G(i). J Biol Chem 2001;276:41479–41485.

    Van Leeuwen FN, Olivo C, Grivell S et al. Rac activation by lysophosphatidic acid LPA1 receptors through the guanine nucleotide exchange factor Tiam1. J Biol Chem 2003;278:400–406.

    Shim MH, Hoover A, Blake N et al. Gene expression profile of primary human CD34+CD38(lo) cells differentiating along the megakaryocyte lineage. Exp Hematol 2004;32:638–648.

    Smas CM, Sul HS. Pref-1, a protein containing EGF-like repeats, inhibits adipocyte differentiation. Cell 1993;73:725–734.

    Laborda J, Sausville EA, Hoffman T et al. dlk, a putative mammalian homeotic gene differentially expressed in small cell lung carcinoma and neuroendocrine tumor cell line. J Biol Chem 1993;268:3817–3820.

    Abdallah BM, Jensen CH, Gutierrez G et al. Regulation of human skeletal stem cells differentiation by Dlk1/Pref-1. J Bone Miner Res 2004;19:841–852.

    Van Limpt VA, Chan AJ, Van Sluis PG et al. High delta-like 1 expression in a subset of neuroblastoma cell lines corresponds to a differentiated chromaffin cell type. Int J Cancer 2003;105:61–69.

    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.

    Okada Y, Nagai R, Sato T et al. Homeodomain proteins MEIS1 and PBXs regulate the lineage-specific transcription of the platelet factor 4 gene. Blood 2003;101:4748–4756.(Martina Komora, Saskia Gü)