Skip to main content
Molecular and Cellular Biology logoLink to Molecular and Cellular Biology
. 2012 Jul;32(13):2628–2644. doi: 10.1128/MCB.00104-12

Krüppel-Like Factor 1 (KLF1), KLF2, and Myc Control a Regulatory Network Essential for Embryonic Erythropoiesis

Christopher J Pang a, Wafaa Lemsaddek f, Yousef N Alhashem a, Cornelius Bondzi a,h, Latasha C Redmond a, Nicolas Ah-Son f, Catherine I Dumur b, Kellie J Archer c, Jack L Haar d, Joyce A Lloyd a,e,, Marie Trudel f,g,
PMCID: PMC3434496  PMID: 22566683

Abstract

The Krüppel-like factor 1 (KLF1) and KLF2 positively regulate embryonic β-globin expression and have additional overlapping roles in embryonic (primitive) erythropoiesis. KLF1−/− KLF2−/− double knockout mice are anemic at embryonic day 10.5 (E10.5) and die by E11.5, in contrast to single knockouts. To investigate the combined roles of KLF1 and KLF2 in primitive erythropoiesis, expression profiling of E9.5 erythroid cells was performed. A limited number of genes had a significantly decreasing trend of expression in wild-type, KLF1−/−, and KLF1−/− KLF2−/− mice. Among these, the gene for Myc (c-Myc) emerged as a central node in the most significant gene network. The expression of the Myc gene is synergistically regulated by KLF1 and KLF2, and both factors bind the Myc promoters. To characterize the role of Myc in primitive erythropoiesis, ablation was performed specifically in mouse embryonic proerythroblast cells. After E9.5, these embryos exhibit an arrest in the normal expansion of circulating red cells and develop anemia, analogous to KLF1−/− KLF2−/− embryos. In the absence of Myc, circulating erythroid cells do not show the normal increase in α- and β-like globin gene expression but, interestingly, have accelerated erythroid cell maturation between E9.5 and E11.5. This study reveals a novel regulatory network by which KLF1 and KLF2 regulate Myc to control the primitive erythropoietic program.

INTRODUCTION

There are two developmental phases of erythropoiesis: primitive (embryonic) and definitive (adult). In mice, primitive erythropoiesis initiates in the yolk sac as early as embryonic day 7.5 (E7.5) and definitive erythropoiesis begins in the fetal liver by E11.5. The molecular mechanisms that control primitive erythropoiesis are generally less well understood than those that control definitive erythropoiesis. At E9.5, primitive proerythroblast cells synchronously enter the bloodstream from the yolk sac and follow a stepwise developmental program (48). In the circulation, these nucleated proerythroblasts proliferate until approximately E11.5 (18). In parallel, these cells undergo significant upregulation in the expression of embryonic globin, as well as other red cell genes, including surface protein glycophorin A (GPA), heme biosynthesis, and iron transport genes (18, 37, 40). Additionally, these primitive erythroid cells go through cellular and morphological changes in the maturation process from proerythroblasts to orthochromatophilic erythroblasts involving cytoskeletal reorganization, nuclear condensation, and enucleation between E12.5 and E16.5 (27, 41).

Understanding the roles of key transcription factors in primitive erythropoiesis will define molecular mechanisms controlling this process. Krüppel-like factors (KLFs) are a family of transcription factors that bind GC-rich sequences such as CACCC elements via their three carboxy-terminal Cys2/His2 zinc fingers (9, 59). The 17 members in the KLF gene family have important functions in cell differentiation, proliferation, and tissue development (47). Erythroid KLF (EKLF or KLF1) was the first family member to be identified in mice and humans (49) and is a master regulator of definitive erythropoiesis. It is expressed specifically in erythroid cells and positively regulates the adult β-globin gene (20, 49). KLF1−/− mice develop fatal anemia during fetal liver erythropoiesis, due to a defect in the maturation of red blood cells, and die by E16 (11, 52, 58). In KLF1−/− mice, definitive erythroid cells have reduced amounts of transcripts encoding major red cell membrane proteins and heme synthesis enzymes (22, 34, 51). KLF1 is necessary for normal chromatin looping between the β-globin locus control region enhancer and the adult β-globin promoter (21). Furthermore, KLF1 is required for the coassociation of KLF1-regulated genes in nuclear transcription factories (70).

KLF1 is also important in embryonic erythropoiesis. KLF1 directly binds to and positively regulates the human ε- and γ-globin promoters and the mouse Ey- and βh1-globin gene promoters in embryonic erythroid cells (2). KLF1−/− primitive red blood cells have an accumulation of Heinz bodies reminiscent of α-globin chain aggregation and defective membrane morphology (22, 34). Recent evidence indicates that certain mutations in the human KLF1 gene correlate with hereditary persistence of fetal hemoglobin (3, 10). In contrast to its direct positive effect in embryonic cells, KLF1 has an indirect repressive effect on the γ-globin gene in adult cells, most likely via upregulation of BCL11A (10, 81).

In addition to KLF1, its close relative KLF2 (formerly lung KLF or LKLF) is expressed in erythroid cells (6, 7, 14, 46). KLF2 is important for normal primitive erythropoiesis in the yolk sac and, like KLF1, positively regulates mouse and human embryonic β-like globin gene expression in vivo (2). KLF2−/− erythroid cells have abnormal morphology by E10.5 (6); subsequently, embryos develop vascular endothelial cell and heart defects and die by E14.5 (42, 76). Microarray gene expression assays showed that in KLF2−/− mouse E9.5 erythroid precursor cells, 89 genes are significantly downregulated, including genes enriched in erythroid cells (63), such as those that encode the cell signaling factors CD24a antigen, cytotoxic T-lymphocyte-associated protein 2 alpha (Ctla2α), adenylate cyclase 7, and reelin (60). KLF2 can partially compensate for KLF1 function in regulating the mouse embryonic β-globin genes, because KLF1 and KLF2 double knockout (KO) mice die earlier in development at a critical phase of primitive erythropoiesis (5).

KLF1−/− KLF2−/− (double KO) embryos have features that are unique compared to those of single KO mice (5). The embryos are more anemic at E10.5, and Ey- and βh1-globin mRNA levels are considerably lower in double KO embryonic yolk sacs than in either KLF1−/− or KLF2−/− mice. The erythroid cells in E9.5 KLF1−/− KLF2−/− embryos have severely aberrant shapes due to cytoplasmic blebbing and also have altered cell maturation. The more severe blood cell abnormalities in double than in single KOs predict that red cell genes other than that for globin are also coordinately regulated by KLF1 and KLF2 during primitive erythropoiesis.

Only a few genes, other than those for KLF1 and KLF2, are known to specifically control primitive erythropoiesis in mammals. Flk-1 (4), vascular endothelial growth factor (VEGF/VEGF-A) (45), SCL/Tal1 (45, 69), LMO2/Rbtn2 (77), and Gfi-1b (68) are involved in embryonic erythropoiesis, but these factors have more global roles in the development of other hematopoietic lineages and in endothelial cells. GATA1 and GATA2 are specifically involved in embryonic erythropoiesis. Mice lacking GATA1 or GATA2 die between E10.5 and E11.5, exhibit pallor, and have embryonic erythroid cells arrested at an early proerythroblast-like stage or in reduced numbers (28, 75). However, little is known about the gene interaction networks controlled by these factors in primitive erythroid cells.

To identify regulatory pathways and interactions of major significance in primitive erythropoiesis, we investigated its mechanistic control by KLF1 and KLF2. Expression profiling of wild-type (WT), KLF1−/−, and KLF1−/− KLF2−/− E9.5 erythroid cells revealed a limited number of genes expressed with a decreasing trend in these three genotypes. Among these genes, that for Myc was of particular interest, because it is synergistically regulated by KLF1 and KLF2 and mapped at a central node of the most significant regulatory network identified. Importantly, KLF1 and KLF2 bind to and transactivate the promoters of the Myc gene. The role of Myc in embryonic erythropoiesis was then functionally characterized by specific ablation in primitive proerythroblast cells. The absence of Myc induced a block in the normal expansion of the erythroid cell population and in the increase of globin gene expression after E9.5. Interestingly, circulating Myc null erythroid cells displayed early-onset maturation that did not preclude severe anemia in the embryos. Taken together, this study reveals a regulatory network involving KLF1, KLF2, and Myc that is essential for the normal developmental program of primitive erythropoiesis.

MATERIALS AND METHODS

Animals.

The KLF1 KO mouse model was generated by Perkins et al. (58), and mice were obtained from Jackson Laboratory. KLF2 KO mice were generated by Wani et al. (76). KLF1 and KLF2 double KO mice were generated by Basu et al. (5). These mouse models were bred to an FVB/N genetic background to allow valid comparisons of the microarray data.

Characterization of ErGFPcre (EpoRCrein) knock-in and Mycfl/fl mutant mice (provided by U. Klingmüller, S. Philipsen, and F. Alt) was previously described (17, 33). To generate double transgenic mice (Mycfl/fl; EpoR-Cre), EpoRCrein and Mycfl/fl mice were crossed and the progeny were bred with Mycfl/fl or Mycfl/+ mice. Genomic analysis for the KLF1 KO, KLF2 KO, Mycflox, and EpoRCre transgene was performed by PCR (33). The primers for the Mycflox PCR were as follows: forward, 5′ GCCCCTGAATTGCTAGGAAGACTG 3′; reverse, 5′ CCGACCGGGTCCGAGTCCCTATT 3′. The conditions were 94°C for 5 min; 30 cycles of 94°C for 30 s, 55°C for 40 s, and 72°C for 50 s; and 72°C for 7 min. Transgenic mice were selected as homozygous for the murine β-globin haplotype “diffuse.” All experiments conformed to the standards of the Canadian Council on Animal Care and the Virginia Commonwealth University Institutional Animal Care and Use Committee.

Cellular and histological analyses.

For red cell counts, blood from the embryo and yolk sac (WT, KLF1−/−, KLF2−/−, and KLF1−/− KLF2−/−) or from the embryo only (control and Mycfl/fl; EpoR-Cre) was allowed to drain into 4°C 1× phosphate-buffered saline for 15 min. Cells were counted with a hemocytometer. Cytospins were done for 7 min using a Shandon apparatus, and cells were fixed and stained with Giemsa. The control mice for all Mycfl/fl; EpoR-Cre experiments were of the genotypes Mycfl/+ and Mycfl/fl. Heterozygous KO mice were used in timed matings to obtain E9.5 WT, KLF1−/−, KLF2−/−, and KLF1−/− KLF2−/− embryonic yolk sacs, which were embedded in freezing medium and cryosectioned for laser capture microdissection (LCM) (63).

LCM.

The ArcturusXT LCM instrument (Applied Biosystems, Inc.) was used to isolate erythroid cells from yolk sac cryosections obtained from WT, KLF1−/−, and KLF1−/− KLF2−/− embryos. The manufacturer's protocol was followed in preparing the instrument, isolating the cells, and visually inspecting the isolated erythroid cells on the LCM cap. RNA extractions from the LCM-purified samples yielded 10 to 20 ng of RNA. RNA quality was assessed by capillary electrophoresis, and intact samples were processed for microarray hybridization as previously described (64).

Microarrays and data analysis.

Fifteen micrograms of labeled erythroid cell cRNA was fragmented, and 10 μg was hybridized to Affymetrix GeneChip Mouse Genome 430A 2.0 arrays for 18 h (Affymetrix Inc.). The robust multiarray average (RMA) algorithm was used to obtain probe set expression summaries (36). Thereafter, control probe sets were removed, leaving 22,626 probe sets for statistical analysis. A moderated t test (71) was used to compare the 8 WT and 3 KLF1−/− samples using the limma Bioconductor package (29, 71) in the R programming environment (http://www.R-project.org) and to identify the probe sets that were significantly differentially expressed. The Jonckheere-Terpstra trend test (35), a nonparametric test for ordered differences among classes, was performed to identify probe sets having significantly decreasing expression across the 8 WT, 3 KLF1−/−, and 3 KLF1−/− KLF2−/− samples.

qRT-PCR.

Quantitative reverse transcription-PCR (qRT-PCR) was done with RNA extracted from mouse peripheral blood cells (E9.5, E10.5, or E11.5) or K562 cells with TRIzol reagent. cDNA was prepared from total RNA using the iScript cDNA synthesis kit (Bio-Rad, Inc.) or RT with Moloney murine leukemia virus reverse transcriptase (8). The NCBI database (http://www.ncbi.nih.gov) was used to establish that the primers are gene specific. qRT-PCR experiments were performed using the ABI Prism 7300 system (Applied Biosystems, Inc.) or Mx3005P quantitative PCR (qPCR) using SYBR green chemistry. A dissociation curve was used to verify that only a single product was amplified. Mouse cyclophilin A, ribosomal protein S16, or human cyclophilin A mRNA was used as an internal standard for normalization of input cDNA, as appropriate (8). qRT-PCR was performed in duplicate or triplicate for each biological replicate. Relative values were calculated using MxPro v4.01 software. Statistical analyses of qRT-PCR data were performed with a two-sample Student t test, and all findings were judged to be significant using an α ≤ 0.05 level of significance. The forward and reverse primers used in the qRT-PCR assays for the mouse genes were as follows: Gmpr, GGCAGAAGCTGAAACTCT and TCCACGTCCCCTTTGTAA; Pklr, ACGACTCAACTTCTCCCA and GCAAAACTTTCAGCCGC; Tfrc, AAAATGTGAAGCTCATTGTGAAA and CAACACCAGCACCCAAA; Ddx3x, GACCTGCCTAGTGATATCGA and AAGATCCAGTAAATCCTTTGTGA; Myc, TGCTGCATGAGGAGACA and TCGGGATGGAGATGAGC; Ctse, CGAGTGTCAATGAACCCCT and TGCAGTACACAGAAGGGA; Csda, CGAGGACGCGGAGAA and TCTTTGGTGTCATTTCGGTT; cyclophilin A, CACAAACGGTTCCCAGTT and ACCTTCCCAAAGACCACA; Gapdh, GACAACTTTGGCATTGTGG and AGTGGATGCAGGGATGA; εy, CAAGCTACATGTGGATCCTGAGAA and GCCGAAGTGACTAGCCAAAAC; βh1, AGGCAGCTATCACAAGCATCTG and AACTTGTCAAAGAATCTCTGAGTCCAT; ζ, CGAGCTGCATGCCTACAT and GCCATTGTGACCAGCAGACA. The qRT-PCR primers used for GPA were described previously (8). The human gene forward and reverse primers for qRT-PCR were as follows: KLF2, TGCCATCTGTGCGATCGT and GGCTACATGTGCCGTTTCATG; Myc, GACTCCAGCGCCTTCTC and CTTCCTCATCTTCTTGTTCCTCC; cyclophilin A, CCGAGGAAAACCGTGTACTATTAG and TGCTGTCTTTGGGACCTTG.

Chromatin immunoprecipitation (ChIP) assay.

The evolutionarily conserved region (ECR) browser interface was used to navigate through alignments of the human and mouse genes for Myc (56). An ECR is a 100-bp region of at least 70% similarity between mouse and human gene sequences. Mulan was used to align the mouse and human Myc promoters (55). The user-defined motif search tool of rVISTA within the Mulan interface was used to find KLF1 (CCNCNCCC) (25) and KLF2 binding sites (CCACCC or CCGCCC) (39) within ECRs. The website http://www.dcode.org was used to generate the schematic representation in Fig. 4.

Fig 4.

Fig 4

ChIP assays at evolutionarily conserved KLF1 and KLF2 binding sites in the promoters of the mouse gene for Myc. The consensus site for KLF1 binding is CCNCNCCC, and that for KLF2 is CCRCCC (CCACCC or CCGCCC). Each tick mark at the bottom of the diagram represents an ECR between the mouse and human genes for Myc. The ECR sequences are designated intergenic, intron, or coding. P1 (promoter 1) of the gene for Myc is at the left and includes the −126 KLF1/KLF2 consensus site, and P2 is to the right and includes the +2074 consensus binding site. These −126 and +2074 binding sites are within ECRs. ChIP assay values obtained with antibodies specific for KLF1 and KLF2 were compared to that obtained with IgG, which was set to 1. **, P = 0.0158; *, P = 0.0405 (unpaired t tests). The arrows indicate transcription start sites.

ChIP assays were performed with E11.5 erythroid cells as previously described (2). Chromatin was precipitated with anti-KLF1 (Abcam catalog no. AB-2483), anti-KLF2 (KLF2_Ng) (39), and nonspecific rabbit or goat IgG antibodies. Specific primers surrounding the putative KLF binding sites were used to determine KLF1 and KLF2 relative enrichment, which was normalized to IgG. Specific primers for the promoter of mouse β-actin were used as a negative control. The sequences of the primers used for qPCR are as follows: Myc P1 (5′ promoter), TCCTCTTTCCCCGGCTC and TCCTCCTCTCGCTTCCC; Myc P2 (3′ promoter), AGTAAAAGAGTGCATGCCTCC and GTACCCCAATCCTGAACCAC; ACTB (β-actin), ACCCCATTGAACATGGCATT and TGTAGAAGGTGTGGTGCCAGAT.

Transfection assays.

K562 cells (human myeloid leukemia; ATCC CCL-243) were grown and maintained at 37°C and 5% CO2 in RPMI 1640 supplemented with 10% fetal bovine serum. K562 cells were cotransfected with a Myc promoter/luciferase reporter construct, a Renilla luciferase control reporter (pRL-TK; Promega, Inc.), and a KLF1 cDNA expression construct (49). The human Myc promoter/luciferase fusion gene constructs were obtained from Addgene (31, 32). Transfection assays were performed using Amaxa Nucleofector Solution V (Lonza Bio, Inc.) according to the company's optimized protocol for K562 (ATCC) cells. The Dual-Luciferase reporter assay system (Promega, Inc.) was used to measure firefly and Renilla luciferase activities with a TD-20120 luminometer (Turner Designs, Inc.) at 48 h after transfection. The promoterless pBv-luc construct was transfected as a negative control. Three separate transfections were performed, and the mean of the trials was determined. For KLF2 knockdown transient-transfection assays, 5 × 105 K562 cells were transfected with Scramble small interfering RNA (siRNA) or siRNA Hs-KLF2-7 (Qiagen) at 10 nm and 20 nm using Lipofectamine RNAi-Max (Invitrogen) according to the manufacturer's recommendations. Cells were harvested at 36 to 48 h posttransfection. The methods for subsequent qRT-PCR are described above. The statistical analyses of transfection assay results were performed using a two-sample Student t test, and all findings were judged to be significant when α was ≤0.05.

Microarray data accession number.

The raw data from the microarrays (.CEL files) have been deposited in GEO under accession number GSE36427.

RESULTS

Expression profiling of KLF1−/− and KLF1−/− KLF2−/− embryonic erythroid cells.

To explore the complex network of genes controlled by KLF1 and KLF2 in the primitive erythropoietic program, gene expression profiling was undertaken. We reasoned that genes important for KLF1−/− KLF2−/−-specific phenotypes are synergistically regulated by KLF1 and KLF2. E9.5 embryos were selected for this study because KLF1−/− KLF2−/− embryos are normal in gross appearance at that time point, unlike at E10.5 (5). E9.5 erythroid precursor cells were isolated from embryonic yolk sacs by LCM because it facilitated the enrichment of erythroid precursor cells. Total RNA from 8 WT, 3 KLF1−/−, and 3 KLF1−/− KLF2−/− samples were used for Affymetrix 430A 2.0 microarray assays (6365).

WT and KLF1−/− samples were compared using a moderated t test, and 146 genes were identified as being significantly differentially expressed. The genes that are downregulated in KLF1−/− compared to WT samples are indicated by a negative n-fold change in Table 1, and those upregulated are indicated by a positive n-fold change. Of the 47 genes that are significantly downregulated in embryonic erythropoiesis, many are also downregulated in adult KLF1−/− erythroid cells, such as those for the transferrin receptor (Tfrc), erythrocyte protein band 4.9 (Epb4.9), and α-hemoglobin-stabilizing protein (Ahsp) (34). However, some genes, such as those for E2F2, p18 (Cdkn2c), and p21 (Cdkn1a) (61, 72), that are dysregulated in KLF1−/− definitive erythroid cells are unaltered in primitive erythroid cells. On the other hand, a number of genes are significantly downregulated in KLF1−/− compared to WT primitive erythroid cells, including those for pyruvate kinase liver and red blood cell (Pklr), adenylate cyclase 7 (Adcy7), and Myc.

Table 1.

Genes that are up- and downregulated in E9.5 KLF1−/− versus WT yolk sac erythroid cellsa

AffyID Entrez ID Gene symbol Expression summary for:
Fold change P value
WT KLF1−/−
1419175_a_at 12231 Btn1a1 6.94 9.00 4.19 0.0000205
1449360_at 12984 Csf2rb2 6.49 8.22 3.33 0.0065116
1421811_at 21825 Thbs1 8.86 10.58 3.30 0.0041092
1434853_x_at 54484 Mkrn1 9.75 11.41 3.16 0.0046076
1449280_at 71690 Esm1 6.07 7.66 3.02 0.0000491
1460241_a_at 20454 St3gal5 6.41 8.00 3.00 0.0073785
1418480_at 57349 Ppbp 6.55 8.08 2.89 0.0000965
1456014_s_at 108101 Fermt3 8.98 10.48 2.84 0.0007647
1436905_x_at 16792 Laptm5 8.05 9.47 2.68 0.0076590
1455504_a_at NA NA 9.41 10.82 2.67 0.0023565
1460232_s_at NA NA 7.31 8.69 2.59 0.0008638
1451425_a_at 54484 Mkrn1 8.51 9.86 2.54 0.0026376
1418835_at 21664 Phlda1 8.86 10.19 2.51 0.0014728
1432004_a_at 13430 Dnm2 5.19 6.50 2.48 0.0041875
1424966_at 94346 Tmem40 6.31 7.56 2.36 0.0055598
1435386_at 22371 Vwf 5.52 6.76 2.35 0.0018190
1450852_s_at 14062 F2r 9.92 11.15 2.35 0.0043781
1416811_s_at NA NA 6.85 8.07 2.33 0.0018710
1418412_at 21987 Tpd52l1 6.73 7.93 2.30 0.0002513
1451263_a_at 11770 Fabp4 7.76 8.94 2.25 0.0006915
1423062_at 16009 Igfbp3 6.55 7.71 2.23 0.0004194
1448471_a_at 13024 Ctla2a 9.62 10.75 2.19 0.0097492
1437726_x_at 12260 C1qb 6.06 7.15 2.12 0.0018801
1448954_at 78593 Nrip3 6.17 7.22 2.07 0.0007463
1416488_at 12452 Ccng2 8.24 9.29 2.07 0.0004951
1448736_a_at 15452 Hprt 10.18 11.20 2.02 0.0023947
1450344_a_at 19218 Ptger3 6.15 7.15 1.99 0.0036792
1439259_x_at 105501 Abhd4 9.43 10.42 1.98 0.0006879
1422798_at 66797 Cntnap2 7.90 8.88 1.97 0.0058018
1426970_a_at NA NA 7.15 8.10 1.93 0.0043149
1422474_at 18578 Pde4b 6.97 7.91 1.92 0.0042260
1422977_at 14724 Gp1bb 6.23 7.15 1.90 0.0000247
1416882_at 67865 Rgs10 9.10 10.02 1.89 0.0006363
1452352_at 13025 Ctla2b 8.62 9.54 1.89 0.0079715
1451196_at 383295 Ypel5 7.97 8.89 1.89 0.0008458
1427168_a_at 12818 Col14a1 7.32 8.23 1.89 0.0012275
1434059_at 230088 B230312A22Rik 7.36 8.24 1.84 0.0034893
1451453_at 13143 Dapk2 6.85 7.72 1.83 0.0037859
1433593_at 383295 Ypel5 5.58 6.44 1.81 0.0011630
1420664_s_at 19124 Procr 9.01 9.86 1.81 0.0080605
1418435_at 54484 Mkrn1 7.99 8.80 1.76 0.0076558
1435275_at 333182 Cox6b2 6.90 7.71 1.76 0.0058952
1424923_at 20715 Serpina3g 6.99 7.79 1.74 0.0036619
1426454_at 11857 Arhgdib 8.86 9.65 1.73 0.0075277
1423319_at 15242 Hhex 7.72 8.51 1.73 0.0023488
1425158_at 57246 Tbx20 5.45 6.24 1.72 0.0095330
1448995_at 56744 Pf4 11.21 11.96 1.68 0.0064027
1422444_at 16403 Itga6 7.25 7.98 1.66 0.0019243
1438261_at 56222 Cited4 6.32 7.05 1.65 0.0002694
1415874_at 24063 Spry1 8.32 9.04 1.65 0.0005373
1415949_at 12876 Cpe 6.99 7.71 1.64 0.0009311
1423306_at 106878 2010002N04Rik 7.48 8.19 1.64 0.0016944
1417214_at 80718 Rab27b 5.42 6.11 1.61 0.0016003
1433964_s_at 108101 Fermt3 6.55 7.23 1.61 0.0065248
1419259_at 20163 Rsu1 10.12 10.79 1.60 0.0031843
1417023_a_at 11770 Fabp4 6.47 7.14 1.59 0.0037652
1426450_at 224860 Plcl2 7.69 8.34 1.57 0.0005064
1450016_at 12450 Ccng1 8.91 9.55 1.56 0.0059483
1448657_a_at 56812 Dnajb2 8.41 9.03 1.54 0.0031640
1452358_at 24004 Rai2 3.93 4.55 1.53 0.0000153
1427040_at 16543 Mdfic 8.38 8.98 1.52 0.0062468
1452366_at 234356 Csgalnact1 6.14 6.74 1.52 0.0073674
1448694_at 16476 Jun 6.84 7.43 1.50 0.0008474
1421268_at 22234 Ugcg 6.49 7.07 1.50 0.0062467
1434087_at 17769 Mthfr 6.96 7.54 1.50 0.0082196
1416315_at 105501 Abhd4 7.33 7.91 1.50 0.0065987
1417978_at 66892 Eif4e3 7.72 8.30 1.49 0.0018258
1423212_at 13619 Phc1 8.22 8.79 1.49 0.0074183
1455220_at 212398 Frat2 7.35 7.92 1.48 0.0019347
1421633_a_at 12950 Hapln1 5.95 6.51 1.47 0.0086383
1426734_at 224093 Fam43a 6.99 7.53 1.46 0.0063603
1417426_at 19073 Srgn 7.89 8.43 1.46 0.0098012
1419298_at 269823 Pon3 5.49 6.02 1.44 0.0032081
1418104_at 78593 Nrip3 5.32 5.83 1.43 0.0048456
1428794_at 432572 Specc1 7.03 7.54 1.42 0.0032746
1420124_s_at 102791 Tcta 7.39 7.90 1.42 0.0025608
1425570_at 27218 Slamf1 6.83 7.33 1.41 0.0089085
1415850_at 19414 Rasa3 7.01 7.50 1.40 0.0049996
1416714_at 15900 Irf8 5.98 6.46 1.40 0.0004215
1416843_at 18582 Pde6d 6.72 7.20 1.39 0.0057671
1431597_a_at 78593 Nrip3 4.44 4.90 1.38 0.0007931
1423286_at 12404 Cbln1 7.91 8.36 1.37 0.0053469
1425506_at 107589 Mylk 8.11 8.56 1.36 0.0066188
1448134_at 27355 X99384 7.79 8.24 1.36 0.0040177
1426624_a_at 66090 Ypel3 8.03 8.48 1.36 0.0098438
1418144_a_at 18720 Pip5k1a 7.39 7.82 1.36 0.0053209
1425125_at 18302 Oit3 6.72 7.16 1.35 0.0034544
1435383_x_at 17984 Ndn 11.13 11.57 1.35 0.0051937
1422699_at 11684 Alox12 5.37 5.80 1.35 0.0096007
1437401_at 16000 Igf1 6.44 6.86 1.34 0.0042858
1434555_at 11737 Anp32a 9.53 9.95 1.33 0.0032632
1435382_at 17984 Ndn 11.08 11.49 1.33 0.0087450
1449402_at 60322 Chst7 6.27 6.67 1.32 0.0037640
1422817_at 14729 Gp5 6.54 6.94 1.32 0.0072048
1436448_a_at 19224 Ptgs1 5.83 6.23 1.32 0.0032874
1437849_x_at 67416 Armcx2 9.95 10.35 1.32 0.0051815
1417363_at 22719 Zfp61 5.67 6.06 1.31 0.0090568
1455180_at 102371 Gcom1 5.55 5.93 1.31 0.0066221
1416935_at 22368 Trpv2 6.15 6.53 1.30 0.0057705
1427007_at 74131 Sash3 7.24 7.62 1.30 0.0070703
1417755_at 106021 Topors 5.54 5.88 1.27 0.0086579
1448024_at 18162 Npr3 6.12 6.46 1.27 0.0059614
1417523_at 56193 Plek 6.52 6.86 1.26 0.0014985
1448443_at 20713 Serpini1 5.44 5.75 1.24 0.0041357
1455367_at 213236 Dnd1 5.46 5.75 1.22 0.0050743
1428816_a_at 14461 Gata2 6.86 7.15 1.22 0.0071113
1427287_s_at 16439 Itpr2 5.18 5.44 1.20 0.0060864
1426604_at 24014 Rnasel 4.38 4.63 1.18 0.0096496
1450442_at 11519 Add2 7.44 7.11 −1.26 0.0046469
1422866_at 12817 Col13a1 6.77 6.43 −1.26 0.0015366
1455035_s_at 67134 Nop56 9.69 9.29 −1.32 0.0029171
1428390_at 72515 Wdr43 9.75 9.33 −1.33 0.0069107
1429080_at 67973 Mphosph10 9.02 8.60 −1.34 0.0017344
1419660_at 67008 1600012F09Rik 8.85 8.40 −1.36 0.0027637
1424021_at 65103 Arl6ip6 8.09 7.63 −1.37 0.0087204
1427954_at 270802 BC048403 4.82 4.36 −1.38 0.0095131
1420918_at 170755 Sgk3 6.97 6.51 −1.38 0.0061668
1417675_a_at 100019 Mdn1 7.68 7.17 −1.42 0.0098793
1449623_at 232223 Txnrd3 6.54 6.01 −1.45 0.0063133
1424437_s_at 192663 Abcg4 6.73 6.19 −1.45 0.0023347
1424858_at 217666 L2hgdh 7.48 6.91 −1.49 0.0063305
1424722_at 71775 1300017J02Rik 7.94 7.36 −1.49 0.0078258
1436990_s_at NA NA 10.46 9.85 −1.52 0.0045047
1460371_at 72630 Hspa12b 7.80 7.19 −1.53 0.0085880
1421883_at 15569 Elavl2 4.72 4.09 −1.55 0.0070508
1420339_at 76890 Memo1 8.46 7.82 −1.56 0.0019363
1434959_at 13363 Dhh 7.28 6.51 −1.71 0.0000812
1422920_at 57255 Cldn13 7.03 6.21 −1.76 0.0048145
1451229_at 232232 Hdac11 6.72 5.89 −1.78 0.0045228
1424117_at 414077 BC056474 7.03 6.18 −1.80 0.0052470
1449424_at 27260 Plek2 8.34 7.47 −1.83 0.0025433
1417092_at 19228 Pth1r 6.10 5.22 −1.84 0.0083709
1452324_at 19296 Pvt1 8.26 7.38 −1.84 0.0001237
1424942_a_at 17869 Myc 7.35 6.46 −1.85 0.0021150
1437614_x_at 224454 Zdhhc14 7.76 6.86 −1.87 0.0032367
1438975_x_at 224454 Zdhhc14 7.81 6.84 −1.95 0.0010981
1418591_at 58233 Dnaja4 7.58 6.59 −1.99 0.0014377
1432332_a_at 110959 Nudt19 8.25 7.19 −2.09 0.0009313
1448300_at 66447 Mgst3 9.75 8.68 −2.09 0.0023340
1460223_a_at 13829 Epb4.9 7.45 6.30 −2.22 0.0004694
1437502_x_at 12484 Cd24a 10.12 8.94 −2.27 0.0001713
1460444_at 109689 Arrb1 6.05 4.83 −2.33 0.0049112
1448182_a_at 12484 Cd24a 9.31 8.05 −2.40 0.0011112
1448530_at 66355 Gmpr 8.13 6.85 −2.42 0.0001778
1438619_x_at 224454 Zdhhc14 7.45 6.16 −2.44 0.0000852
1416034_at 12484 Cd24a 8.64 7.27 −2.59 0.0000683
1438711_at 18770 Pklr 8.63 7.21 −2.66 0.0000103
1451415_at 69068 1810011O10Rik 8.14 6.71 −2.68 0.0000060
1418600_at 16596 Klf1 10.67 9.15 −2.87 0.0013763
1452661_at 22042 Tfrc 11.40 9.87 −2.89 0.0017499
1456307_s_at 11513 Adcy7 8.62 7.02 −3.05 0.0000228
1422930_at 78369 Icam4 8.33 6.70 −3.09 0.0000331
1417184_s_at NA NA 11.67 9.97 −3.24 0.0011955
1435945_a_at 16534 Kcnn4 10.26 8.46 −3.48 0.0000054
1427866_x_at 15130 Hbb-b2 8.07 6.15 −3.79 0.0048748
1418909_at 27028 Ermap 10.17 8.11 −4.16 0.0013191
1420468_at 66772 Asb17 7.24 4.93 −4.96 0.0000063
1449077_at 170812 Ahsp 12.98 8.11 −29.35 0.0000000
a

Mean probe set expression summaries for WT and KLF−/− cells were calculated by RMA. Some genes were detected by multiple AffyID probe sets, and there are multiple entries for these genes. Positive n-fold values indicate genes that are upregulated in KLF1−/− versus WT erythroid cells; negative n-fold values indicate genes downregulated in KLF1−/− versus WT erythroid cells.

To define genes that might be synergistically regulated by KLF1 and KLF2, the Jonckheere-Terpstra trend test was used to identify probe sets having significantly decreasing expression across the WT, KLF1−/−, and KLF1−/− KLF2−/− E9.5 embryonic erythroid cell samples. There were 110 probe sets corresponding to 101 unique genes that had a significant decreasing trend of expression in WT, KLF1−/−, and KLF1−/− KLF2−/− erythroid cell samples (Table 2). The major functional classifications of these genes include homeostasis, regulation of apoptosis, hemopoiesis, and positive regulation of biosynthetic and macromolecule metabolic processes (Fig. 1). Because KLF1 and KLF2 have generally positive roles in gene regulation, we focused on genes that are down- rather than upregulated in mutants, to increase the likelihood of discovering genes that are directly regulated by KLF1 and KLF2. Several of the downregulated genes encode important red cell membrane and cytoskeletal proteins, such as Tfrc, adducin, Epb4.9, and the solute carrier 4 anion exchanger (SLc4a1), indicating previously unrecognized roles for KLF1 and KLF2 in coordinately regulating these genes in primitive cells. Decreased expression of these cell membrane and cytoskeletal genes may influence the observed abnormal cell morphology in embryonic erythroid cells, which is more severe in KLF1−/− KLF2−/− cells than in KLF1−/− or KLF2−/− cells. Certain red cell enzyme genes have more pronounced downregulation in KLF1−/− KLF2−/− cells than in KLF1−/− cells, including Adcy7 and Pklr.

Table 2.

Genes that are significantlya decreased in expression across WT, KLF1−/−, and KLF1−/− KLF2−/− embryonic erythroid cells

Gene symbol Description P value AffyID
Elavl2 ELAVc 0.000518 1421883_at
Gmpr3 GMPR 0.000518 1448530_at
Zdhhc14 Zinc finger, DHHC domain containing 14 0.000518 1438619_x_at
Arrb1 Arrestin, beta 1 0.000518 1460444_at
Sgk3 Serum/glucocorticoid regulated kinase 3 0.000518 1420918_at
Cd24ab CD24a antigen 0.000797 1437502_x_at
Memo1 Mediator of cell motility 1 0.000797 1420339_at
Klf1 KLF1 (erythroid) 0.000797 1418600_at
1600012F09Rik RIKEN cDNA 1600012F09 gene 0.000797 1419660_at
Hdac11 Histone deacetylase 11 0.000797 1451229_at
Pvt1 Plasmacytoma variant translocation 1 0.001211 1452324_at
Eraf Erythroid-associated factor 0.001211 1449077_at
Tfrc TFRC 0.001211 1452661_at
Pklr PKLR 0.001211 1438711_at
Orc2l Origin recognition complex, subunit 2 like (Saccharomyces cerevisiae) 0.001211 1418226_at
Mmaa Methylmalonic aciduria (cobalamin deficiency) type A 0.001211 1417857_at
Dhh Desert hedgehog 0.001211 1434959_at
Icam4 Intercellular adhesion molecule 4, Landsteiner-Wiener blood group 0.001211 1422930_at
Epb4.9 Erythrocyte protein band 4.9 0.001211 1460223_a_at
Chchd10 Coiled-coil-helix-coiled-coil-helix domain containing 10 0.001211 1436990_s_at
Nudt19 Nudixd-type motif 19 0.001812 1432332_a_at
Wdr43 WD repeat domain 43 0.001812 1428390_at
Ankrd28 Ankyrin repeat domain 28 0.001812 1454801_at
Kcnn4 Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4 0.001812 1435945_a_at
Mphosph10 M-phase phosphoprotein 10 (U3 small nucleolar ribonucleoprotein) 0.001812 1429080_at
Nudcd1 NudC domain containing 1 0.001812 1429655_at
Nudt19 Nudix-type motif 19 0.001812 1434216_a_at
Pth1r Parathyroid hormone 1 receptor 0.001812 1417092_at
Usp59 Ubiquitin-specific peptidase 49 0.001812 1453037_at
Add2 Adducin 2 (beta) 0.001812 1450442_at
Ddx3x DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 3, X linked 0.001812 1416467_at
AU021092 Expressed sequence AU021092 0.002673 1437661_at
BC048403 cDNA sequence BC048403 0.002673 1427954_at
Asb17 Ankyrin repeat and SOCS box-containing 17 0.002673 1420468_at
Cldn13 Claudin 13 0.002673 1422920_at
Zdhhc14 Zinc finger, DHHC domain containing 14 0.002673 1437614_x_at
Hspa12b Heat shock protein 12B 0.002673 1460371_at
Dnaja4 DnaJ (Hsp40) homolog, subfamily A, member 4 0.002673 1418591_at
Slc4a1 Solute carrier family 4 (anion exchanger), member 1 0.002673 1416464_at
Atg5 Autophagy-related 5 (yeast) 0.002673 1418235_at
Myc Myelocytomatosis oncogene 0.002673 1424942_a_at
Pfkp Phosphofructokinase, platelet 0.002673 1430634_a_at
BC048355 cDNA sequence BC048355 0.002673 1460713_at
Exosc2 Exosome component 2 0.002673 1426630_at
Nme3 Nonmetastatic cells 3, protein expressed in 0.002673 1448905_at
Irf4 Interferon regulatory factor 4 0.002673 1421174_at
AA517858 Expressed sequence AA517858 0.003229 1420121_at
Adcy7 Adenylate cyclase 7 0.003888 1456307_s_at
BC056474 cDNA sequence BC056474 0.003888 1424117_at
Hbb-b1/Hbb-b2 Hemoglobin, beta adult major chain/hemoglobin, beta adult minor chain 0.003888 1417184_s_at
Cd24ab CD24a antigen 0.003888 1416034_at
Abcg4 ATP-binding cassette, subfamily G (WHITE), member 4 0.003888 1424437_s_at
Elavl2 ELAV 0.003888 1421882_a_at
Dck Deoxycytidine kinase 0.003888 1439012_a_at
Ctse Cathepsin E 0.003888 1418989_at
Pklr PKLR 0.003888 1421258_a_at
Ermap Erythroblast membrane-associated protein 0.003888 1418909_at
2810453I06Rik RIKEN cDNA 2810453I06 gene 0.003888 1418389_at
Zdhhc14 Zinc finger, DHHC domain containing 14 0.003888 1438975_x_at
1110007M04Rik RIKEN cDNA 1110007M04 gene 0.003888 1427997_at
Gcnt2 Glucosaminyl (N-acetyl) transferase 2, I-branching enzyme 0.003888 1430826_s_at
1300017J02Rik RIKEN cDNA 1300017J02 gene 0.003888 1424722_at
Fgf13 Fibroblast growth factor 13 0.003888 1418498_at
Atp13a2 ATPase type 13A2 0.003888 1452747_at
Txnrd3 Thioredoxin reductase 3 0.003888 1449623_at
Myo19 Myosin XIX 0.003888 1451183_at
Cd24a CD24a antigen 0.005577 1448182_a_at
Nop10 NOP10 ribonucleoprotein homolog (yeast) 0.005577 1423210_a_at
Hbb-b2 Hemoglobin, beta adult minor chain 0.005577 1427866_x_at
Slc4a1 Solute carrier family 4 (anion exchanger), member 1 0.005577 1434502_x_at
Xpo5 Exportin 5 0.005577 1451468_s_at
Mapk6 Mitogen-activated protein kinase 6 0.005577 1419169_at
Rhd Rh blood group, D antigen 0.005577 1417049_at
Mgst3 Microsomal glutathione S-transferase 3 0.005577 1448300_at
Traf6 Tumor necrosis factor receptor-associated factor 6 0.005577 1435350_at
Clps Colipase, pancreatic 0.005577 1438612_a_at
Dnaja4 DnaJ (Hsp40) homolog, subfamily A, member 4 0.005577 1434196_at
Tlcd1 TLC domain containing 1 0.005577 1452132_at
Olfr78 Olfactory receptor 78 0.005577 1421507_at
Nol5a Nucleolar protein 5A 0.005577 1455035_s_at
Igfals Insulin-like growth factor binding protein, acid-labile subunit 0.005577 1422826_at
Traf4 Tumor necrosis factor receptor-associated factor 4 0.005577 1416571_at
Rrp1b rRNA processing 1 homolog B (S. cerevisiae) 0.005577 1452119_at
Srfbp1 Serum response factor binding protein 1 0.005577 1420509_at
Tmem49 Transmembrane protein 49 0.005577 1421491_a_at
Grin2b Glutamate receptor, ionotropic, NMDA2B (epsilon 2) 0.006644 1422223_at
Usp38 Ubiquitin-specific peptidase 38 0.007888 1428592_s_at
Bco2 Beta-carotene oxygenase 2 0.007888 1421221_at
Arl6ip6 ADP-ribosylation factor-like 6 interacting protein 6 0.007888 1424021_at
Sphk1 Sphingosine kinase 1 0.007888 1451596_a_at
Frrs1 Ferric-chelate reductase 1 0.007888 1423465_at
Pigq Phosphatidylinositol glycan anchor biosynthesis, class Q 0.007888 1437999_x_at
Plek2 Pleckstrin 2 0.007888 1449424_at
Ppara Peroxisome proliferator-activated receptor alpha 0.007888 1449051_at
Ela3 Elastase 3, pancreatic 0.007888 1415883_a_at
Tcp11 T-complex protein 11 0.007888 1420730_a_at
Unknown Unknown 0.007888 1425499_at
Josd2 Josephin domain containing 2 0.007888 1449046_a_at
1810029B16Rik RIKEN cDNA 1810029B16 gene 0.007888 1423289_a_at
Slc25a37 Solute carrier family 25, member 37 0.007888 1417750_a_at
Pde3a Phosphodiesterase 3A, cGMP inhibited 0.007888 1431914_at
Echdc1 Enoyl coenzyme A hydratase domain containing 1 0.007888 1419552_at
Csda Cold shock domain protein A 0.007888 1451012_a_at
Pura Purine-rich element binding protein A 0.007888 1449934_at
Btg2 B-cell translocation gene 2, antiproliferative 0.007888 1448272_at
2610209A20Rik RIKEN cDNA 2610209A20 gene 0.007888 1423357_at
Ndn Necdin 0.007888 1456575_at
Isoc1 Isochorismatase domain containing 1 0.007888 1425051_at
Foxm1 Forkhead box M1 0.007888 1448833_at
1600014C10Rik RIKEN cDNA 1600014C10 gene 0.007888 1436289_x_at
a

P < 0.01.

b

Included are the products of 101 unique genes, but some were detected by multiple AffyID probe sets and there are multiple entries for these.

c

ELAV, embryonic lethal, abnormal vision, Drosophila-like 2 (Hu antigen B).

d

Nudix, nucleoside diphosphate-linked moiety X.

Fig 1.

Fig 1

Functional classifications of genes exhibiting significantly decreasing expression across WT, KLF1−/−, and KLF1−/− KLF2−/− embryonic erythroid cells. Functional gene categories were determined with GO (Gene Ontology) using DAVID (Database Annotation Visualization and Integrated Discovery, http://david.abcc.ncifcrf.gov/). Eleven of the 101 genes identified were not registered by DAVID. The graph illustrates categories with at least 5 genes. Some genes are in multiple categories. The values on the x axis are numbers of genes. ncRNA, noncoding RNA.

An embryonic erythroid cell regulatory network controlled by KLF1 and KLF2.

Figure 2A shows the values for the RMA expression summaries for 7 selected genes that are downregulated with a decreasing monotonic trend in WT, KLF1−/−, and KLF1−/− KLF2−/− primitive erythroid precursor cells. These genes were chosen for further analysis because their P values were relatively low (Table 2), and except for that of the gene for Ddx3x, their expression is enriched in E9.5 yolk sac embryonic erythroid cells compared to epithelial cells, based on our previous data (63). The selected genes encode GMP reductase (Gmpr); Pklr; Tfrc; DEAD/H box polypeptide 3, X linked (Ddx3x); Myc; cathepsin E (Ctse); and cold shock protein A (Csda).

Fig 2.

Fig 2

(A) RMA expression summaries for selected genes regulated by KLF1 and KLF2. LCM was performed to collect independent E9.5 WT, KLF1−/−, and KLF1−/− KLF2−/− mouse erythroid precursor cells. Peptidylprolyl isomerase A (PPIA) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNAs were used as negative controls that are not regulated by KLF1 or KLF2. The average and normalized RMA expression summary is indicated on the y axis. Black bars indicate WT (n = 8) samples, gray bars indicate KLF1−/− (n = 3) samples, and white bars indicate KLF1−/− KLF2−/− (n = 3) samples. The error bars indicate the standard errors. (B) qRT-PCR verification of KLF1- and KLF2-regulated genes identified by microarray analyses. Independent samples of E10.5 WT, KLF1−/−, KLF2−/−, and KLF1−/− KLF2−/− peripheral blood cells were collected. qRT-PCR was performed with at least 100 erythroid cells per assay. GAPDH mRNA was used to normalize the data. The expression amounts in the mutants are expressed relative to those in the WT (100%). PPIA mRNA was used as a negative control. Black bars indicate WT samples, striped bars indicate KLF2−/− samples, gray bars indicate KLF1−/− samples, and white bars indicate KLF1−/− KLF2−/− samples. The values shown are mean of at least 3 independent samples. The error bars indicate the standard errors. Asterisks indicate significant difference from the WT (P < 0.05).

To validate that the 7 genes are differentially expressed in WT, KLF2−/−, KLF1−/−, and KLF1−/− KLF2−/− cells, qRT-PCR analysis was performed with E10.5 peripheral blood cells (Fig. 2B). To avoid bias, the samples used for qRT-PCR were obtained independently from those used for the microarray analyses. The amount of mRNA in the WT was taken as 100%, and the expression in mutants was compared to that in the WT. For each gene, the qRT-PCR results confirm a decreasing trend in WT, KLF1−/−, and KLF1−/− KLF2−/− erythroid cells. For 6 of the 7 genes (except that for Gmpr), expression was significantly lower in KLF1−/− KLF2−/− than in WT cells. The consistency between the qRT-PCR and microarray analyses strengthens the validity of the microarrays. In Fig. 2B, it is possible to directly compare KLF2−/− to the other genotypes, and KLF2 has a positive role in gene expression, but in general, its effect is more modest than that of KLF1. This may be explained by the uniquely KLF1-mediated transcription factories that coordinate the transcription of highly transcribed erythroid cell genes, including those for globin and cell membrane structural proteins (70).

To determine which genes might be the cause of the more severe KLF1−/− KLF2−/− erythroid cell phenotype compared to that of single KOs, ingenuity pathway analysis (IPA) of the 101 unique genes in Table 2 was used to discover gene networks. Seventeen networks were identified by IPA, but one was by far the most significant, scoring 46, indicating a P value of 1E-46. The functions of this IPA network include behavior, cell cycle, connective tissue development, and function (Fig. 3). Five of the 7 genes which were verified by qRT-PCR appear in this network (those for Myc, Tfrc, Pklr, Ddx3x, and Csda). The gene for Myc has an integral role in the network, forming a nexus, whereas some of the other genes appear to be more peripheral. Furthermore, as shown in Fig. 2B, Myc mRNA is reduced by approximately 2-fold in KLF2−/− erythroid cells and by 3-fold in KLF1−/− erythroid cells but by 10-fold in KLF1−/− KLF2−/− erythroid cells, compared to that in the WT. Therefore, KLF1 and KLF2 have synergistic roles in Myc regulation, as the reduction in the amount of Myc mRNA in the double KO is more than additive compared to the single KOs. The number of circulating erythroid cells at E10.5 is reduced in KLF1−/− (4.5 ± 0.9 × 105, n = 8; P < 0.003) and KLF1−/− KLF2−/− (2.6 ± 0.4 × 105, n = 4; P < 0.002) embryos and yolk sacs compared to that in WT embryos and yolk sacs (8.7 ± 0.7 × 105, n = 29). These data are consistent with a role for Myc in the proliferation of primitive erythroid cells.

Fig 3.

Fig 3

IPA network of genes coordinately regulated by KLF1 and KLF2. The 101 genes from Table 1, which had a significant decreasing trend of expression in WT, KLF1−/−, and KLF1−/− KLF2−/− erythroid cell samples, were subjected to IPA analysis. The most significant network is shown. It scored 46, indicating that the overall P value is 1E-46; P values for individual genes are shown in Table 2. The network functions include behavior, cell cycle, connective tissue development, and function. Solid lines indicate direct relationships, and dashed lines indicate indirect relationships. Genes with statistically significant P values from the microarray assays are shaded, and darker shading represents lower P values.

The Mycg gene is directly regulated by KLF1 and KLF2.

ChIP assays were used to determine if KLF1 and KLF2 bind to consensus sites in the Myc promoters in WT E11.5 circulating mouse red blood cells. The consensus sites that were selected for the ChIP assays reside in regions evolutionarily conserved between the mouse and human genes. The results in Fig. 4 indicate that KLF1 binds to a CACCC element in the P1 promoter of the gene for Myc located at base position −126 (significant 7-fold enrichment compared to the IgG control). This validates ChIP-seq experiments with definitive erythroid cells which suggest that KLF1 binds near the Myc gene (60). Interestingly, KLF2 also binds to the gene for Myc in primitive erythroid cells (Fig. 4) but at the P2 promoter at base position +2074 (significant 2-fold enrichment compared to IgG). Both the P1 and P2 promoters of the Myc gene are utilized in erythroid cells (66).

To determine whether KLF1 and KLF2 directly regulate the gene for Myc, transient-transfection assays were performed with K562 cells, a human cell line representing the embryonic/fetal erythroid cell compartment. As shown in Fig. 5A, a KLF1 cDNA expression vector transactivates two different Myc gene promoter-luciferase fusion constructs containing the KLF1 binding site at P1 (Del-2269 and Del-352) by 2.2- and 3.4-fold, respectively, indicating a direct effect of KLF1 on the regulation of the Myc gene. Because KLF2 (80), unlike KLF1 (20), is already expressed in K562 cells, knockdown experiments were undertaken with two KLF2 siRNAs. As shown in Fig. 5B for one of these, 5- or 10-fold KLF2 knockdown in K562 cells was achieved compared to the Scramble control siRNA, and there was a correlative 2- or 3-fold reduction in Myc mRNA. Reduced Myc mRNA was observed with the two independent KLF2 siRNAs, suggesting that KLF2 positively regulates the expression of the Myc gene.

Fig 5.

Fig 5

(A) KLF1 transactivates the promoters of the gene for Myc in K562 cells. The black bars represent transfections with the luciferase reporter gene only, and the gray bars represent transfections which additionally have the expression plasmid pSG5-mEKLF. pBv-luc is a promoterless luciferase construct. Del-2269 and Del-352 are two different Myc promoter-luciferase fusion constructs. Data are the mean values from at least 3 independent samples. Error bars indicate standard deviations. **, P < 0.0001; *, P = 0.0034. The same total amount of DNA was used in each transfection by including the pSG5 empty vector in the transfections without the expression construct. (B) Knockdown indicates that KLF2 regulates Myc expression in K562 cells. K562 cells were transfected with Scramble siRNA, KLF2 siRNA (10 nm), or KLF2 siRNA (20 nm). qRT-PCR assay of KLF2 (black bars) and Myc (c-Myc, white bars) mRNAs in transfected cells was performed using cyclophilin A mRNA as an internal standard. The KLF2-to-cyclophilin A mRNA (siRNA Scramble) and Myc-to-cyclophilin A mRNA (siRNA Scramble) ratios were taken as 100%. Error bars show standard deviations. Data are the mean values of 2 independent samples. An asterisk indicates significant difference from siRNA Scramble (P < 0.05).

In vivo characterization of the role of Myc in embryonic erythropoiesis.

To determine whether Myc downregulation in KLF1−/− KFL2−/− embryos plays a critical role in embryonic erythropoiesis, Myc expression was specifically suppressed in the late stages of erythropoiesis and maturation, because Myc null embryos have severe developmental abnormalities and die in mid-gestation before E10 (16). Cell-autonomous Myc ablation was induced by mating mice carrying the Myc conditional allele (Mycfl/fl) and mice with the erythropoietin receptor Cre allele (EpoR-Cre), which express Cre from the proerythroblast onward (44). The Mycfl/fl; EpoR-Cre mice are alive at E11.5 (n = 8/8) but die by E12.5 (n = 6/6). Similar to KLF1−/− KLF2−/− embryos, the Mycfl/fl; EpoR-Cre mice were indistinguishable from controls at E9.5 but pale and apparently anemic at E10.5 (n = 26) and 11.5 (n = 19) (Fig. 6).

Fig 6.

Fig 6

Phenotypes of control and Mycfl/fl; EpoR-Cre embryos. At E10.5, the Mycfl/fl; EpoR-Cre embryo and yolk sac (right) are very pale and vessel arborization is difficult to define compared to that in controls (left). Images were obtained with a Leica MZ12 stereomicroscope. Magnification, ×2. At E11.5, dissected Mycfl/fl; EpoR-Cre embryos (right), relative to controls (left), appeared to have a growth delay, exhibited a drastic decrease in erythroid cells within the vasculature, and were severely anemic, which is indicative of erythroid cell deficiency. Images were obtained with a Leica MZ12 stereomicroscope. Magnification, ×1.25.

Quantification of circulating primitive erythroid cells was carried out with E9.5 to E11.5 control and Mycfl/fl; EpoR-Cre embryos (Table 3). The number of cells in controls increased markedly between E9.5 and E10.5 by ∼14-fold (P < 0.0002) and between E10.5 and E11.5 by an additional ∼4-fold (P < 0.05), concordant with previous studies (18). In contrast, the Mycfl/fl; EpoR-Cre erythroid cell count did not increase with age. At E9.5, cell counts in Mycfl/fl; EpoR-Cre and normal embryos were not significantly different. However, E10.5 and E11.5 Mycfl/fl; EpoR-Cre embryos showed a progressive and significant reduction in cell counts, relative to control embryos (P < 0.002 and P < 0.01, respectively), consistent with their severely anemic appearance. The low circulating erythroid cell counts in Mycfl/fl; EpoR-Cre embryos at E11.5 are consistent with a potential role for Myc in the expansion and/or maturation of the circulating primitive erythroid cell population.

Table 3.

Circulating erythroid cell counts in control and Mycfl/fl; EpoR-Cre embryos

Embryonic stage n Control (104 cells) n Mycfl/fl; EpoR-Cre (104 cells)
E9.5 6 1.7 ± 0.4a 2 2.1 ± 0.4a
E10.5 6 24.6 ± 3.9 4 1.9 ± 1.4b
E11.5 9 99.5 ± 22.8 5 2.0 ± 0.0c
a

The values shown are mean numbers of circulating erythroid cells ± the standard error of the mean.

b

P < 0.002 (in comparison with controls).

c

P < 0.01 (in comparison with controls).

Cytospin preparations of embryonic peripheral blood at E10.5 and E11.5 were stained with Giemsa dye (Fig. 7A), and erythroid cell maturation was monitored. The circulating erythroid cells from controls were mainly basophilic erythroblasts at E10.5 and E11.5. Interestingly, blood from Mycfl/fl; EpoR-Cre embryos from the same litters displayed a change in Giemsa reactivity and a more mature phenotype at both E10.5 and E11.5, resembling E13.5 orthochromatophilic erythroblasts (27). The E10.5 and E11.5 Mycfl/fl; EpoR-Cre cells have more condensed nuclei, a lower nucleus-to-cytoplasm ratio, and a higher-than-normal proportion of cells with nuclei at the periphery (Fig. 7B). In contrast to KLF1−/− KLF2−/− cells (5), there is no evidence of cytoplasmic blebbing or cell shape anomalies in Mycfl/fl; EpoR-Cre circulating blood cells, suggesting that this phenotype is controlled not by the gene for Myc but rather by other genes regulated by KLF1 and KLF2. Nevertheless, the finding that Mycfl/fl; EpoR-Cre embryos have more mature cells suggests that the absence of Myc in the late stages of primitive erythropoiesis promotes earlier erythroid cell maturation than normally observed in development.

Fig 7.

Fig 7

(A) Cytological analysis of circulating primitive erythroid cells. Giemsa-stained cytospin preparations of peripheral blood from E10.5 and E11.5 control (left) and Mycfl/fl; EpoR-Cre (right) embryos. Primitive blood cell preparations of the control embryos at E10.5 and E11.5 exhibited numerous large erythroid cells with features characteristic of basophilic erythroblasts. In contrast, Mycfl/fl; EpoR-Cre embryos had altered Giemsa reactivity and a more orthochromatophilic erythroblast appearance. The Mycfl/fl; EpoR-Cre cells also displayed more condensed and eccentric nuclei in comparison to those of controls, indicating that the Mycfl/fl; EpoR-Cre cells were more mature. Images were obtained with a Zeiss Axiophot microscope. Magnification, ×64. (B) Nuclear-to-cell morphometric analysis indicates a more mature erythroid cell profile in Mycfl/fl; EpoR-Cre embryos. Histograms of ratios of nuclear to cell surface areas of circulating erythroid cells from WT controls (n = 3) and Mycfl/fl; EpoR-Cre embryos (n = 3) at E10.5 and E11.5 were obtained for a minimum of 103 primitive erythroid cells (range, 103 to 203). Importantly, the ratio of nuclear to cell surface areas in Mycfl/fl; EpoR-Cre embryos was more than 2-fold lower than that of controls at E10.5 and E11.5 (****; P < 0.0001). In controls, the ratio of nucleus to cell surface area decreased significantly between E10.5 to E11.5 (P < 0.006).

To evaluate the role of Myc in the primitive erythroid cell developmental program, we monitored the mRNA encoding the erythroid cell-specific membrane marker GPA in the peripheral blood of E9.5 to E11.5 controls and Mycfl/fl; EpoR-Cre embryos (Fig. 8A). Controls have an approximately 3- and 5-fold increased GPA/S16 ratio at E10.5 and E11.5 compared to that at E9.5, consistent with a previous report (37). Strikingly, Mycfl/fl; EpoR-Cre embryos exhibited similar GPA/S16 ratios at E10.5 and E11.5, compared to E9.5. Hence Mycfl/fl; EpoR-Cre E9.5 to E11.5 circulating erythroid cells maintain stationary GPA mRNA amounts and an unchanged cell count, whereas normal embryos show increases in both parameters.

Fig 8.

Fig 8

(A) Quantitative analyses of mRNA expression in Mycfl/fl; EpoR-Cre and control embryos. Shown is the erythroid-cell-specific GPA expression detected by real-time PCR and normalized to S16 from peripheral blood of Mycfl/fl; EpoR-Cre and control mice at E9.5, E10.5, and E11.5. GPA transcript levels increased significantly in controls, whereas in Mycfl/fl; EpoR-Cre embryos, the levels were virtually unchanged from E9.5. (B) Quantification of embryonic (Ey, βh1, ζ) and adult β-globin expression. qRT-PCR was performed with peripheral blood at E9.5 and E11.5 and normalized to S16. A significant increase in globin gene expression was observed in control embryos from E9.5 to E11.5, while in Mycfl/fl; EpoR-Cre embryos, the expression of none of these genes was increased and expression at E11.5 was comparable to that at E9.5. Data are presented as means ± standard deviations (n ≥ 6). Asterisks indicate significant difference from the control. *, P < 0.02; **, P < 0.01; ***, P < 0.0001.

To determine whether Myc deficiency could be dysregulating endogenous embryonic α-like and β-like globin gene expression, real-time PCR was performed to quantify embryonic globin mRNA levels in E9.5 and E11.5 peripheral blood. Figure 8B shows that the expression of the embryonic Ey, βh1, and ζ-globin genes is similar in control and Mycfl/fl; EpoR-Cre embryos at E9.5. At E11.5, the expression of all three of these genes in controls increased by ∼8-, 3-, and 5-fold, respectively, consistent with proliferating and maturing circulating erythroid cells. In contrast, E11.5 Mycfl/fl; EpoR-Cre embryos had levels similar to those measured at E9.5, thereby exhibiting less globin expression than controls (P < 0.007 for each globin). Further, the adult murine β-globin gene was also assessed in Mycfl/fl; EpoR-Cre and control embryos. While the expression of the gene for β-globin was also significantly increased in controls between E9.5 and 11.5, levels were unchanged for Mycfl/fl; EpoR-Cre, leading to reduced expression in comparison to that in controls. In fact, the globin gene expression profiles of Mycfl/fl; EpoR-Cre appear almost identical between E9.5 and E11.5. Although the circulating primitive erythroid cells in Mycfl/fl; EpoR-Cre embryos display a developmental block in globin gene expression and switching, there is an apparent accelerated maturation, as well as impaired expansion, of the cell population and this provides a rationale for the severe anemia seen at E11.5.

DISCUSSION

To elucidate the complex network of genes controlled by KLF1 and KLF2 in the primitive erythropoietic program, a global transcriptional analysis, followed by functional characterization, was undertaken. The regulatory network that was uncovered reveals potential mechanisms controlling the processes of differentiation, expansion, and maturation of the primitive erythroid cell population. A pathway of major significance was discovered that involves KLF1, KLF2, and Myc. Our data indicate that KLF1 and KLF2 directly regulate the gene for Myc, which, in turn, controls some of the phenotypes observed in KLF1−/− KLF2−/− embryos. Importantly, cell-autonomous Myc ablation at the proerythroblast stage demonstrates that Myc is an essential contributor to the developmental program of primitive erythropoiesis.

A unique expression profiling approach comparing WT, KLF1−/−, and KLF1−/− KLF2−/− erythroid cells was devised to enhance the identification of key regulators in primitive erythropoiesis. A number of genes that are now recognized to be coordinately regulated by KLF1 and KLF2 in primitive erythroid cells (this report and reference 62), were also identified in expression profiling studies of KLF1−/− definitive erythroid cells (22, 34, 61, 73). These include Epb4.9 and Tfrc (22, 34, 73), CD24a antigen and Kcnn4 (22), microsomal glutathione S-transferase 3 (Mgst3), solute carrier family 25 member 27 (Slc25a37), and erythroblast membrane-associated protein (Ermap) (73). These targets represent a subclass of genes directly or indirectly controlled by KLF1 and KLF2 in primitive erythropoiesis and by KLF1 in definitive erythropoiesis. This correlates with the fact that KLF1 and KLF2 expression is similar in primitive erythroid cells, whereas KLF1 expression is upregulated in definitive erythroid cells (2). Relatively few genes were determined to be coordinately regulated by KLF1 and KLF2, and some of these are important red cell genes, like those for Tfrc, Pklr, and Myc. Of these genes, that for Myc is synergistically regulated by KLF1 and KLF2. In addition, Myc is central to the pathway of genes regulated by KLF1 and KLF2 and is a critical determinant of primitive erythropoiesis.

The transcription factor Myc plays an important role in development and hematopoiesis (19, 50). Misexpression of Myc is associated with various forms of human cancer, as well as other genetic diseases (43, 57, 74, 82). Numerous studies have implicated Myc in the regulation of stem cell self-renewal, lineage differentiation, and cell proliferation and growth (e.g., 13, 15, 24). These cellular processes/functions are ongoing during primitive erythropoiesis, consistent with the observed expression and involvement of Myc.

The role of Myc in erythropoiesis has previously been explored. Several reports examined the role of ectopic Myc overexpression and suggested, despite conflicting outcomes, that Myc alters the differentiation rate (1, 78), self-renewal potential (12, 78), cell cycle arrest, proliferation, and terminal maturation (1, 38, 67). Discrepancies between these studies may result from the use of different erythroid cell culture systems at various differentiation stages or from dose-dependent responses (38). The relevance of these results to the in vivo activity of Myc in primitive erythropoiesis has not been addressed.

Epiblast-restricted Myc conditional ablation leads to fetal demise at approximately E11.5, and the yolk sac blood islands appear to have a reduced number of red blood cells (23). However, it was not clear whether this is due to the absence of Myc in blood cells or in other cell types. Hematopoietic Myc conditional KO mice generated using Vav-iCre precluded analysis of primitive erythropoiesis because the Vav gene promoter is activated during definitive erythropoiesis (30). In the present study, we demonstrated that Myc has a cell-autonomous role in primitive erythroid cells in vivo by specifically ablating the Myc gene at the proerythroblast stage. These results also provide the first evidence that Myc is an essential regulator in the late stages of embryonic erythroid cell maturation. Indeed, the constant number of erythroblasts in Mycfl/fl; EpoR-Cre between E9.5 and E11.5 suggests that Myc plays a crucial role in controlling terminal cell division in circulating primitive erythroid cells. The finding that there are fewer circulating erythroid cells at E10.5 in both KLF1−/− KLF2−/− and Mycfl/fl; EpoR-Cre embryos is consistent with the observed hierarchical regulation of Myc by KLF1 and KLF2. In the context of the enormous growth of the embryo between E9.5 and E11.5, failure to expand the synchronous circulating primitive erythroid cell population likely leads to embryonic death in both Mycfl/fl; EpoR-Cre and KLF1−/− KLF2−/− mice. In addition, Mycfl/fl; EpoR-Cre circulating primitive erythroid cells resemble orthochromatophilic erythroblasts of normal morphology, with nuclear condensation and asymmetric nuclear localization. This accelerated erythroid cell maturation suggests that upon cell cycle arrest, the onset of terminal maturation is triggered in circulating Mycfl/fl; EpoR-Cre erythroid cells. This correlates with the reported requirement of Myc downregulation for terminal maturation in definitive erythroid cells (38).

Mycfl/fl; EpoR-Cre circulating cells fail to upregulate the embryonic globin genes between E9.5 and E11.5, even though they appear to mature morphologically, and it is likely that this contributes to the paler appearance of embryos. In addition, the circulating Mycfl/fl; EpoR-Cre cells do not initiate the embryonic-to-adult switch normally detected by E11.5. Consequently, the absence of Myc in primitive proerythroblasts appears to abrogate not only the developmental program but also indirectly the globin developmental switching pattern. Taken together, our results indicate that the absence of KLF1 and KLF2 in KO mice leads to downregulation of Myc, and this, in turn, causes a reduced number, an abnormal developmental program, and accelerated maturation of primitive erythroid cells after E9.5.

Pathway analysis mapped Myc to a nexus in the KLF1 and KLF2 regulatory network in embryonic erythroid cells. Interestingly, Myc had previously been shown to regulate genes important in red cells, such as the genes for the Tfrc, deoxycytidine kinase (dck), nucleolar protein 5A, Mgst3, and Foxm1 (26, 53). These genes are also coordinately regulated by KLF1 and KLF2 in primitive erythroid cells. The similarity in gene targets identified for KLF1 and KLF2 in embryonic erythropoiesis, and for KLF1 in definitive erythropoiesis (60, 61, 73), suggests a potential in vivo functional role for Myc in definitive cells. Significantly, the more severe erythroid cell abnormalities observed in KLF1−/− KLF2−/− compared to Mycfl/fl; EpoR-Cre primitive erythroid cells likely stem from the dysregulation of genes other than that for Myc. Nevertheless, the overlap in gene targets is consistent with the hierarchical role of KLF1 and KLF2 in regulating Myc in embryonic erythropoiesis.

Our interaction network model of primitive erythropoiesis has at its core the coordinate roles of KLF1 and KLF2 in the direct regulation of the Myc gene. An important subset of genes in the broader network could be indirectly controlled by KLF1 and KLF2 but directly regulated by Myc. KLF1 and KLF2, as well as Myc, have roles in the expansion and maturation of the erythroid cell population. While KLF1 and KLF2 have been shown to directly stimulate globin gene expression in primitive erythroid cells, Myc may potentially influence hemoglobin production indirectly, by modulating heme synthesis or iron-controlling genes (54, 79). KLF1, KLF2, and Myc apparently regulate genes controlling cytoskeleton and membrane formation, and this could be directly or indirectly required for the expansion of primitive erythroid cells. These experiments integrate, for the first time, the roles of KLF1, KLF2, and Myc in driving primitive erythropoiesis.

In summary, we have defined a regulatory network controlled by KLF1 and KLF2 in primitive erythroid cells. The Myc gene is a direct target that is synergistically regulated by KLF1 and KLF2. Myc is an essential regulator in primitive erythroid cells and controls the expansion of the circulating erythroid cell population, the developmental program, and the kinetics of erythroid cell maturation. The identification of the KLF1, KLF2, and Myc network provides mechanistic insights into the dynamic sequential events that control the developmental program of primitive erythropoiesis.

ACKNOWLEDGMENTS

We thank Jerry B Lingrel for his kind gift of the KLF2 KO mice and James Bieker for the KLF1 cDNA expression construct. We are grateful to Gordon Ginder and Stephen Sawyer for ongoing discussions and critical advice.

This work was supported by NIH U54 HL090516 to J.A.L., NIH F31 DK072548 to L.C.R., NIH VCU Massey Cancer Center support grant P30 CA016059, a Canadian Blood Services/CIHR fellowship to W.L., and Canadian Blood Services/CIHR grant award 210399 to M.T.

Footnotes

Published ahead of print 7 May 2012

REFERENCES

  • 1. Acosta JC, et al. 2008. Myc inhibits p27-induced erythroid differentiation of leukemia cells by repressing erythroid master genes without reversing p27-mediated cell cycle arrest. Mol. Cell. Biol. 28:7286–7295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Alhashem YN, et al. 2011. Transcription factors KLF1 and KLF2 positively regulate embryonic and fetal beta-globin genes through direct promoter binding. J. Biol. Chem. 286:24819–24827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Arnaud L, et al. 2010. A dominant mutation in the gene encoding the erythroid transcription factor KLF1 causes a congenital dyserythropoietic anemia. Am. J. Hum. Genet. 87:721–727 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Baron MH. 2003. Embryonic origins of mammalian hematopoiesis. Exp. Hematol. 31:1160–1169 [DOI] [PubMed] [Google Scholar]
  • 5. Basu P, et al. 2007. EKLF and KLF2 have compensatory roles in embryonic {beta}-globin gene expression and primitive erythropoiesis. Blood 110:3417–3425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Basu P, et al. 2005. KLF2 is essential for primitive erythropoiesis and regulates the human and murine embryonic beta-like globin genes. Blood 106:2566–2571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Basu P, et al. 2004. Evolutionary conservation of KLF transcription factors and functional conservation of human gamma-globin gene regulation in chicken. Genomics 84:311–319 [DOI] [PubMed] [Google Scholar]
  • 8. Beauchemin H, Trudel M. 2009. Evidence for a bigenic chromatin subdomain in regulation of the fetal-to-adult hemoglobin switch. Mol. Cell. Biol. 29:1635–1648 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bieker JJ. 2001. Krüppel-like factors: three fingers in many pies. J. Biol. Chem. 276:34355–34358 [DOI] [PubMed] [Google Scholar]
  • 10. Borg J, et al. 2010. Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin. Nat. Genet. 42:801–805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Coghill E, et al. 2001. Erythroid Krüppel-like factor (EKLF) coordinates erythroid cell proliferation and hemoglobinization in cell lines derived from EKLF null mice. Blood 97:1861–1868 [DOI] [PubMed] [Google Scholar]
  • 12. Cory S, Maekawa T, McNeall J, Metcalf D. 1991. Murine erythroid cell lines derived with c-myc retroviruses respond to leukemia-inhibitory factor, erythropoietin, and interleukin 3. Cell Growth Differ. 2:165–172 [PubMed] [Google Scholar]
  • 13. Couillard M, Trudel M. 2009. C-myc as a modulator of renal stem/progenitor cell population. Dev. Dyn. 238:405–414 [DOI] [PubMed] [Google Scholar]
  • 14. Crossley M, et al. 1996. Isolation and characterization of the cDNA encoding BKLF/TEF-2, a major CACCC-box-binding protein in erythroid cells and selected other cells. Mol. Cell. Biol. 16:1695–1705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Dang CV. 1999. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol. Cell. Biol. 19:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Davis AC, Wims M, Spotts GD, Hann SR, Bradley A. 1993. A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev. 7:671–682 [DOI] [PubMed] [Google Scholar]
  • 17. de Alboran I, et al. 2001. Analysis of C-MYC function in normal cells via conditional gene-targeted mutation. Immunity 14:45–55 [DOI] [PubMed] [Google Scholar]
  • 18. De la Chapelle A, Fantoni A, Marks PA. 1969. Differentiation of mammalian somatic cells: DNA and hemoglobin synthesis in fetal mouse yolk sac erythroid cells. Proc. Natl. Acad. Sci. U. S. A. 63:812–819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Delgado MD, Leon J. 2010. Myc roles in hematopoiesis and leukemia. Genes Cancer 1:605–616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Donze D, Townes TM, Bieker JJ. 1995. Role of erythroid Krüppel-like factor in human gamma- to beta-globin gene switching. J. Biol. Chem. 270:1955–1959 [DOI] [PubMed] [Google Scholar]
  • 21. Drissen R, et al. 2004. The active spatial organization of the beta-globin locus requires the transcription factor EKLF. Genes Dev. 18:2485–2490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Drissen R, von Lindern M, et al. 2005. The erythroid phenotype of EKLF-null mice: defects in hemoglobin metabolism and membrane stability. Mol. Cell. Biol. 25:5205–5214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Dubois NC, et al. 2008. Placental rescue reveals a sole requirement for c-Myc in embryonic erythroblast survival and hematopoietic stem cell function. Development 135:2455–2465 [DOI] [PubMed] [Google Scholar]
  • 24. Eilers M, Eisenman RN. 2008. Myc's broad reach. Genes Dev. 22:2755–2766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Feng WC, Southwood CM, Bieker JJ. 1994. Analyses of beta-thalassemia mutant DNA interactions with erythroid Krüppel-like factor (EKLF), an erythroid cell-specific transcription factor. J. Biol. Chem. 269:1493–1500 [PubMed] [Google Scholar]
  • 26. Fernandez PC, et al. 2003. Genomic targets of the human c-Myc protein. Genes Dev. 17:1115–1129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Fraser ST, Isern J, Baron MH. 2007. Maturation and enucleation of primitive erythroblasts during mouse embryogenesis is accompanied by changes in cell-surface antigen expression. Blood 109:343–352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH. 1996. Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1. Proc. Natl. Acad. Sci. U. S. A. 93:12355–12358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Gentleman RC, et al. 2004. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5:R80 doi:10.1186/gb-2004-5-10-r80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. He C, et al. 2008. c-myc in the hematopoietic lineage is crucial for its angiogenic function in the mouse embryo. Development 135:2467–2477 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. He TC, Chan TA, Vogelstein B, Kinzler KW. 1999. PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell 99:335–345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. He TC, et al. 1998. Identification of c-MYC as a target of the APC pathway. Science 281:1509–1512 [DOI] [PubMed] [Google Scholar]
  • 33. Heinrich AC, Pelanda R, Klingmuller U. 2004. A mouse model for visualization and conditional mutations in the erythroid lineage. Blood 104:659–666 [DOI] [PubMed] [Google Scholar]
  • 34. Hodge D, et al. 2006. A global role for EKLF in definitive and primitive erythropoiesis. Blood 107:3359–3370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Hollander M, Wolfe DA. 1999. Nonparametric statistical methods, 2nd edition Wiley-Interscience, New York, NY [Google Scholar]
  • 36. Irizarry RA, et al. 2003. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 31:e15 doi:10.1093/nar/gng015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Isern J, et al. 2011. Single-lineage transcriptome analysis reveals key regulatory pathways in primitive erythroid progenitors in the mouse embryo. Blood 117:4924–4934 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Jayapal SR, et al. 2010. Down-regulation of Myc is essential for terminal erythroid maturation. J. Biol. Chem. 285:40252–40265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Jiang J, et al. 2008. A core Klf circuitry regulates self-renewal of embryonic stem cells. Nat. Cell Biol. 10:353–360 [DOI] [PubMed] [Google Scholar]
  • 40. Kingsley PD, et al. 2006. “Maturational” globin switching in primary primitive erythroid cells. Blood 107:1665–1672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kingsley PD, Malik J, Fantauzzo KA, Palis J. 2004. Yolk sac-derived primitive erythroblasts enucleate during mammalian embryogenesis. Blood 104:19–25 [DOI] [PubMed] [Google Scholar]
  • 42. Kuo CT, et al. 1997. The LKLF transcription factor is required for normal tunica media formation and blood vessel stabilization during murine embryogenesis. Genes Dev. 11:2996–3006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Lanoix J, D'Agati V, Szabolcs M, Trudel M. 1996. Dysregulation of cellular proliferation and apoptosis mediates human autosomal dominant polycystic kidney disease (ADPKD). Oncogene 13:1153–1160 [PubMed] [Google Scholar]
  • 44. Maetens M, et al. 2007. Distinct roles of Mdm2 and Mdm4 in red cell production. Blood 109:2630–2633 [DOI] [PubMed] [Google Scholar]
  • 45. Martin R, et al. 2004. SCL interacts with VEGF to suppress apoptosis at the onset of hematopoiesis. Development 131:693–702 [DOI] [PubMed] [Google Scholar]
  • 46. Matsumoto N, et al. 2006. Developmental regulation of yolk sac hematopoiesis by Krüppel-like factor 6. Blood 107:1357–1365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. McConnell BB, Yang VW. 2010. Mammalian Krüppel-like factors in health and diseases. Physiol. Rev. 90:1337–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. McGrath KE, Koniski AD, Malik J, Palis J. 2003. Circulation is established in a stepwise pattern in the mammalian embryo. Blood 101:1669–1676 [DOI] [PubMed] [Google Scholar]
  • 49. Miller IJ, Bieker JJ. 1993. A novel, erythroid cell-specific murine transcription factor that binds to the CACCC element and is related to the Krüppel family of nuclear proteins. Mol. Cell. Biol. 13:2776–2786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Murphy MJ, Wilson A, Trumpp A. 2005. More than just proliferation: Myc function in stem cells. Trends Cell Biol. 15:128–137 [DOI] [PubMed] [Google Scholar]
  • 51. Nilson DG, Sabatino DE, Bodine DM, Gallagher PG. 2006. Major erythrocyte membrane protein genes in EKLF-deficient mice. Exp. Hematol. 34:705–712 [DOI] [PubMed] [Google Scholar]
  • 52. Nuez B, Michalovich D, Bygrave A, Ploemacher R, Grosveld F. 1995. Defective haematopoiesis in fetal liver resulting from inactivation of the EKLF gene. Nature 375:316–318 [DOI] [PubMed] [Google Scholar]
  • 53. Obaya AJ, Kotenko I, Cole MD, Sedivy JM. 2002. The proto-oncogene c-myc acts through the cyclin-dependent kinase (Cdk) inhibitor p27(Kip1) to facilitate the activation of Cdk4/6 and early G(1) phase progression. J. Biol. Chem. 277:31263–31269 [DOI] [PubMed] [Google Scholar]
  • 54. O'Donnell KA, et al. 2006. Activation of transferrin receptor 1 by c-Myc enhances cellular proliferation and tumorigenesis. Mol. Cell. Biol. 26:2373–2386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Ovcharenko I, et al. 2005. Mulan: multiple-sequence local alignment and visualization for studying function and evolution. Genome Res. 15:184–194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ovcharenko I, Nobrega MA, Loots GG, Stubbs L. 2004. ECR Browser: a tool for visualizing and accessing data from comparisons of multiple vertebrate genomes. Nucleic Acids Res. 32:W280–W286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Pelengaris S, Khan M. 2003. The many faces of c-MYC. Arch. Biochem. Biophys. 416:129–136 [DOI] [PubMed] [Google Scholar]
  • 58. Perkins AC, Sharpe AH, Orkin SH. 1995. Lethal beta-thalassaemia in mice lacking the erythroid CACCC-transcription factor EKLF. Nature 375:318–322 [DOI] [PubMed] [Google Scholar]
  • 59. Philipsen S, Suske G. 1999. A tale of three fingers: the family of mammalian Sp/XKLF transcription factors. Nucleic Acids Res. 27:2991–3000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Pilon AM, et al. 2011. Genome-wide ChIP-Seq reveals a dramatic shift in the binding of the transcription factor erythroid Krüppel-like factor during erythrocyte differentiation. Blood 118:e139–e148 doi:10.1182/blood-2011-05-355107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Pilon AM, et al. 2008. Failure of terminal erythroid differentiation in EKLF-deficient mice is associated with cell cycle perturbation and reduced expression of E2F2. Mol. Cell. Biol. 28:7394–7401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Redmond LC, et al. 2011. Krüppel-like factor 2 regulated gene expression in mouse embryonic yolk sac erythroid cells. Blood Cells Mol. Dis. 47:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Redmond LC, Dumur CI, Archer KJ, Haar JL, Lloyd JA. 2008. Identification of erythroid-enriched gene expression in the mouse embryonic yolk sac using microdissected cells. Dev. Dyn. 237:436–446 [DOI] [PubMed] [Google Scholar]
  • 64. Redmond LC, et al. 2006. Isolation of erythroid cells from the mouse embryonic yolk sac by laser capture microdissection and subsequent microarray hybridization. Blood Cells Mol. Dis. 37:27–32. [DOI] [PubMed] [Google Scholar]
  • 65. Redmond LC, Pang CJ, Dumur CI, Haar JL, Lloyd JA. Laser capture microdissection of embryonic cells and preparation of RNA for microarray assays. Methods Mol. Biol., in press [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Riggs KJ, et al. 1993. Yin-yang 1 activates the c-myc promoter. Mol. Cell. Biol. 13:7487–7495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Rylski M, et al. 2003. GATA-1-mediated proliferation arrest during erythroid maturation. Mol. Cell. Biol. 23:5031–5042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Saleque S, Cameron S, Orkin SH. 2002. The zinc-finger proto-oncogene Gfi-1b is essential for development of the erythroid and megakaryocytic lineages. Genes Dev. 16:301–306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Sánchez MJ, Bockamp EO, Miller J, Gambardella L, Green AR. 2001. Selective rescue of early haematopoietic progenitors in Scl(−/−) mice by expressing Scl under the control of a stem cell enhancer. Development 128:4815–4827 [DOI] [PubMed] [Google Scholar]
  • 70. Schoenfelder S, et al. 2010. Preferential associations between co-regulated genes reveal a transcriptional interactome in erythroid cells. Nat. Genet. 42:53–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Smyth GK. 12 February 2004. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3:Article 3. doi:10.2202/1544-6115.1027. [DOI] [PubMed] [Google Scholar]
  • 72. Tallack MR, Keys JR, Humbert PO, Perkins AC. 2009. EKLF/KLF1 controls cell cycle entry via direct regulation of E2f2. J. Biol. Chem. 284:20966–20974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Tallack MR, et al. 2010. A global role for KLF1 in erythropoiesis revealed by ChIP-seq in primary erythroid cells. Genome Res. 20:1052–1063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Thivierge C, et al. 2006. Overexpression of PKD1 causes polycystic kidney disease. Mol. Cell. Biol. 26:1538–1548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Tsai FY, et al. 1994. An early haematopoietic defect in mice lacking the transcription factor GATA-2. Nature 371:221–226 [DOI] [PubMed] [Google Scholar]
  • 76. Wani MA, Means RTJ, Lingrel JB. 1998. Loss of LKLF function results in embryonic lethality in mice. Transgenic Res. 7:229–238 [DOI] [PubMed] [Google Scholar]
  • 77. Warren AJ, et al. 1994. The oncogenic cysteine-rich LIM domain protein rbtn2 is essential for erythroid development. Cell 78:45–57 [DOI] [PubMed] [Google Scholar]
  • 78. Wilson A, et al. 2004. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev. 18:2747–2763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Wu KJ, Polack A, Della-Favera R. 1999. Coordinated regulation of iron-controlling genes, H-ferritin and IRP2, by c-MYC. Science 283:676–679 [DOI] [PubMed] [Google Scholar]
  • 80. Zhang P, et al. 2005. A functional screen for Krüppel-like factors that regulate the human gamma-globin gene through the CACCC promoter element. Blood Cells Mol. Dis. 35:227–235 [DOI] [PubMed] [Google Scholar]
  • 81. Zhou D, Liu K, Sun CW, Pawlik KM, Townes TM. 2010. KLF1 regulates BCL11A expression and gamma- to beta-globin gene switching. Nat. Genet. 42:742–744 [DOI] [PubMed] [Google Scholar]
  • 82. Zimmerman KA, et al. 1986. Differential expression of myc family genes during murine development. Nature 319:780–783 [DOI] [PubMed] [Google Scholar]

Articles from Molecular and Cellular Biology are provided here courtesy of Taylor & Francis

RESOURCES