Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin

Joseph Borg, Petros Papadopoulos, Marianthi Georgitsi, Laura Gutiérrez, Godfrey Grech, Pavlos Fanis, Marios Phylactides, Annemieke J M H Verkerk, Peter J van der Spek, Christian A Scerri, Wilhelmina Cassar, Ruth Galdies, Wilfred van Ijcken, Zeliha Ozgür, Nynke Gillemans, Jun Hou, Marisa Bugeja, Frank G Grosveld, Marieke von Lindern, Alex E Felice, George P Patrinos, Sjaak Philipsen, Joseph Borg, Petros Papadopoulos, Marianthi Georgitsi, Laura Gutiérrez, Godfrey Grech, Pavlos Fanis, Marios Phylactides, Annemieke J M H Verkerk, Peter J van der Spek, Christian A Scerri, Wilhelmina Cassar, Ruth Galdies, Wilfred van Ijcken, Zeliha Ozgür, Nynke Gillemans, Jun Hou, Marisa Bugeja, Frank G Grosveld, Marieke von Lindern, Alex E Felice, George P Patrinos, Sjaak Philipsen

Abstract

Hereditary persistence of fetal hemoglobin (HPFH) is characterized by persistent high levels of fetal hemoglobin (HbF) in adults. Several contributory factors, both genetic and environmental, have been identified but others remain elusive. HPFH was found in 10 of 27 members from a Maltese family. We used a genome-wide SNP scan followed by linkage analysis to identify a candidate region on chromosome 19p13.12-13. Sequencing revealed a nonsense mutation in the KLF1 gene, p.K288X, which ablated the DNA-binding domain of this key erythroid transcriptional regulator. Only family members with HPFH were heterozygous carriers of this mutation. Expression profiling on primary erythroid progenitors showed that KLF1 target genes were downregulated in samples from individuals with HPFH. Functional assays suggested that, in addition to its established role in regulating adult globin expression, KLF1 is a key activator of the BCL11A gene, which encodes a suppressor of HbF expression. These observations provide a rationale for the effects of KLF1 haploinsufficiency on HbF levels.

Conflict of interest statement

Declaration of competing financial interests

The authors declare no competing financial interests.

Figures

Figure 1. Chromosome 19 locus linked to…
Figure 1. Chromosome 19 locus linked to HPFH in a Maltese family
a) The Maltese HPFH pedigree. HbF levels are indicated as percentage of total Hb (%HbF). HPFH individuals are shown as half-filled symbols. b) LOD scores derived from genome-wide linkage analysis. The putative HPFH locus on chr. 19 is indicated by an arrow. pLOD = parametric LOD score; MPT = multi point test; cM = centiMorgan. c) Sequence analysis of KLF1. HPFH individuals were heterozygous for two mutations (arrows; Supplementary Table 1). The predicted effects of the mutations on KLF1 protein are shown below.
Figure 2. KLF1 target genes are downregulated…
Figure 2. KLF1 target genes are downregulated in wt/KLF1 p.K288X HEPs
a) RNA isolated from HEPs derived from normal (wt/wt) and HPFH (wt/KLF1 p.K288X) family members was used for genome-wide expression analysis. Deregulated genes common between wt/wt and wt/KLF1 p.K288X and mouse wt/wt versus Klf1 null mutant erythroid progenitors (Supplementary Table 2) were used for cluster analysis. b) Validation of key target genes by qPCR. Expression levels of BCL11A were normalized using GAPDH as a reference. Expression levels of HBG1/HBG2 (HBG) were calculated as ratio to HBA1/HBA2 (HBA) expression. Medians are indicated by red lines in the box plots. Asterisk: p=0.0209.
Figure 3. Increased HBG1/HBG2 expression after knockdown…
Figure 3. Increased HBG1/HBG2 expression after knockdown of KLF1 in normal HEPs
a) HEPs derived from normal donors were transduced with shRNA-expressing lentiviruses. Cells were harvested five days after transduction, and nuclear extracts prepared. Top panels: KLF1 protein expression assessed by Western blot analysis. Bottom panels: BCL11A protein levels were reduced upon KLF1 knockdown. NPM1 served as a loading control. None = mock transduction; TRC = control non-specific shRNA; sh1 and sh2 = two independent shRNAs targeting KLF1. A non-specific band is indicated (AHSP is a known KLF1 target gene and serves as a positive control. Expression levels of AHSP and BCL11A were normalized using GAPDH as a reference. Expression levels of HBG were calculated as ratio to total β-like globin expression (HBG + HBB) expression. Medians are indicated by red lines. Circles: points outside the range of the error bars. Asterisk: p=0.020; double asterisks: p<0.003.
Figure 4. Expression of exogenous KLF1 in…
Figure 4. Expression of exogenous KLF1 in HPFH HEPs
a) HEPs derived from individual II-5 were transduced with lentiviral constructs expressing GFP, KLF1 truncated at amino acid 288 (TR) or full-length KLF1 (FL). Seven days after transduction, nuclear extracts were prepared and expression of BCL11A and KLF1 was assessed by Western blot. NPM1 served as a loading control. b) RNA was isolated from II-5 HEPs seven days after transduction with the indicated lentiviruses, and was used for quantitative S1 nuclease protection assays to measure globin mRNA expression. Arrows indicate protected fragments diagnostic for HBA1/HBA2 (HBA), HBG1/HBG2 (HBG) and HBB mRNAs. c) Quantitation of data shown in b) by Phosphorimager analysis.
Figure 5. KLF1 binds to the promoter…
Figure 5. KLF1 binds to the promoter of the BCL11A gene in vivo
a) Schematic drawings of the promoter areas of the BCL11A,HBB and RASSF1A genes. Positions of potential KLF1 binding sites (CACC boxes) and PCR primers used are indicated. Arrows indicate transcription start sites. b) ChIP analysis of KLF1 binding to the BCL11A promoter in human fetal liver cells and adult HEPs. The HBB promoter served as a positive control .RASSF1A was used as a negative control, and the unrelated CD71 antibody served as a control for the specificity of the KLF1 antibody. Asterisk: p<0.05; double asterisks: p<0.01. Error bars: standard deviation.

References

    1. Serjeant G. Geographic heterogeneity of sickle cell disease. In: Steinberg MH, Forget BG, Higgs DR, Nagel RL, editors. Disorders of Hemoglobin. Cambridge University Press; Cambrige: 2001. pp. 895–906.
    1. Bieker JJ. Probing the onset and regulation of erythroid cell-specific gene expression. Mt Sinai J Med. 2005;72:333–8.
    1. Sankaran VG, et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322:1839–42.
    1. Stamatoyannopoulos G, Grosveld F. Hemoglobin switching. In: Stamatoyannopoulos G, Majerus PW, Perlmutter RM, Varmus H, editors. The Molecular Basis of Blood Diseases. WB Saunders Company; Philadelphia PA: 2001. pp. 135–182.
    1. Thein SL, Menzel S, Lathrop M, Garner C. Control of fetal hemoglobin: new insights emerging from genomics and clinical implications. Hum Mol Genet. 2009;18:R216–23.
    1. Craig JE, et al. Dissecting the loci controlling fetal haemoglobin production on chromosomes 11p and 6q by the regressive approach. Nat Genet. 1996;12:58–64.
    1. Gilman JG, Huisman TH. DNA sequence variation associated with elevated fetal G gamma globin production. Blood. 1985;66:783–7.
    1. Close J, et al. Genome annotation of a 1.5 Mb region of human chromosome 6q23 encompassing a quantitative trait locus for fetal hemoglobin expression in adults. BMC Genomics. 2004;5:33.
    1. Garner C, et al. Haplotype mapping of a major quantitative-trait locus for fetal hemoglobin production, on chromosome 6q23. Am J Hum Genet. 1998;62:1468–74.
    1. Lettre G, et al. DNA polymorphisms at the BCL11A, HBS1L-MYB, and beta-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease. Proc Natl Acad Sci U S A. 2008;105:11869–74.
    1. Menzel S, et al. A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15. Nat Genet. 2007;39:1197–9.
    1. Abecasis GR, Cherny SS, Cookson WO, Cardon LR. Merlin--rapid analysis of dense genetic maps using sparse gene flow trees. Nat Genet. 2002;30:97–101.
    1. Hoffmann K, Lindner TH. easyLINKAGE-Plus--automated linkage analyses using large-scale SNP data. Bioinformatics. 2005;21:3565–7.
    1. Leykin I, et al. Comparative linkage analysis and visualization of high-density oligonucleotide SNP array data. BMC Genet. 2005;6:7.
    1. Singleton BK, Burton NM, Green C, Brady RL, Anstee DJ. Mutations in EKLF/KLF1 form the molecular basis of the rare blood group In(Lu) phenotype. Blood. 2008;112:2081–8.
    1. Miller IJ, Bieker JJ. A novel, erythroid cell-specific murine transcription factor that binds to the CACCC element and is related to the Kruppel family of nuclear proteins. Mol Cell Biol. 1993;13:2776–86.
    1. Feng WC, Southwood CM, Bieker JJ. Analyses of beta-thalassemia mutant DNA interactions with erythroid Kruppel-like factor (EKLF), an erythroid cell-specific transcription factor. J Biol Chem. 1994;269:1493–500.
    1. Leberbauer C, et al. Different steroids co-regulate long-term expansion versus terminal differentiation in primary human erythroid progenitors. Blood. 2005;105:85–94.
    1. Pilon AM, et al. Failure of terminal erythroid differentiation in EKLF-deficient mice is associated with cell cycle perturbation and reduced expression of E2F2. Mol Cell Biol. 2008;28:7394–401.
    1. Moffat J, et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell. 2006;124:1283–98.
    1. Antoniou M. Induction of erythroid-specific expression in murine erythroleukemia (MEL) cell lines. Methods in Molecular Biology. 1991;7:421–434.
    1. Muhlemann O, Eberle AB, Stalder L, Zamudio Orozco R. Recognition and elimination of nonsense mRNA. Biochim Biophys Acta. 2008;1779:538–49.
    1. Donze D, Townes TM, Bieker JJ. Role of erythroid Kruppel-like factor in human gamma- to beta-globin gene switching. J Biol Chem. 1995;270:1955–9.
    1. Drissen R, et al. The active spatial organization of the beta-globin locus requires the transcription factor EKLF. Genes Dev. 2004;18:2485–90.
    1. Zhou D, Pawlik KM, Ren J, Sun CW, Townes TM. Differential binding of erythroid Krupple-like factor to embryonic/fetal globin gene promoters during development. J Biol Chem. 2006;281:16052–7.
    1. Bottardi S, Ross J, Pierre-Charles N, Blank V, Milot E. Lineage-specific activators affect beta-globin locus chromatin in multipotent hematopoietic progenitors. EMBO J. 2006;25:3586–95.
    1. Wijgerde M, et al. The role of EKLF in human beta-globin gene competition. Genes Dev. 1996;10:2894–902.
    1. Drissen R, et al. The erythroid phenotype of EKLF-null mice: defects in hemoglobin metabolism and membrane stability. Mol Cell Biol. 2005;25:5205–14.
    1. Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res. 1988;16:1215.
    1. Trifillis P, Ioannou P, Schwartz E, Surrey S. Identification of four novel delta-globin gene mutations in Greek Cypriots using polymerase chain reaction and automated fluorescence-based DNA sequence analysis. Blood. 1991;78:3298–305.
    1. Craig JE, Barnetson RA, Prior J, Raven JL, Thein SL. Rapid detection of deletions causing delta beta thalassemia and hereditary persistence of fetal hemoglobin by enzymatic amplification. Blood. 1994;83:1673–82.
    1. Higgs DR, Wood WG. Genetic complexity in sickle cell disease. Proc Natl Acad Sci U S A. 2008;105:11595–6.
    1. O'Connell JR, Weeks DE. PedCheck: a program for identification of genotype incompatibilities in linkage analysis. Am J Hum Genet. 1998;63:259–66.
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–8.
    1. Follenzi A, Sabatino G, Lombardo A, Boccaccio C, Naldini L. Efficient gene delivery and targeted expression to hepatocytes in vivo by improved lentiviral vectors. Hum Gene Ther. 2002;13:243–60.
    1. Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat Biotechnol. 1997;15:871–5.
    1. Andrews NC, Faller DV. A rapid micropreparation technique for extraction of DNA-binding proteins from limiting numbers of mammalian cells. Nucleic Acids Res. 1991;19:2499.
    1. Follows GA, et al. Epigenetic consequences of AML1-ETO action at the human c-FMS locus. EMBO J. 2003;22:2798–809.

Source: PubMed

3
Subscribe