The genome-wide molecular signature of transcription factors in leukemia

转录因子 运行x1 生物 表观遗传学 染色质 造血 抄写(语言学) GATA转录因子 遗传学 基因 细胞生物学 发起人 基因表达 计算生物学 干细胞 语言学 哲学
作者
Koen H M Prange,Abhishek A. Singh,Joost H.A. Martens
出处
期刊:Experimental Hematology [Elsevier]
卷期号:42 (8): 637-650 被引量:15
标识
DOI:10.1016/j.exphem.2014.04.012
摘要

Transcription factors control expression of genes essential for the normal functioning of the hematopoietic system and regulate development of distinct blood cell types. During leukemogenesis, aberrant regulation of transcription factors such as RUNX1, CBFβ, MLL, C/EBPα, SPI1, GATA, and TAL1 is central to the disease. Here, we will discuss the mechanisms of transcription factor deregulation in leukemia and how in recent years next-generation sequencing approaches have helped to elucidate the molecular role of many of these aberrantly expressed transcription factors. We will focus on the complexes in which these factors reside, the role of posttranslational modification of these factors, their involvement in setting up higher order chromatin structures, and their influence on the local epigenetic environment. We suggest that only comprehensive knowledge on all these aspects will increase our understanding of aberrant gene expression in leukemia as well as open new entry points for therapeutic intervention. Transcription factors control expression of genes essential for the normal functioning of the hematopoietic system and regulate development of distinct blood cell types. During leukemogenesis, aberrant regulation of transcription factors such as RUNX1, CBFβ, MLL, C/EBPα, SPI1, GATA, and TAL1 is central to the disease. Here, we will discuss the mechanisms of transcription factor deregulation in leukemia and how in recent years next-generation sequencing approaches have helped to elucidate the molecular role of many of these aberrantly expressed transcription factors. We will focus on the complexes in which these factors reside, the role of posttranslational modification of these factors, their involvement in setting up higher order chromatin structures, and their influence on the local epigenetic environment. We suggest that only comprehensive knowledge on all these aspects will increase our understanding of aberrant gene expression in leukemia as well as open new entry points for therapeutic intervention. In eukaryotes, chromatin structure and interaction between transcription factors (TFs) modulates transcription of genes. TFs initiate transcription and control transcriptional elongation and thereby influence gene expression programs and consequently cell state [1Adelman K. Lis J.T. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans.Nat Rev Genet. 2012; 13: 720-731Crossref PubMed Google Scholar, 2Bai X. Kim J. Yang Z. et al.TIF1gamma controls erythroid cell fate by regulating transcription elongation.Cell. 2010; 142: 133-143Abstract Full Text Full Text PDF PubMed Scopus (97) Google Scholar, 3Fuda N.J. Ardehali M.B. Lis J.T. Defining mechanisms that regulate RNA polymerase II transcription in vivo.Nature. 2009; 461: 186-192Crossref PubMed Scopus (224) Google Scholar, 4Lin C.Y. Lovén J. Rahl P.B. et al.Transcriptional amplification in tumor cells with elevated c-Myc.Cell. 2012; 151: 56-67Abstract Full Text Full Text PDF PubMed Scopus (276) Google Scholar, 5Rahl P.B. Lin C.Y. Seila A.C. et al.c-Myc regulates transcriptional pause release.Cell. 2010; 141: 432-445Abstract Full Text Full Text PDF PubMed Scopus (439) Google Scholar]. Through their DNA-binding domain all TFs expressed in a given cell type interact with specific regulatory elements. In this way, they exert control over spatial and temporal expression of genes through a variety of regulatory domains, consequently modulating all complex biological processes, such as cell differentiation, proliferation, and apoptosis [6Maston G.A. Evans S.K. Green M.R. Transcriptional regulatory elements in the human genome.Annu Rev Genomics Hum Genet. 2006; 7: 29-59Crossref PubMed Scopus (262) Google Scholar]. Approximately 10% of genes in the human genome are presumed to code for TFs [7Levine M. Tjian R. Transcription regulation and animal diversity.Nature. 2003; 424: 147-151Crossref PubMed Scopus (696) Google Scholar], with different sets of TFs expressed during different developmental stages. In the same cell type, different TFs can co-localize [8Chen X. Xu H. Yuan P. et al.Integration of external signaling pathways with the core transcriptional network in embryonic stem cells.Cell. 2008; 133: 1106-1117Abstract Full Text Full Text PDF PubMed Scopus (1156) Google Scholar, 9MacArthur S. Li X.-Y. Li J. et al.Developmental roles of 21 Drosophila transcription factors are determined by quantitative differences in binding to an overlapping set of thousands of genomic regions.Genome Biol. 2009; 10: R80Crossref PubMed Scopus (179) Google Scholar], and in different cell types and developmental stages these factors and complexes can bind in a different pattern [10Odom D.T. Zizlsperger N. Gordon D.B. et al.Control of pancreas and liver gene expression by HNF transcription factors.Science. 2004; 303: 1378-1381Crossref PubMed Scopus (762) Google Scholar, 11Lupien M. Eeckhoute J. Meyer C.A. et al.FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription.Cell. 2008; 132: 958-970Abstract Full Text Full Text PDF PubMed Scopus (443) Google Scholar, 12Sandmann T. Jensen L.J. Jakobsen J.S. et al.A temporal map of transcription factor activity: mef2 directly regulates target genes at all stages of muscle development.Dev Cell. 2006; 10: 797-807Abstract Full Text Full Text PDF PubMed Scopus (143) Google Scholar]. For example, in mouse macrophages and splenic B cells, PU.1 (SPI1) binds to a distinct set of cis regulatory elements, which in both cell types are also enriched for the motifs of a distinct group of lineage-specific TFs. Co-localization of different TFs—C/EBPα for macrophages and Oct2 for B cells—at cell-type specific PU.1 sites was subsequently confirmed with chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-seq) [13Heinz S. Benner C. Spann N. et al.Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities.Molecular Cell. 2010; 38: 576-589Abstract Full Text Full Text PDF PubMed Scopus (767) Google Scholar]. Because of the wide variety of TFs expressed in a given cell type, one main question is what the role of each individual TF is and how it functions in the context of the other DNA-binding proteins that are present. Studies in embryonic stem cells (ESCs) and in differentiated cells have suggested that the integrity of cell-type–specific gene expression programs is set up and maintained by only a few TFs (master regulators) [14Graf T. Historical origins of transdifferentiation and reprogramming.Cell Stem Cell. 2011; 9: 504-516Abstract Full Text Full Text PDF PubMed Scopus (66) Google Scholar, 15Ng H.-H. Surani M.A. The transcriptional and signalling networks of pluripotency.Nat Cell Biol. 2011; 13: 490-496Crossref PubMed Scopus (121) Google Scholar, 16Orkin S.H. Hochedlinger K. Chromatin connections to pluripotency and cellular reprogramming.Cell. 2011; 145: 835-850Abstract Full Text Full Text PDF PubMed Scopus (156) Google Scholar, 17Young R.A. Control of the embryonic stem cell state.Cell. 2011; 144: 940-954Abstract Full Text Full Text PDF PubMed Scopus (346) Google Scholar]. For example, it is believed that master TFs in ESCs, such as NANOG, OCT4, and SOX2, are expressed at high levels and exert autoregulation, properties shared with master TFs of other cell types [17Young R.A. Control of the embryonic stem cell state.Cell. 2011; 144: 940-954Abstract Full Text Full Text PDF PubMed Scopus (346) Google Scholar, 18Lee T.I. Young R.A. Transcriptional regulation and its misregulation in disease.Cell. 2013; 152: 1237-1251Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar]. Also hematopoiesis is classically thought to be governed by a small fraction of crucial TFs that are lineage specific and interact among themselves to control cell state [19Iwasaki H. Akashi K. Myeloid lineage commitment from the hematopoietic stem cell.Immunity. 2007; 26: 726-740Abstract Full Text Full Text PDF PubMed Scopus (171) Google Scholar], such as RUNX1, C/EBPα, and SPI1 in myeloid [20Ichikawa M. Asai T. Saito T. et al.AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis.Nat Med. 2004; 10: 299-304Crossref PubMed Scopus (338) Google Scholar, 21Tenen D.G. Hromas R. Licht J.D. Zhang D.E. Transcription factors, normal myeloid development, and leukemia.Blood. 1997; 90: 489-519Crossref PubMed Google Scholar, 22Vangala R.K. Heiss-Neumann M.S. Rangatia J.S. et al.The myeloid master regulator transcription factor PU.1 is inactivated by AML1-ETO in t(8;21) myeloid leukemia.Blood. Am Soc Hematol. 2003; 101: 270-277Google Scholar] and IKZF1, E2A, and EBF1 in B-lymphocyte development [23Tijchon E. Havinga J. van Leeuwen F.N. Scheijen B. B-lineage transcription factors and cooperating gene lesions required for leukemia development.Leukemia. 2013; 27: 541-552Crossref PubMed Scopus (16) Google Scholar]. However, it has also been suggested that a large number of TFs constitute a complex multi-tier gene regulatory network to control hematopoiesis [24Müller F.-J. Laurent L.C. Kostka D. et al.Regulatory networks define phenotypic classes of human stem cell lines.Nature. 2008; 455: 401-405Crossref PubMed Scopus (193) Google Scholar, 25Amit I. Garber M. Chevrier N. et al.Unbiased reconstruction of a mammalian transcriptional network mediating pathogen responses.Science. 2009; 326: 257-263Crossref PubMed Scopus (236) Google Scholar, 26Suzuki H. Forrest A.R.R. van Nimwegen E. et al.The transcriptional network that controls growth arrest and differentiation in a human myeloid leukemia cell line.Nat Genet. 2009; 41: 553-562Crossref PubMed Scopus (229) Google Scholar, 27Novershtern N. Subramanian A. Lawton L.N. et al.Densely interconnected transcriptional circuits control cell states in human hematopoiesis.Cell. 2011; 144: 296-309Abstract Full Text Full Text PDF PubMed Scopus (232) Google Scholar]. These two theories have reopened the debate on the coordinated mechanistic role of TFs in regulating hematopoiesis. Is hematopoiesis indeed modulated by a small set of master regulators or dependent on interactions between several different sets of TFs? Also, how is transcription regulated by differential usage of TF modules and how are other molecular constituents involved? It has become clear that although TFs are key proteins in regulating gene expression, it is further governed through synergistic contributions from several entities, including interaction between the TFs, post-transcriptional modifications, and the structure of the chromatin template (Fig. 1) [18Lee T.I. Young R.A. Transcriptional regulation and its misregulation in disease.Cell. 2013; 152: 1237-1251Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar]. For example, chromatin regulators, such as complexes of the SWI/SNF family and histone-modifying enzymes, assist in mobilizing and modifying nucleosomes during gene transcription and silencing [18Lee T.I. Young R.A. Transcriptional regulation and its misregulation in disease.Cell. 2013; 152: 1237-1251Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar, 28Clapier C.R. Cairns B.R. The biology of chromatin remodeling complexes.Annu Rev Biochem. 2009; 78: 273-304Crossref PubMed Scopus (676) Google Scholar, 29Hargreaves D.C. Crabtree G.R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms.Cell Res. 2011; 21: 396-420Crossref PubMed Scopus (254) Google Scholar]. Indeed in several diseases and syndromes, including distinct classes of leukemias, regulatory elements, chromatin regulators, and noncoding RNAs can be mutated [18Lee T.I. Young R.A. Transcriptional regulation and its misregulation in disease.Cell. 2013; 152: 1237-1251Abstract Full Text Full Text PDF PubMed Scopus (85) Google Scholar], suggesting that knowledge of the context in which TFs operate is imperative in understanding the molecular mechanisms governing gene expression in physiologic and diseased states. The last few decades have witnessed enormous advancements in DNA-sequencing technology, from Sanger sequencing to its automation and finally the advent of next-generation sequencing (NGS) methods. Especially NGS has led to the identification of many new regulatory sequences and insight into transcriptional machinery and chromatin regulators and has provided novel insights into gene regulatory mechanisms in normal but also diseased cells. Application of different NGS methods has made great contributions to the field of hematology. For example whole-genome sequencing (WGS) for detection of single nucleotide variants (SNV), insertions, deletions, and copy number variations has facilitated the sequencing of the first cancer genome [30Ley T.J. Mardis E.R. Ding L. et al.DNA sequencing of a cytogenetically normal acute myeloid leukaemia genome.Nature. 2008; 456: 66-72Crossref PubMed Scopus (670) Google Scholar] and identification of mutations in several types of leukemia [31Puente X.S. Pinyol M. Quesada V. et al.Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia.Nature. 2011; 475: 101-105Crossref PubMed Scopus (526) Google Scholar]. Mate-pair sequencing has allowed simultaneous detection of mutations, copy number variations, and structural rearrangements and contributed to the unfolding of molecular mechanisms in leukemia [32Buijs A. Poot M. van der Crabben S. et al.Elucidation of a novel pathogenomic mechanism using genome-wide long mate-pair sequencing of a congenital t(16;21) in a series of three RUNX1-mutated FPD/AML pedigrees.Leukemia. 2012; 26: 2151-2154Crossref PubMed Scopus (7) Google Scholar]. Whole-exome sequencing (WES) for studying exons and untranslated regions have assisted with the identification of mutations—including potential driver mutations—in several leukemias [33Kim Y. Schulz V.P. Satake N. et al.Whole-exome sequencing identifies a novel somatic mutation in MMP8 associated with a t(1;22)-acute megakaryoblastic leukemia.Leukemia. 2014; 28: 945-948Crossref PubMed Scopus (1) Google Scholar, 34Cancer Genome Atlas Research NetworkGenomic and epigenomic landscapes of adult de novo acute myeloid leukemia.N Engl J Med. 2013; 368: 2059-2074Crossref PubMed Google Scholar], including hairy cell leukemia (HCL) [35Tiacci E. Trifonov V. Schiavoni G. et al.BRAF mutations in hairy-cell leukemia.N Engl J Med. 2011; 364: 2305-2315Crossref PubMed Scopus (326) Google Scholar] and multiple myeloma [36Chapman M.A. Lawrence M.S. Keats J.J. et al.Initial genome sequencing and analysis of multiple myeloma.Nature. 2011; 471: 467-472Crossref PubMed Scopus (519) Google Scholar]. RNA sequencing for the detection of transcripts, quantification of expressed transcripts, and detection of mutations has contributed to the identification of functionally relevant leukemic fusion genes [37Lilljebjörn H. Agerstam H. Orsmark-Pietras C. et al.RNA-seq identifies clinically relevant fusion genes in leukemia including a novel MEF2D/CSF1R fusion responsive to imatinib.Leukemia. 2014; 28: 977-979Crossref PubMed Scopus (5) Google Scholar], and ChIP-sequencing to detect genomic sites occupied by proteins/histone modifications has led to genome-wide identification of binding sites of mutated transcription factors [38Mandoli A. Singh A.A. Jansen P.W. et al.CBFB-MYH11/RUNX1 together with a compendium of hematopoietic regulators, chromatin modifiers and basal transcription factors occupies self-renewal genes in inv(16) acute myeloid leukemia.Leukemia. 2014; 28: 770-778Crossref PubMed Scopus (8) Google Scholar, 39Martens J.H.A. Mandoli A. Simmer F. et al.ERG and FLI1 binding sites demarcate targets for aberrant epigenetic regulation by AML1-ETO in acute myeloid leukemia.Blood. Am Soc Hematol. 2012; 120: 4038-4048Google Scholar, 40Martens J.H.A. Stunnenberg H.G. The molecular signature of oncofusion proteins in acute myeloid leukemia. FEBS Letters.Fed Eur Biochem Soc. 2010; 584: 2662-2669Abstract Full Text Full Text PDF PubMed Scopus (32) Google Scholar, 41Ptasinska A. Assi S.A. Mannari D. et al.Depletion of RUNX1/ETO in t(8;21) AML cells leads to genome-wide changes in chromatin structure and transcription factor binding.Leukemia. 2012; 26: 1829-1841Crossref PubMed Scopus (35) Google Scholar]. Together with whole-genome bisulphite sequencing (WGBS), this allowed for the first time a comprehensive analysis of full leukemia epigenomes [42Pei L. Choi J.-H. Liu J. et al.Genome-wide DNA methylation analysis reveals novel epigenetic changes in chronic lymphocytic leukemia.Epigenetics. 2012; 7: 567-578Crossref PubMed Scopus (29) Google Scholar]. These NGS technologies facilitate characterization of the molecular pathology of different diseases, and they have also been used to differentiate related diseases on the basis of mutations in genes of driver pathways [43Braggio E. Egan J.B. Fonseca R. Stewart A.K. Lessons from next-generation sequencing analysis in hematological malignancies.Blood Cancer J. 2013; 3: e127Crossref PubMed Scopus (11) Google Scholar], gene expression profiles [34Cancer Genome Atlas Research NetworkGenomic and epigenomic landscapes of adult de novo acute myeloid leukemia.N Engl J Med. 2013; 368: 2059-2074Crossref PubMed Google Scholar], and epigenetic patterns [44Kulis M. Heath S. Bibikova M. et al.Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia.Nat Genet. 2012; 44: 1236-1242Crossref PubMed Scopus (105) Google Scholar]. Next we further discuss what has been learned from these genome-wide analyses, with a specific focus on leukemia-associated TFs and the context in which they operate. TFs control expression of genes essential for the normal functioning of the hematopoietic system and regulate development of distinct blood cell types. In the event of genetic perturbations, including deletions, insertions, and translocations, the molecular roles of these TFs can be altered, resulting in uncontrolled proliferation of immature blood cell lineages and sometimes depletion of one or more blood cell lineage. Throughout the years many TFs important for various stages of hematopoietic development have been discovered and molecularly characterized. Many of these were initially identified as being mutated in hematologic disorders. Given their importance to disease, genome-wide profiles were generated for many of these TFs in recent years aiming to further elucidate their role in normal and aberrant hematopoiesis. These genome-wide datasets were mostly submitted to the Gene Expression Omnibus (GEO) online repository (www.ncbi.nlm.nih.gov/geo/) and can be identified through GSE accession numbers. Examples include the core binding factors, MLL, C/EBPα, SPI1, GATA, and TAL1 (Fig. 2). The core binding factor (CBF) family consists of two types of proteins: CBFα, which represents a variable DNA-binding subunit, and CBFβ, a nonDNA-binding subunit thought to stabilize the DNA binding of the α subunit. The CBFα subunit can in distinct cell lineages be represented by one of three proteins: RUNX1, RUNX2 (CBFα1), or RUNX3 (CBFα3) [45Markova E.N. Kantidze O.L. Razin S.V. Transcriptional regulation and spatial organisation of the human AML1/RUNX1 gene.J Cell Biochem. 2011; 112: 1997-2005Crossref PubMed Scopus (9) Google Scholar]. RUNX1 and CBFβ are needed at several stages of hematopoiesis, such as megakaryocytic differentiation, platelet formation, erythropoiesis, B-cell development, and T-cell development [46de Bruijn M.F.T.R. Speck N.A. Core-binding factors in hematopoiesis and immune function.Oncogene. 2004; 23: 4238-4248Crossref PubMed Scopus (136) Google Scholar]. In addition, RUNX1 and CBFβ are common targets of chromosomal translocations. For example, in acute myeloid leukemia (AML) the translocation t(8; 21) involves a fusion of the AML1 and ETO genes and its expression results in the chimeric protein AML1-ETO [47Meyers S. Lenny N. Hiebert S.W. The t(8;21) fusion protein interferes with AML-1B-dependent transcriptional activation.Mol Cell Biol. 1995; 15: 1974-1982Crossref PubMed Google Scholar], whereas inversion of a part of chromosome 16 results in the fusion of the CBFβ and MYH11 genes, coding the chimeric protein CBFβ-MYH11 [48Lutterbach B. Hou Y. Durst K.L. Hiebert S.W. The inv(16) encodes an acute myeloid leukemia 1 transcriptional corepressor.Proc Natl Acad Sci USA. 1999; 96: 12822-12827Crossref PubMed Scopus (75) Google Scholar]. The two CBFs are reported to be mutated in 5–10% of AMLs and function by interfering with the normal CBF gene program [40Martens J.H.A. Stunnenberg H.G. The molecular signature of oncofusion proteins in acute myeloid leukemia. FEBS Letters.Fed Eur Biochem Soc. 2010; 584: 2662-2669Abstract Full Text Full Text PDF PubMed Scopus (32) Google Scholar]. MLL fusions are reported in approximately 10% of acute leukemias (AL). In ALs, the MLL protein is reported to fuse with one of >50 identified partner genes and generates a chimeric protein [49Dou Y. Hess J.L. Mechanisms of transcriptional regulation by MLL and its disruption in acute leukemia.Int J Hematol. 2008; 87: 10-18Crossref PubMed Scopus (37) Google Scholar, 50Mitterbauer-Hohendanner G. Mannhalter C. The biological and clinical significance of MLL abnormalities in haematological malignancies.Eur J Clin Invest. 2004; 34: 12-24Crossref PubMed Google Scholar, 51Krivtsov A.V. Armstrong S.A. MLL translocations, histone modifications and leukaemia stem-cell development.Nat Rev Cancer. 2007; 7: 823-833Crossref PubMed Scopus (449) Google Scholar]. Often, a super elongation complex (SEC) component (like ENL or AF4) is a partner in MLL fusions [52Smith E. Lin C. Shilatifard A. The super elongation complex (SEC) and MLL in development and disease.Genes Dev. 2011; 25: 661-672Crossref PubMed Scopus (112) Google Scholar], and it is speculated that the SEC part of the fusion anomalously stabilizes itself at MLL target genes, consequently modifying the normal MLL transcriptional program [52Smith E. Lin C. Shilatifard A. The super elongation complex (SEC) and MLL in development and disease.Genes Dev. 2011; 25: 661-672Crossref PubMed Scopus (112) Google Scholar, 53Luo Z. Lin C. Shilatifard A. The super elongation complex (SEC) family in transcriptional control.Nat Rev Mol Cell Biol. 2012; 13: 543-547Crossref PubMed Scopus (63) Google Scholar, 54Marschalek R. Mixed lineage leukemia: roles in human malignancies and potential therapy.FEBS J. 2010; 277: 1822-1831Crossref PubMed Scopus (32) Google Scholar, 55Slany R.K. The molecular biology of mixed lineage leukemia.Haematologica. 2009; 94: 984-993Crossref PubMed Scopus (124) Google Scholar] and inducing leukemogenesis. Moreover, MLL fusion proteins are believed to cause changes in chromatin structure through altering histone 3 lysine 4 trimethylation (H3K4me3) and H3K79me2 deposition [56Muyrers-Chen I. Rozovskaia T. Lee N. et al.Expression of leukemic MLL fusion proteins in Drosophila affects cell cycle control and chromosome morphology.Oncogene. 2004; 23: 8639-8648Crossref PubMed Scopus (19) Google Scholar, 57Guenther M.G. Lawton L.N. Rozovskaia T. et al.Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia.Genes Dev. 2008; 22: 3403-3408Crossref PubMed Scopus (104) Google Scholar]. C/EBPα controls differentiation and proliferation of myeloid cells [21Tenen D.G. Hromas R. Licht J.D. Zhang D.E. Transcription factors, normal myeloid development, and leukemia.Blood. 1997; 90: 489-519Crossref PubMed Google Scholar], and mutations have been associated with AML [58Gombart A.F. Hofmann W.-K. Kawano S. et al.Mutations in the gene encoding the transcription factor CCAAT/enhancer binding protein alpha in myelodysplastic syndromes and acute myeloid leukemias.Blood. 2002; 99: 1332-1340Crossref PubMed Scopus (208) Google Scholar, 59Pabst T. Mueller B.U. Zhang P. et al.Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia.Nat Genet. 2001; 27: 263-270Crossref PubMed Scopus (543) Google Scholar]. N-terminal mutations in CEBPα inhibit the expression of the p42 isoform of the C/EBPα protein, resulting in loss of apoptosis and failed myeloid differentiation [59Pabst T. Mueller B.U. Zhang P. et al.Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia.Nat Genet. 2001; 27: 263-270Crossref PubMed Scopus (543) Google Scholar, 60Schuster M.B. Frank A.-K. Bagger F.O. Rapin N. Vikesaa J. Porse B.T. Lack of the p42 form of C/EBPα leads to spontaneous immortalization and lineage infidelity of committed myeloid progenitors.Exp Hematol. 2013; 41: 882-893.e16Abstract Full Text Full Text PDF PubMed Scopus (4) Google Scholar], whereas C-terminal mutations disrupt the DNA-binding domain [58Gombart A.F. Hofmann W.-K. Kawano S. et al.Mutations in the gene encoding the transcription factor CCAAT/enhancer binding protein alpha in myelodysplastic syndromes and acute myeloid leukemias.Blood. 2002; 99: 1332-1340Crossref PubMed Scopus (208) Google Scholar, 61Preudhomme C. Sagot C. Boissel N. et al.Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA).Blood. 2002; 100: 2717-2723Crossref PubMed Scopus (314) Google Scholar] and increase premalignant HSCs [62Bereshchenko O. Mancini E. Moore S. et al.Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPalpha mutant AML.Cancer Cell. 2009; 16: 390-400Abstract Full Text Full Text PDF PubMed Scopus (63) Google Scholar]. Both N- and C-terminal mutations contribute to aggressive leukemia and are reported to be mutated in 10% of AMLs [58Gombart A.F. Hofmann W.-K. Kawano S. et al.Mutations in the gene encoding the transcription factor CCAAT/enhancer binding protein alpha in myelodysplastic syndromes and acute myeloid leukemias.Blood. 2002; 99: 1332-1340Crossref PubMed Scopus (208) Google Scholar, 59Pabst T. Mueller B.U. Zhang P. et al.Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia.Nat Genet. 2001; 27: 263-270Crossref PubMed Scopus (543) Google Scholar, 63Snaddon J. Smith M.L. Neat M. et al.Mutations of CEBPA in acute myeloid leukemia FAB types M1 and M2.Genes Chromosomes Cancer. 2003; 37: 72-78Crossref PubMed Scopus (76) Google Scholar]. Moreover, a decreased expression of C/EBPα is observed in both t(8; 21) [59Pabst T. Mueller B.U. Zhang P. et al.Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia.Nat Genet. 2001; 27: 263-270Crossref PubMed Scopus (543) Google Scholar] and inv(16) [64Cilloni D. Carturan S. Gottardi E. et al.Down-modulation of the C/EBPalpha transcription factor in core binding factor acute myeloid leukemias.Blood. 2003; 102: 2705-2706Crossref PubMed Scopus (24) Google Scholar, 65Helbling D. Mueller B.U. Timchenko N.A. et al.CBFB-SMMHC is correlated with increased calreticulin expression and suppresses the granulocytic differentiation factor CEBPA in AML with inv(16).Blood. 2005; 106: 1369-1375Crossref PubMed Scopus (59) Google Scholar], suggesting that chromosomal translocations as well as the level of CEBPα expression are critical for leukemogenesis. SPI1, which is mutated in 5–7% of AMLs [66Mueller B.U. Pabst T. Osato M. et al.Heterozygous PU.1 mutations are associated with acute myeloid leukemia.Blood. 2002; 100: 998-1007Crossref PubMed Scopus (152) Google Scholar], regulates the proliferation and differentiation of myeloid progenitors [67DeKoter R.P. Walsh J.C. Singh H. PU.1 regulates both cytokine-dependent proliferation and differentiation of granulocyte/macrophage progenitors.EMBO J. 1998; 17: 4456-4468Crossref PubMed Scopus (206) Google Scholar] and is crucial for cell cycle regulation. In murine myeloid cells, for example, PU.1 expression leads to an exit from the cell cycle, thereby blocking proliferation and inducing differentiation [68Ziliotto R. Gruca M.R. Podder S. et al.PU.1 promotes cell cycle exit in the murine myeloid lineage associated with downregulation of E2F1.Exp Hematol. 2014; 42: 204-217.e1Abstract Full Text Full Text PDF PubMed Scopus (6) Google Scholar]. Sustained expression of PU.1 is necessary to modulate the transcription of cell cycle regulators and maintain the HSC population [69Staber P.B. Zhang P. Ye M. et al.Sustained PU.1 levels balance cell-cycle regulators to prevent exhaustion of adult hematopoietic stem cells.Mol Cell. 2013; 49: 934-946Abstract Full Text Full Text PDF PubMed Scopus (25) Google Scholar]. Increased and reduced expression of SPI1 directs commitment to the myeloid lineage and formation of immature eosinophils, respectively [70Engel I. Murre C. Transcription factors in hematopoiesis.Curr Opin Genet Dev. 1999; 9: 575-579Crossref PubMed Scopus (20) Google Scholar]. The expression of SPI1 is deregulated in AML by action of oncofusion proteins. For example, SPI1 is hyperactivated in the presence of MOZ-TIF2 and hypoactivated in the presence of PML-RARα [71Shima Y. Kitabayashi I. Deregulated transcription factors in leukemia.Int J Hematol. 2011; 94: 134-141Crossref PubMed Scopus (13) Google Scholar, 72Wang K. Wang P. Shi J. et al.PML/RARalpha targets promoter regions containing PU.1 consensus and RARE half sites in acute promyelocytic leukemia.Cancer Cell. 2010; 17: 186-197Abstract Full Text Full Text PDF PubMed Scopus (67) Google Scholar]. The members of the GATA zinc finger TF family, such as GATA1, GATA2, and GATA3, are involved in the regulation of hematopoiesis [73Cantor A.B. Orkin S.H. Hematopoietic development: a balancing act.Curr Opin Genet Dev. 2001; 11: 513-519Crossref PubMed Scopus (155) Google Scholar]. The expression of
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
2秒前
3秒前
LYZSh发布了新的文献求助10
3秒前
阔达的黑猫完成签到 ,获得积分10
5秒前
小跳发布了新的文献求助10
6秒前
鲤鱼人英完成签到,获得积分10
6秒前
谢书南发布了新的文献求助10
6秒前
8秒前
8秒前
cc应助科研通管家采纳,获得10
8秒前
Orange应助科研通管家采纳,获得10
8秒前
狮子卷卷完成签到,获得积分10
9秒前
军军问问张完成签到,获得积分20
10秒前
执着柔发布了新的文献求助10
10秒前
12秒前
13秒前
YXHTCM完成签到,获得积分10
13秒前
mrz完成签到,获得积分10
14秒前
谢书南完成签到,获得积分10
15秒前
执着柔完成签到,获得积分10
17秒前
scl发布了新的文献求助10
18秒前
21秒前
宁静致远完成签到,获得积分10
22秒前
幽默珩发布了新的文献求助10
23秒前
吴小苏发布了新的文献求助10
25秒前
26秒前
路漫漫完成签到,获得积分10
27秒前
28秒前
上官若男应助pigpara采纳,获得10
30秒前
上官若男应助幽默珩采纳,获得10
31秒前
路漫漫发布了新的文献求助10
32秒前
FashionBoy应助哈哈哈的一笑采纳,获得100
33秒前
33秒前
搜集达人应助路漫漫采纳,获得10
38秒前
大气的画板完成签到,获得积分10
39秒前
端庄的小蝴蝶完成签到,获得积分10
39秒前
41秒前
ZDF完成签到,获得积分20
41秒前
汉堡包应助KSung采纳,获得10
42秒前
44秒前
高分求助中
Microlepidoptera Palaearctica, Volumes 1 and 3 - 13 (12-Volume Set) [German] 1122
Дружба 友好报 (1957-1958) 1000
The Data Economy: Tools and Applications 1000
Mantiden - Faszinierende Lauerjäger – Buch gebraucht kaufen 600
PraxisRatgeber Mantiden., faszinierende Lauerjäger. – Buch gebraucht kaufe 600
A Dissection Guide & Atlas to the Rabbit 600
Revolution und Konterrevolution in China [by A. Losowsky] 500
热门求助领域 (近24小时)
化学 医学 生物 材料科学 工程类 有机化学 生物化学 物理 内科学 纳米技术 计算机科学 化学工程 复合材料 基因 遗传学 催化作用 物理化学 免疫学 量子力学 细胞生物学
热门帖子
关注 科研通微信公众号,转发送积分 3108991
求助须知:如何正确求助?哪些是违规求助? 2759776
关于积分的说明 7657017
捐赠科研通 2414379
什么是DOI,文献DOI怎么找? 1281314
科研通“疑难数据库(出版商)”最低求助积分说明 618565
版权声明 599445