Advertisement
Review article| Volume 83, P74-84, March 2020

ASXL1 mutation in clonal hematopoiesis

  • Takeshi Fujino
    Affiliations
    Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
    Search for articles by this author
  • Toshio Kitamura
    Correspondence
    Offprint requests to: Toshio Kitamura, MD, PhD, Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 1088639, Japan
    Affiliations
    Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
    Search for articles by this author
Open AccessPublished:January 13, 2020DOI:https://doi.org/10.1016/j.exphem.2020.01.002

      Highlights

      • The epigenetic regulator ASXL1 is frequently mutated in CH and in a wide range of myeloid neoplasms.
      • ASXL1 mutations could contribute to pathogenesis through loss-of-function, dominant-negative, or gain-of-function mutations.
      • Aberrant histone modifications, signal transduction, and autoimmunity may be responsible for ASXL1 mutation-induced CH.
      Recent advances in DNA sequencing technologies have enhanced our knowledge about several diseases. Coupled with easy accessibility to blood samples, hematology plays a leading role in understanding the process of carcinogenesis. Clonal hematopoiesis (CH) with somatic mutations is observed in at least 10% of people over 65 years of age, without apparent hematologic disorders. CH is associated with increased risk of hematologic malignancies, which is indicative of a pre-malignant condition. Therefore, a better understanding of CH will help elucidate the mechanism of multi-step tumorigenesis in the hematopoietic system. Somatic mutations of ASXL1 are frequently detected in CH and myeloid malignancies. Although ASXL1 does not have any catalytic activity, it is involved in multiple histone modifications including H3K4me3, H3K27me3, and H2AK119Ub, suggesting its function as a scaffolding protein. Most ASXL1 mutations detected in CH and myeloid malignancies are frameshift or nonsense mutations of the last exon, generating a C-terminally truncated protein. Deletion of Asxl1 or expression of mutant ASXL1 in mice alters histone modifications and facilitates aberrant gene expression, resulting in myeloid transformation. On the contrary, these mice exhibit impaired functioning of hematopoietic stem cells (HSCs), suggesting the negative effects of ASXL1 mutations on stem cell function. Thus, how ASXL1 mutations induce a clonal advantage of hematopoietic cells and subsequent CH development has not been elucidated. Here, we have reviewed the current literature that enhances our understanding of ASXL1, including its mutational landscape, function, and involvement of its mutation in pathogenesis of CH and myeloid malignancies. Finally, we discuss the potential causes of CH harboring ASXL1 mutations with our latest knowledge.

      Mutational landscape of ASXL1 in CH and myeloid malignancies

      The presence of clonal hematopoiesis (CH) was first recognized around 50 years ago [
      • Oni SB
      • Osunkoya BO
      • L L
      Paroxysmal nocturnal hemoglobinuria: evidence for monoclonal origin of abnormal red cells.
      ]. In females, one of the two X chromosomes is randomly inactivated; this is termed X-inactivation or lyonization. The occurrence of a biased X-inactivation, toward either the paternal or maternal X chromosome, is called X-inactivation skewing (XIS). XIS has been used to analyze clonality since the 1960s. In 1994, Fey et al. [
      • Fey MF
      • Liechti-Gallati S
      • Rohr A Von
      • et al.
      Clonality and X-inactivation patterns in hematopoietic cell populations detected by the highly informative M27 beta DNA probe.
      ] reported that XIS occurred in the peripheral blood of healthy women and was more prevalent in elderly women (75–96 years old) than in younger women (20–58 years old). Busque et al. [
      • Busque L
      • Mio R
      • Mattioli J
      • et al.
      Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age.
      ] suggested that increased XIS in healthy individuals undergoing aging represents clonal expansion of hematopoietic cells, that is, CH. Considering the increased incidence of hematologic malignancies during aging, they also predicted that CH could be a premalignant state resulting from somatic mutations that confer a clonal advantage on hematopoietic stem cells (HSCs).
      Advances in next-generation sequence technologies have enabled us to detect somatic mutation events with higher sensitivity. In 2014, three groups simultaneously reported that clonal expansion of blood cells harboring somatic mutations was present in about 10% of healthy individuals older than 65 years of age without abnormality in their hematologic parameters [
      • Xie M
      • Lu C
      • Wang J
      • et al.
      Age-related mutations associated with clonal hematopoietic expansion and malignancies.
      ,
      • Genovese G
      • Kähler AK
      • Handsaker RE
      • et al.
      Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
      ,
      • Jaiswal S
      • Fontanillas P
      • Flannick J
      • et al.
      Age-related clonal hematopoiesis associated with adverse outcomes.
      ]. These studies also identified somatic mutations in epigenetic regulators, including DNMT3A, ASXL1, and TET2, to be among the most frequently mutated genes in individuals with CH. Given that genes mutated in CH are similar to those in myeloid malignancies, and that CH is associated with an increased risk of subsequent hematologic malignancies [
      • Genovese G
      • Kähler AK
      • Handsaker RE
      • et al.
      Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
      ,
      • Jaiswal S
      • Fontanillas P
      • Flannick J
      • et al.
      Age-related clonal hematopoiesis associated with adverse outcomes.
      ], CH is considered a premalignant condition, as previously mentioned. Moreover, Jaiswal et al. [
      • Jaiswal S
      • Natarajan P
      • Silver AJ
      • et al.
      Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease.
      ] reported that individuals with CH are at an increased risk of developing cardiovascular diseases and atherosclerosis, and mutations in DNMT3A, TET2, ASXL1, and JAK2 were recurrently detected in them. Studies suggest that macrophage-driven inflammatory responses are responsible for the accelerated atherosclerosis seen in these patients [
      • Jaiswal S
      • Natarajan P
      • Silver AJ
      • et al.
      Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease.
      ,
      • Wang W
      • Liu W
      • Fidler T
      • et al.
      Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in JAK2V617F mice.
      ,
      • Fuster Jose J
      • MacLauchlan S
      • Zuriaga MA
      • et al.
      Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice.
      ].
      In addition to CH, somatic mutations in the ASXL1 gene are frequently detected in various types of myeloid malignancies including myelodysplastic syndromes (MDS, 14%−23%) [
      • Scott BL
      • Deeg HJ
      Myelodysplastic syndromes.
      ,
      • Bejar R
      • Stevenson K
      • Abdel-Wahab O
      • et al.
      Clinical effect of point mutations in myelodysplastic syndromes.
      ,
      • Papaemmanuil E
      • Gerstung M
      • Malcovati L
      • et al.
      Clinical and biological implications of driver mutations in myelodysplastic syndromes.
      ,
      • Haferlach T
      • Nagata Y
      • Grossmann V
      • et al.
      Landscape of genetic lesions in 944 patients with myelodysplastic syndromes.
      ,
      • Boultwood J
      • Perry J
      • Pellagatti A
      • et al.
      Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia.
      ,
      • Chen TC
      • Hou HA
      • Chou WC
      • et al.
      Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome.
      ,
      • Thol F
      • Friesen I
      • Damm F
      • et al.
      Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes.
      ], chronic myelomonocytic leukemia (CMML, 40%−49%) [
      • Gelsi-Boyer V
      • Trouplin V
      • Roquain J
      • et al.
      ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia.
      ,
      • Itzykson R
      • Kosmider O
      • Renneville A
      • et al.
      Prognostic score including gene mutations in chronic myelomonocytic leukemia.
      ,
      • Itzykson R
      • Solary E
      An evolutionary perspective on chronic myelomonocytic leukemia.
      ], myeloproliferative neoplasms (MPN, 5%−11%) [
      • Vannucchi AM
      • Lasho TL
      • Guglielmelli P
      • et al.
      Mutations and prognosis in primary myelofibrosis.
      ,
      • Nangalia J
      • Massie CE
      • Baxter EJ
      • et al.
      Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2.
      ,
      • Lundberg P
      • Karow A
      Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms.
      ,
      • Schischlik F
      • Kralovics R
      Mutations in myeloproliferative neoplasms—their significance and clinical use.
      ], and acute myeloid leukemia (AML, 5%−17%) [
      • Boultwood J
      • Perry J
      • Pellagatti A
      • et al.
      Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia.
      ,
      • Papaemmanuil E
      • Gerstung M
      • Bullinger L
      • et al.
      Genomic classification and prognosis in acute myeloid leukemia.
      ,
      • Mengistu G
      • Balcha F
      • Britton S
      Acute myeloid leukemia.
      ,
      • Meyer SC
      • Levine RL
      Translational implications of somatic genomics in acute myeloid leukaemia.
      ,
      The Cancer Genome Atlas Research Network
      Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia.
      ,
      • Welch JS
      • Ley TJ
      • Link DC
      • et al.
      The origin and evolution of mutations in acute myeloid leukemia.
      ,
      • Marcucci G
      • Haferlach T
      • Döhner H
      Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications.
      ,
      • Metzeler KH
      • Becker H
      • Maharry K
      • et al.
      ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category.
      ]. Mutations in ASXL1 are related to poor prognosis of these diseases. It has been found that the majority of patients with ASXL1 mutations have other mutations at the same time. Mutations in ASXL1 co-exist with those in epigenetic factors (IDH2, EZH2), splicing factors (SRSF2, U2AF1), signal transduction molecules (JAK2, NRAS, SETBP1), transcription factor (RUNX1), and components of cohesion complex (STAG2) [
      • Papaemmanuil E
      • Gerstung M
      • Malcovati L
      • et al.
      Clinical and biological implications of driver mutations in myelodysplastic syndromes.
      ,
      • Haferlach T
      • Nagata Y
      • Grossmann V
      • et al.
      Landscape of genetic lesions in 944 patients with myelodysplastic syndromes.
      ,
      • Chen TC
      • Hou HA
      • Chou WC
      • et al.
      Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome.
      ,
      • Schnittger S
      • Eder C
      • Jeromin S
      • et al.
      ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome.
      ,
      • Tyner JW
      • Tognon CE
      • Bottomly D
      • et al.
      Functional genomic landscape of acute myeloid leukaemia.
      ,
      • Gerstung M
      • Pellagatti A
      • Malcovati L
      • et al.
      Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes.
      ]. On the other hand, ASXL1 mutations and DNMT3A, NPM1, FLT3-ITD, and WT1 mutations are mutually exclusive [
      • Haferlach T
      • Nagata Y
      • Grossmann V
      • et al.
      Landscape of genetic lesions in 944 patients with myelodysplastic syndromes.
      ,
      • Schnittger S
      • Eder C
      • Jeromin S
      • et al.
      ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome.
      ,
      • Tyner JW
      • Tognon CE
      • Bottomly D
      • et al.
      Functional genomic landscape of acute myeloid leukaemia.
      ]. Of note, recent studies have reported that CH is detected in half of the patients with aplastic anemia (AA) in whom ASXL1 mutations are common [
      • Yoshizato T
      • Dumitriu B
      • Hosokawa K
      • et al.
      Somatic mutations and clonal hematopoiesis in aplastic Anemia.
      ]. As patients with AA sometimes develop MDS or AML, and genes mutated in these diseases seem to be overlapping, it is possible that MDS and AML could develop from the same CH clones as AA in some patients. Taken together, ASXL1 mutations are closely involved with the development of CH and myeloid malignancies, suggesting that they are among the earliest events in the process of malignant transformation.

      ASXL1 as an epigenetic regulator

      The ASXL1 gene was identified as one of the three human homologues of the Drosophila Asx gene and it functions as an epigenetic regulator [
      • Fisher CL
      • Berger J
      • Randazzo F
      • Brock HW
      A human homologue of Additional sex combs, ADDITIONAL SEX COMBS-LIKE 1, maps to chromosome 20q11.
      ]. ASXL1 consists of 1,541 amino acid residues, and harbors the ASXH domain in the N-terminal region and the plant homeodomain (PHD) domain in the C-terminal region (Figure 1) [
      • Schnittger S
      • Eder C
      • Jeromin S
      • et al.
      ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome.
      ,
      • Chou WC
      • Chou SC
      • Liu CY
      • et al.
      TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics.
      ,
      • Tan M
      • Ng IKS
      • Chen Z
      • et al.
      Clinical implications of DNMT3A mutations in a Southeast Asian cohort of acute myeloid leukaemia patients.
      ,
      • Hou HA
      • Kuo YY
      • Liu CY
      • et al.
      DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications.
      ,
      • Chou WC
      • Huang HH
      • Hou HA
      • et al.
      Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations.
      ]. The former is required for interaction with carboxy-terminal hydrolase BAP1 [
      • Balasubramani A
      • Larjo A
      • Bassein JA
      • et al.
      Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex.
      ]. The latter binds to chromatin and DNA [
      • Sanchez R
      • Zhou MM
      The PHD finger: a versatile epigenome reader.
      ], suggesting the importance of this domain in epigenetic regulation.
      Figure 1
      Figure 1Localization of DNMT3A, TET2, and ASXL1 mutations in AML patients. Mutations in DNMT3A and TET2 include missense, nonsense, or frameshift mutations. They tend to spread throughout the genes, except for DNMT3A R882 hotspot mutation. Nonsense or frameshift mutations in the ASXL1 gene are concentrated in N-terminus of the last exon, generating a C-terminally truncated form of ASXL1 (Mutant ASXL1). Each mutation is depicted as a circle (white: frame shift mutation, gray: missense mutation, black: nonsense mutation). Hotspot mutations are indicated in boldface. ASXH=Asx homology; ASXN=Asx N-terminal; Cys-rich=cysteine-rich; DSBH=double stranded β-helix; MTase=methyltransferase; NR box=nuclear receptor co-regulator binding motif; PHD=plant homeodomain; PWWP=proline–tryptophan–tryptophan–proline; ZNF=zinc finger.
      In Drosophila, expression of genes required for somitogenesis during the embryonic stage is regulated by both Trithorax group (TrxG) and Polycomb group (PcG) complexes [
      • Kennison JA
      • Tamkun JW
      Dosage-dependent modifiers of Polycomb and Antennapedia mutations in Drosophila.
      ,
      • Simon J
      • Chiang A
      • Bender W
      Ten different Polycomb group genes are required for spatial control of the abdA and AbdB homeotic products.
      ,
      • Kennison JA
      Transcriptional activation of Drosophila homeotic genes form distant regulatory elements.
      ,
      • Pirrotta V
      PcG complexes and chromatin silencing.
      ,
      • Schuettengruber B
      • Bourbon HM
      • Di Croce L
      • Cavalli G
      Genome regulation by Polycomb and Trithorax: 70 years and counting.
      ]. The TrxG complex trimethylates histone H3 at lysine 4 (H3K4me3), resulting in transcriptional activation. The PcG complex is divided into two groups: Polycomb repressive complex 1 (PRC1) and Polycomb repressive complex 2 (PRC2). The former ubiquitinates histone H2A at lysine 119 (H2AK119Ub), and the latter trimethylates histone H3 at lysine 27 (H3K27me3). Both modifications repress the expression of the target genes [
      • Schuettengruber B
      • Bourbon HM
      • Di Croce L
      • Cavalli G
      Genome regulation by Polycomb and Trithorax: 70 years and counting.
      ,
      • Wang H
      • Wang L
      • Erdjument-Bromage H
      • Vidal M
      • Tempst P
      • Jones RS ZY
      Role of histone H2A ubiquitination in Polycomb silencing.
      ,
      • de Napoles M
      • Mermoud JE
      • Wakao R
      • et al.
      Polycomb group proteins ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation.
      ,
      • Wang L
      • Brown JL
      • Cao R
      • Zhang Y
      • Kassis JA
      • Jones RS
      Hierarchical recruitment of polycomb group silencing complexes.
      ]. Deletion of Asx in Drosophila exhibits both TrxG- and PcG-deficient phenotypes [
      • Milne TA
      • Sinclair DAR
      • Brock HW
      The Additional sex combs gene of Drosophila is required for activation and repression of homeotic loci, and interacts specifically with Polycomb and super sex combs.
      ], suggesting that Asx is indispensable for histone modifications by TrxG and PcG proteins.
      In addition to Drosophila Asx, recent studies have uncovered the relationship between ASXL1 and histone modifications in mammals. Abdel-Wahab et al. [
      • Abdel-Wahab O
      • Adli M
      • LaFave LM
      • et al.
      ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression.
      ] reported that ASXL1 bound to components of PRC2 including EZH2 and EED. ASXL1 loss resulted in global exclusion of H3K27me3, implying that ASXL1 cooperated with PRC2 complex to trimethylate H3K27. Scheuermann et al. [
      • Scheuermann JC
      • De Ayala Alonso AG
      • Oktaba K
      • et al.
      Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB.
      ] have shown that ASXL1 and BAP1 form a complex, termed Polycomb repressive de-ubiquitinase (PR-DUB). PR-DUB complex removes monoubiquitin from H2AK119Ub to derepress genes targeted by PRC1. Intriguingly, mutations in components of PRC1 (BCOR and BCORL1) and PRC2 (EZH2, EED, and SUZ12) complexes are recurrently detected in myeloid malignancies, suggesting that ASXL1 mutations induce leukemogenesis via alteration of normal histone modifications. Of note, in patients with ASXL1 mutations, concurrent mutations in EZH2 are frequently detected [
      • Haferlach T
      • Nagata Y
      • Grossmann V
      • et al.
      Landscape of genetic lesions in 944 patients with myelodysplastic syndromes.
      ,
      • Chen TC
      • Hou HA
      • Chou WC
      • et al.
      Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome.
      ], suggesting that ASXL1 mutations contribute to malignant transformation through mechanisms other than PRC2 function.
      Lastly, it has been reported that ASXL1 loss in mice results in reduced global levels of H3K4me3 through unknown mechanisms [
      • Wang J
      • Li Z
      • He Y
      • et al.
      Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
      ]. We have previously revealed that knockdown of ASXL1 decreases the level of H3K4me3 in human leukemic cells and proposed that the ASXL1–OGT–HCFC1 complex collaborates with MLL5 to trimethylate H3K4 [
      • Inoue D
      • Fujino T
      • Sheridan P
      • et al.
      A novel ASXL1–OGT axis plays roles in H3K4 methylation and tumor suppression in myeloid malignancies.
      ]. Although the exact mechanism remains to be fully elucidated, these studies strongly suggest the involvement of ASXL1 in the trimethylation of H3K4. Collectively, ASXL1 is involved in various histone modifications, such as H3K27me3, H2AK119Ub, and H3K4me3, to regulate gene expression, and might function as a scaffold for epigenetic regulators.

      Roles of ASXL1 in hematopoiesis

      As described earlier, ASXL1 is involved in various histone modifications to maintain proper gene expression. To clarify the role of ASXL1 in normal hematopoiesis and the impact of ASXL1 deficiency on the pathogenesis of myeloid malignancies, Asxl1-deficient mice have been generated by several groups (Table 1) [
      • Wang J
      • Li Z
      • He Y
      • et al.
      Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
      ,
      • Fisher CL
      • Pineault N
      • Brookes C
      • et al.
      Loss-of-function additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia.
      ,
      • Abdel-Wahab O
      • Gao J
      • Adli M
      • et al.
      Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo.
      ].
      Table 1Overview of genetically engineered mouse models
      Mouse modelPeripheral bloodHSPCsDiseaseHistone modificationFeatureCitation
      Constitutive Asxl1knockout mouseNo changes were observedSlight reduction of GM and GEMM colony-forming abilityNo hematopoietic diseases were observed (58 wk)No dataReduced T cells in thymus Reduced B cells in bone marrow, splenomegaly with increased myeloid cells
      • Fisher CL
      • Pineault N
      • Brookes C
      • et al.
      Loss-of-function additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia.
      Vav-Cre/Mx1-Cre conditional Asxl1 knockout mouseProgressive leukocytopenia and anemia (>6 mo), dysplasiaIncreased LT-HSCs, decreased repopulation abilityMDS-like disease (after transplantation)Reduction in H3K27me3 levelsDefect in erythroid differentiation, increased expression of HoxA7, HoxA9, and p16 Ink4a
      • Abdel-Wahab O
      • Gao J
      • Adli M
      • et al.
      Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo.
      Constitutive Asxl1 knockout mousePancytopenia, myeloid skewing, dysplasiaDecreased LSK cells, increased GMPs, decreased repopulation abilityMDS-like diseaseReduction in H3K4me3 and H3K27me3 levelsIncreased expression of HoxA5, HoxA7, HoxA9, and HoxA10
      • Wang J
      • Li Z
      • He Y
      • et al.
      Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
      Asxl1Y588X transgenic mouseAnemia, thrombocytosis, myeloid skewingIncreased LSK cells and GMPs, decreased MEPs, increased repopulation abilityAML, MPN, and MDS-like diseasesIncreased levels of H3K112Ac and H3K27AcIncreased chromatin accessibility, mutant ASXL1 gains interaction with BRD4 to increase expression of Prdm16
      • Yang H
      • Kurtenbach S
      • Guo Y
      • et al.
      Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies.
      Constitutive Asxl1G643fs knock-in mouse (inserted into Asxl1 locus)Slight increase in number of RBCs and WBCs (18 mo)Decreased repopulation abilityNo hematopoietic diseases were observed (18 mo)Changes in distribution of H3K27me3Increased expression of MN1, increase in the number of DEGs associated with upregulation of signatures of HSC, KRAS pathway, and MEK pathway
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      Constitutive Asxl1G643fs knock-in mouse (inserted into Asxl1 locus)Leukocytopenia and thrombocytosis (12 mo), increased myeloid cellsDecreased LT-HSCs, CMPs, and GMPsMDS/MPN-like diseaseDecreased H2AK119Ub level at p16 Ink4a locusMutant ASXL1 lost ability to interact with BMI1, leading to depression of PRC1 target genes
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      Conditional ASXL1E635fs knock-in mouse (inserted into Rosa26 locus)Decreased RBC, thrombocytosis and myeloid skewing (>70 wk)Decreased LT-HSCs, decreased repopulation abilityNo hematopoietic diseases were observed (18 mo), increased susceptibility to leukemic transformationReduction in H3K4me3 and H2AK119Ub levelsDefect in erythroid differentiation, downregulation of erythroid differentiation-related genes
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      Wang et al. [
      • Wang J
      • Li Z
      • He Y
      • et al.
      Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
      ] generated an Asxl1 conventional knockout mouse and demonstrated that heterogeneous loss of Asxl1 led to the development of an MDS-like disease. In these mice, Asxl1 loss resulted in impaired function of HSCs associated with pancytopenia and myeloid-biased differentiation. Moreover, Linc-kit+ cells in Asxl1 knockout mice decreased global levels of H3K4me3 and H3K27me3. Abdel-Wahab et al. [
      • Abdel-Wahab O
      • Gao J
      • Adli M
      • et al.
      Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo.
      ] bred Cre-mediated conditional Asxl1 knockout mice with interferon-inducible Mx1-Cre transgenic mice or hematopoietic lineage-specific Vav-Cre transgenic mice. Conditional deletion of Asxl1 in mice resulted in age-dependent leukocytopenia and anemia with dysplasia in the context of impaired differentiation. Asxl1 deficiency caused an increase in HSCs, but impaired self-renewal of HSCs associated with increased apoptosis. Bone marrow transplantation of Asxl1-deficient cells led to the development of lethal MDS-like disease. In these mice, Asxl1 loss resulted in global reduction of H3K27me3, followed by de-repression of posterior HoxA genes and p16INK4a, suggesting a dysfunction of PRC2.
      These studies suggest that ASXL1 is indispensable for normal differentiation of blood cells and maintenance of HSC function, presumably through epigenetic regulations including trimethylation of H3K4 and H3K27. Importantly, these knockout mouse models suggest that ASXL1 mutations could cause hematologic malignancies by loss of function.

      ASXL1 mutations gain functions

      A series of studies using Asxl1 knockout mice closely recapitulate the pathology of human MDS, implying loss of function as a consequence of ASXL1 mutations. Apart from the knockout mouse model, several studies have revealed that the expression of mutant ASXL1 in mice induces the development of MDS-like disease, raising a question about whether the loss of ASXL1 function is necessary and sufficient for the pathogenesis of myeloid malignancies as well as CH.
      Here, we summarize mutations in epigenetic regulators including DNMT3A, TET2, and ASXL1 (Figure 1). Mutations of TET2 seem to be dispersed across the coding region, suggesting loss-of-function mutations, which supports the validity of using a knockout mouse as a model for TET2-mutated diseases [
      • Chou WC
      • Chou SC
      • Liu CY
      • et al.
      TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics.
      ]. DNMT3A mutations are probably mixtures of the frequent missense mutations (R882C/H/S) and dispersed mutations, suggesting gain-of-function and loss-of-function mutations, respectively [
      • Tan M
      • Ng IKS
      • Chen Z
      • et al.
      Clinical implications of DNMT3A mutations in a Southeast Asian cohort of acute myeloid leukaemia patients.
      ,
      • Hou HA
      • Kuo YY
      • Liu CY
      • et al.
      DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications.
      ,
      • Ley TJ
      • Ding L
      • Walter MJ
      • et al.
      DNMT3A mutations in acute myeloid leukemia.
      ]. On the other hand, most mutations in ASXL1 are frameshift or nonsense mutations in the N-terminal region of the last exon, generating a C-terminally truncated form of ASXL1 lacking PHD domain (hereinafter referred to as mutant ASXL1) [
      • Schnittger S
      • Eder C
      • Jeromin S
      • et al.
      ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome.
      ]. In general, mRNAs containing premature termination codons (PTCs) are degraded by nonsense-mediated mRNA decay (NMD). However, as mutations in ASXL1 are located in the last exon, they are assumed to escape from NMD. In fact, we have found that the C-terminally truncated form of ASXL1 was present in leukemic cell lines by mass spectrometry and western blot analyses [
      • Inoue D
      • Matsumoto M
      • Nagase R
      • et al.
      Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels.
      ]. These results suggest that mutations in ASXL1 may result in dominant-negative or gain-of function mutations, and further raise the issue as to whether a knockout mouse that has been conventionally used is appropriate for a disease model of human ASXL1-mutated diseases.
      We previously investigated if mutant ASXL1 proteins induce myeloid transformation using a mouse transplantation model [
      • Inoue D
      • Kitaura J
      • Togami K
      • et al.
      Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations.
      ]. Mice transplanted with bone marrow cells expressing mutant ASXL1 exhibited progressive pancytopenia, multilineage dysplasia after a long latency, and subsequent overt leukemia in some mice, mimicking MDS in humans. We found that mutant ASXL1 caused de-repression of posterior HoxA genes and miR-125a through inhibition of PRC2-mediated H3K27 trimethylation. De-repression of miR-125a decreased the expression of Clec5a in addition to its known target p53 pathway genes, leading to impaired myeloid differentiation. As is the case with the loss-of-function model, these data suggest that mutant ASXL1 inhibits PRC2 function, followed by upregulation of its target genes in a dominant-negative manner, which contributes to the development of myeloid malignancies.
      As described above, ASXL1 and BAP1 form a DUB complex to de-ubiquitinate H2AK119Ub. Recently, our group and others have revealed that mutant ASXL1 aberrantly enhances the de-ubiquitinase (DUB) activity of BAP1 and altered gene expression [
      • Balasubramani A
      • Larjo A
      • Bassein JA
      • et al.
      Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex.
      ,
      • Asada S
      • Goyama S
      • Inoue D
      • et al.
      Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis.
      ]. Mechanistically, BAP1 mono-ubiquitinated and stabilized mutant ASXL1, which in turn enhanced the DUB activity of BAP1. The overactive mutant ASXL1/BAP1 complex aberrantly de-ubiquitinated H2AK119Ub, followed by de-repression of its target genes including posterior HOXA genes and IRF8, leading to leukemic transformation and impaired myeloid differentiation, respectively. These results indicate that mutations in ASXL1 can contribute to myeloid transformation by means of gain of function.
      Collectively, ASXL1 mutations can provoke malignant transformation through not merely loss-of-function but also dominant-negative and gain-of-function mechanisms.

      Impact of ASXL1 mutations on hematopoiesis in vivo

      It is becoming accepted that the mutant ASXL1 protein is involved in the pathogenesis of myeloid malignancies, and a series of studies using mutant ASXL1 knock-in mice has been published by several groups. In this section, we summarize the characterization of these knock-in mouse models (Table 1) [
      • Yang H
      • Kurtenbach S
      • Guo Y
      • et al.
      Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies.
      ,
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ,
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ,
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      ].
      Hsu et al. [
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ] generated a constitutive knock-in mouse carrying the mouse Asxl1 G643WfsX12 mutation (corresponding to the human G646WfsX12 mutation) in the endogenous Asxl1 gene. Mutant ASXL1 knock-in mice did not exhibit obvious abnormalities in hematologic parameters and histology. Moreover, they did not develop hematologic malignancies during the observation period of 18 months, indicating that an ASXL1 mutation alone is not sufficient to induce hematologic diseases. In these mice, mutant ASXL1 impaired the long-term repopulation potential of HSCs and slightly disrupted distribution of H3K27me3. Uni et al. [
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ] also engineered and analyzed mouse Asxl1 G643WfsX12 constitutive knock-in mice. These mice exhibited age-dependent leukocytopenia and thrombocytosis. The number of LinSca1+c-kit+ (LSK) cells decreased along with increased apoptosis in mutant ASXL1 knock-in mice, suggesting a negative impact of mutant ASXL1 on hematopoietic stem and progenitor cells (HSPCs). Unlike a study by Hsu et al. [
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ], Uni et al.’s [
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ] mice developed MDS/MPN-like diseases associated with dysplasia in myeloid lineage 1.5–2 years after birth. They reported that wild-type ASXL1, but not mutant ASXL1, bound to BMI1, a component of PRC1. The levels of H2AK119Ub in the p16Ink4a locus, which is a known target of PRC1, was decreased, leading to de-repression of p16Ink4a in mutant ASXL1-expressing Lin cells. p16Ink4a loss normalized the number and frequency of Annexin V-positive cells in LSK cells of mutant ASXL1 knock-in mice. These data suggest that mutant ASXL1 disturbs the recruitment of PRC1 to the p16Ink4a locus and induces de-repression of p16Ink4a, resulting in increased apoptosis and a decreased number of HSPCs. We generated a hematopoietic lineage-specific Vav-Cre mutant ASXL1 knock-in mouse harboring a human ASXL1 E635RfsX15 mutation inserted into the Rosa26 locus [
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      ]. Although the mutant mice exhibited impaired erythroid differentiation and age-dependent mild anemia, they did not develop hematologic disorders during the observation period of 70 weeks. Thus, similar to a report from Hsu et al. [
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ], an ASXL1 mutation alone was insufficient for inducing the development of hematologic malignancies. However, they increased susceptibility of leukemogenesis by co-occurring expression of mutant RUNX1 or viral insertional mutagenesis indicative of a pre-malignant state. Similar to other knock-in mouse models, mutant ASXL1 reduced the number and the function of HSCs. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) analysis using c-kit+ cells revealed global reduction in levels of H2AK119Ub and H3K4me3 in mutant ASXL1 knock-in mice. In particular, levels of H3K4me3 at the loci of erythroid differentiation-related genes, such as Id3, Tjp1 and Sox6, were markedly decreased.
      Taken together, these studies suggest that mutant ASXL1 impairs hematopoiesis through dysregulation of epigenetic modifications. The phenotypic differences between mouse models may come from the site of mutations, expression levels, or genetic background (Asxl1 locus-specific knock-in or Rosa26 locus knock-in). In any case, it is likely that a single ASXL1 mutation is not sufficient to induce malignant transformation, raising the possibility that mutant ASXL1 knock-in mice represent a premalignant condition such as CH.

      Mechanistic link between ASXL1 mutations and CH

      Although precise mechanisms underlying ASXL1 mutation-induced CH remain elusive, previous studies left clues for unraveling this question. Here, we discuss how ASXL1 mutations promote the development of CH considering previous studies on ASXL1 and the general cause of CH. Mutations that confer increased self-renewal, decreased apoptosis, or impaired differentiation could promote a growth advantage for HSCs. Furthermore, both cell-intrinsic and -extrinsic factors, including DNA damage, microenvironmental changes, therapy, and autoimmunity, would act as selective pressures to HSCs (Figure 2). Presumably, HSC clones harboring mutations that confer a growth advantage gradually expand over time in the presence of selective pressures, eventually leading to development of CH.
      Figure 2
      Figure 2ASXL1 regulates gene expression through histone modifications including H2AK119Ub, H3K4me3, and H3K27me3. Mutant ASXL1 abrogates trimethylation of H3K4 and H3K27. In addition, mutant ASXL1 collaborates with BAP1 to promote de-ubiquitination of H2AK119Ub. These alterations in histone modifications induce aberrant gene expression, which may lead to a fitness advantage. Alternatively, abnormal histone modifications caused by mutant ASXL1 may accelerate “epigenetic drift.” As mutations in ASXL1 are frequently detected in CH with aplastic anemia, autoimmunity could be involved in a clonal selection of the HSC pool. It is possible that mutant ASXL1 augments signal transduction to induce proliferation of the CH clone. ASXL1-MT=mutant ASXL1; ASXL1-WT=wild-type ASXL1.

      Epigenetic regulation

      Approximately 80% of somatic mutations detected in CH are epigenetic factors including DNMT3A, TET2, and ASXL1 [
      • Xie M
      • Lu C
      • Wang J
      • et al.
      Age-related mutations associated with clonal hematopoietic expansion and malignancies.
      ,
      • Genovese G
      • Kähler AK
      • Handsaker RE
      • et al.
      Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
      ,
      • Jaiswal S
      • Fontanillas P
      • Flannick J
      • et al.
      Age-related clonal hematopoiesis associated with adverse outcomes.
      ]. DNA methylation marks play a pivotal role in the regulation of gene expression. It was reported that loss of Dnmt3a in mice increased self-renewal and impaired differentiation of HSCs [
      • Challen GA
      • Sun D
      • Jeong M
      • et al.
      Dnmt3a is essential for hematopoietic stem cell differentiation.
      ,
      • Challen GA
      • Sun D
      • Mayle A
      • et al.
      Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells.
      ]. Two groups revealed that loss of Tet2 expanded LSK cells and increased self-renewal of HSCs [
      • Quivoron C
      • Couronné L
      • Della Valle V
      • et al.
      TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis.
      ,
      • Moran-Crusio K
      • Reavie L
      • Shih A
      • et al.
      Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation.
      ]. These studies suggest that the perturbation of DNA methylation profiles could result in increased self-renewal and differentiation block, leading to a fitness advantage of the HSC clone. Similarly, ASXL1 mutations were also expected to increase the self-renewal of HSCs. However, previous studies reported impaired function of HSCs in Asxl1-deficient or mutant ASXL1-expressing mice [
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ,
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ,
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      ]. How this discrepancy occurs is yet to be delineated, and is discussed in the next section. It is likely that ASXL1 mutations provoke differentiation block and aberrant proliferation of HSCs through disruption of epigenetic modifications (e.g., H2AK119Ub, H3K4me3, H3K27me3) [
      • Abdel-Wahab O
      • Adli M
      • LaFave LM
      • et al.
      ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression.
      ,
      • Wang J
      • Li Z
      • He Y
      • et al.
      Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
      ,
      • Abdel-Wahab O
      • Gao J
      • Adli M
      • et al.
      Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo.
      ,
      • Inoue D
      • Kitaura J
      • Togami K
      • et al.
      Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations.
      ,
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ,
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ,
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      ], which could contribute to the development of CH in the long term.
      Apart from aforementioned mechanisms, age-related stochastic alteration of epigenetics can affect a fate decision of HSCs. Sun et al. [
      • Sun D
      • Luo M
      • Jeong M
      • et al.
      Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal.
      ] reported that aged HSCs exhibited hypermethylation in genes associated with differentiation and hypomethylation in genes associated with maintenance of stem cell function. Single-cell analyses of HSCs revealed that chromatin modifications became more heterogeneous between individuals and single cells over time [
      • Cheung P
      • Vallania F
      • Warsinske HC
      • et al.
      Single-cell chromatin modification profiling reveals increased epigenetic variations with aging.
      ]. Such an age-related global epigenetic alteration is referred to as epigenetic drift [
      • Hannum G
      • Guinney J
      • Zhao L
      • et al.
      Genome-wide methylation profiles reveal quantitative views of human aging rates.
      ,
      • Fraga MF
      • Ballestar E
      • Paz MF
      • et al.
      Epigenetic differences arise during the lifetime of monozygotic twins.
      ,
      • Boks MP
      • Derks EM
      • Weisenberger DJ
      • et al.
      The relationship of DNA methylation with age, gender and genotype in twins and healthy controls.
      ]. It is possible that epigenetic drift causes continuous alteration of the epigenomic modifications followed by changes in self-renewal and differentiation potential of HSCs. As epigenetic modifications can be transmitted to daughter cells, they could be important contributors to clonal selection in addition to gene mutations. Mutations in epigenetic regulators including ASXL1 could cause diverse changes in epigenetics to accelerate epigenetic abnormality, which may confer a growth advantage to HSCs.

      DNA damage response

      It has been previously reported that chemotherapy and radiation therapy act as exogenous selective pressures to contribute to the development of CH [
      • Coombs CC
      • Zehir A
      • Devlin SM
      • et al.
      Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes.
      ]. Mutations in DNA damage response (DDR)-related genes, including TP53 and PPM1D, are recurrently found at high frequencies in patients with therapy-related AML (tAML) and MDS (tMDS) [
      • Hsu JI
      • Dayaram T
      • Tovy A
      • et al.
      PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy.
      ,
      • Lindsley RC
      • Saber W
      • Mar BG
      • et al.
      Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation.
      ,
      • Christiansen DH
      • Andersen MK
      • Pedersen-Bjergaard J
      Mutations with loss of heterozygosity of p53 are common in therapy-related myelodysplasia and acute myeloid leukemia after exposure to alkylating agents and significantly associated with deletion or loss of 5q, a complex karyotype, and a poor prognosis.
      ]. Studies by Coombs et al. [
      • Coombs CC
      • Zehir A
      • Devlin SM
      • et al.
      Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes.
      ] revealed that after chemotherapy and radiation therapy among cancer patients, the frequency of such individuals with CH harboring TP53 or PPM1D mutations significantly increased. Moreover, Hsu et al. [
      • Hsu JI
      • Dayaram T
      • Tovy A
      • et al.
      PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy.
      ] and Kahn et al. [
      • Kahn JD
      • Miller PG
      • Silver AJ
      • et al.
      PPM1D-truncating mutations confer resistance to chemotherapy and sensitivity to PPM1D inhibition in hematopoietic cells.
      ] reported that PPM1D mutant-expressing cells are more resistant to cytotoxic chemotherapy compared with wild-type cells. These data suggest that cells bearing TP53 or PPM1D mutations tend to acquire a fitness advantage to develop CH under the exposure of external genotoxic stresses. On the other hand, it is unlikely that ASXL1 mutations confer tolerance against genotoxic stresses on hematopoietic cells like TP53 and PPM1D mutations, as they appear to be unrelated to the prior therapy given [
      • Coombs CC
      • Zehir A
      • Devlin SM
      • et al.
      Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes.
      ,
      • Hsu JI
      • Dayaram T
      • Tovy A
      • et al.
      PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy.
      ,
      • Lindsley RC
      • Saber W
      • Mar BG
      • et al.
      Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation.
      ,
      • Gibson CJ
      • Lindsley RC
      • Tchekmedyian V
      • et al.
      Clonal hematopoiesis associated with adverse outcomes after autologous stem-cell transplantation for lymphoma.
      ,
      • Takahashi K
      • Wang F
      • Kantarjian H
      • et al.
      Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case–control study.
      ]. Consistent with this, HSCs expressing mutant ASXL1 exhibit increased susceptibility to ionizing radiation in our knock-in mouse model (unpublished data).

      Oncogene

      Mutations in classic oncogenic genes involved in signal transduction are not common in CH. In this group, the most frequently detected mutation is Janus kinase 2 (JAK2) V617F substitution [
      • Xie M
      • Lu C
      • Wang J
      • et al.
      Age-related mutations associated with clonal hematopoietic expansion and malignancies.
      ,
      • Genovese G
      • Kähler AK
      • Handsaker RE
      • et al.
      Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
      ,
      • Jaiswal S
      • Fontanillas P
      • Flannick J
      • et al.
      Age-related clonal hematopoiesis associated with adverse outcomes.
      ]. JAK2 V617F mutations are found in the majority of MPN patients, especially in polycythemia vera (PV) [
      • Vannucchi AM
      • Lasho TL
      • Guglielmelli P
      • et al.
      Mutations and prognosis in primary myelofibrosis.
      ,
      • Nangalia J
      • Massie CE
      • Baxter EJ
      • et al.
      Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2.
      ,
      • Lundberg P
      • Karow A
      Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms.
      ,
      • Tenedini E
      • Bernardis I
      • Artusi V
      • et al.
      Targeted cancer exome sequencing reveals recurrent mutations in myeloproliferative neoplasms.
      ]. As for ASXL1, possible involvement between ASXL1 mutations and signal transduction pathways remains unclear. Youn et al. [
      • Youn HS
      • Kim TY
      • Park UH
      • et al.
      Asxl1 deficiency in embryonic fibroblasts leads to cellular senescence via impairment of the AKT-E2F pathway and Ezh2 inactivation.
      ] found that Asxl1 forms a ternary complex with Akt1 and p27 in mouse embryonic fibroblasts (MEFs). They reported that Akt was not phosphorylated after IGF-1 stimulation in Asxl1-deficient MEFs, suggesting a potential role of ASXL1 in the PI3K/AKT signaling pathway. It is possible that ASXL1 mutations may alter the activity of proliferation-driving signal transduction pathways to induce clonal advantage in hematopoietic cells.

      Autoimmunity

      Autoimmunity can provide a selective pressure in HSCs to yield clonal selection in the hematopoietic system. AA is considered to be an autoimmune disease, in which autoreactive cytotoxic T cells attack HSCs and eventually lead to bone marrow failure [
      • Young NS
      Aplastic anemia.
      ]. Recently, genome-wide sequencing has revealed CH in half of AA patients [
      • Yoshizato T
      • Dumitriu B
      • Hosokawa K
      • et al.
      Somatic mutations and clonal hematopoiesis in aplastic Anemia.
      ]. It should be noted that ASXL1 mutations are among the most frequently detected events in AA, raising the possibility that they affect T-cell functions or alter sensitivity of HSCs in response to autoimmunity. At present, the reason why ASXL1 is frequently mutated in AA is largely unknown. Future study focused on the multilineage interaction, especially HSCs with immune cells, can elucidate the pathogenesis of AA with ASXL1 mutations as well as mutant ASXL1-induced CH.

      Mutant ASXL1 knock-in mouse as a model for CH

      As described earlier, mutant ASXL1 has a negative impact on HSC function [
      • Hsu YC
      • Chiu YC
      • Lin CC
      • et al.
      The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
      ,
      • Uni M
      • Masamoto Y
      • Sato T
      • Kamikubo Y
      • Arai S
      • Kurokawa M
      Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
      ,
      • Nagase R
      • Inoue D
      • Pastore A
      • et al.
      Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
      ]. In contrast, CH-related mutations in the epigenetic factor Dnmt3a or Tet2 deficiency in mice increased self-renewal of HSCs [
      • Challen GA
      • Sun D
      • Jeong M
      • et al.
      Dnmt3a is essential for hematopoietic stem cell differentiation.
      ,
      • Challen GA
      • Sun D
      • Mayle A
      • et al.
      Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells.
      ,
      • Quivoron C
      • Couronné L
      • Della Valle V
      • et al.
      TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis.
      ,
      • Moran-Crusio K
      • Reavie L
      • Shih A
      • et al.
      Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation.
      ]. Thus, it is likely that the mechanisms underlying the development of CH induced by ASXL1 mutations are distinct from those of DNMT3A and TET2 mutations. To address this discrepancy, we examined the effect of mutant ASXL1 on physiological aging of HSCs by using our knock-in mouse model. Although mutant ASXL1 induces a competitive growth disadvantage in transplanted HSCs, HSCs expressing mutant ASXL1 markedly increased over time with no symptoms of hematologic malignancies in Vav-Cre mutant ASXL1 knock-in mice (unpublished data). This model, however, does not accurately reflect human CH because most HSCs express mutant ASXL1. Therefore, there is a need to assess whether a small population of cells expressing mutant ASXL1 acquire a competitive advantage in native hematopoiesis without transplantation. Collectively, our data suggest that the mutant ASXL1 knock-in mouse is useful for elucidating the pathogenesis of ASXL1 mutation-induced CH. Future research would reveal the precise mechanism underlying CH with ASXL1 mutations, which is expected to help the development of therapeutic interventions against CH.

      Concluding remarks

      ASXL1 is one of the most frequently mutated genes in CH, and its mutations are associated with poor prognosis in myeloid malignancies [
      • Bejar R
      • Stevenson K
      • Abdel-Wahab O
      • et al.
      Clinical effect of point mutations in myelodysplastic syndromes.
      ,
      • Chen TC
      • Hou HA
      • Chou WC
      • et al.
      Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome.
      ,
      • Thol F
      • Friesen I
      • Damm F
      • et al.
      Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes.
      ,
      • Gelsi-Boyer V
      • Trouplin V
      • Roquain J
      • et al.
      ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia.
      ,
      • Itzykson R
      • Kosmider O
      • Renneville A
      • et al.
      Prognostic score including gene mutations in chronic myelomonocytic leukemia.
      ,
      • Papaemmanuil E
      • Gerstung M
      • Bullinger L
      • et al.
      Genomic classification and prognosis in acute myeloid leukemia.
      ,
      • Metzeler KH
      • Becker H
      • Maharry K
      • et al.
      ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category.
      ]. Therefore, elucidating the mechanism by which ASXL1 mutations induce CH and subsequent MDS or AML would help prevent the development of hematologic malignancies.
      With the help of advanced DNA sequencing technologies, we delineated a catalogue of mutations in hematologic malignancies. Since then, several studies have revealed how these mutations contribute to the pathogenesis. Nevertheless, we cannot completely overcome these diseases at present. At the same time, much effort has been devoted to understanding their mutational history as far back as the preclinical stage, revealing the complex and dynamic changes of clones in the process of disease progression [
      • Lundberg P
      • Karow A
      Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms.
      ,
      • Yoshizato T
      • Dumitriu B
      • Hosokawa K
      • et al.
      Somatic mutations and clonal hematopoiesis in aplastic Anemia.
      ,
      • Abelson S
      • Collord G
      • Ng SWK
      • et al.
      Prediction of acute myeloid leukaemia risk in healthy individuals.
      ,
      • Jan M
      • Snyder TM
      • Corces-Zimmerman MR
      • et al.
      Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia.
      ,
      • Shlush LI
      • Zandi S
      • Mitchell A
      • et al.
      Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia.
      ,
      • Walter MJ
      • Shen D
      • Ding L
      • et al.
      Clonal architecture of secondary acute myeloid leukemia.
      ,
      • Ding L
      • Ley TJ
      • Larson DE
      • et al.
      Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing.
      ,
      • Makishima H
      • Yoshizato T
      • Yoshida K
      • et al.
      Dynamics of clonal evolution in myelodysplastic syndromes.
      ,
      • Desai P
      • Mencia-Trinchant N
      • Savenkov O
      • et al.
      Somatic mutations precede acute myeloid leukemia years before diagnosis.
      ]. Notably, CH is detected in virtually all people by middle age if we increase the number of sequence reads [
      • Young AL
      • Challen GA
      • Birmann BM
      • Druley TE
      Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults.
      ], suggesting that CH is a natural effect of aging on the hematopoietic system. So, when, why, and what clones with mutations evolve from CH into malignancy? Is there a clear distinction between CH and subsequent malignant transformation? To fully elucidate this problem, we will need to integratedly analyze multiple layers, including mutation, histone modifications, and gene expression, at a single-cell level and link them to clinical findings over time.
      Apart from CH, recent reports have revealed that normal tissues also accumulate somatic mutations with age, which may confer a selective advantage [
      • Suda K
      • Nakaoka H
      • Yoshihara K
      • et al.
      Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium.
      ,
      • Blokzijl F
      • De Ligt J
      • Jager M
      • et al.
      Tissue-specific mutation accumulation in human adult stem cells during life.
      ,
      • Lee-Six H
      • Øbro NF
      • Shepherd MS
      • et al.
      Population dynamics of normal human blood inferred from somatic mutations.
      ,
      • Yokoyama A
      • Kakiuchi N
      • Yoshizato T
      • et al.
      Age-related remodelling of oesophageal epithelia by mutated cancer drivers.
      ,
      • Yizhak K
      • Aguet F
      • Kim J
      • et al.
      RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues.
      ,
      • Martincorena I
      • Roshan A
      • Gerstung M
      • et al.
      High burden and pervasive positive selection of somatic mutations in normal human skin.
      ,
      • Martincorena I
      • Fowler JC
      • Wabik A
      • et al.
      Somatic mutant clones colonize the human esophagus with age.
      ]. However, the mutational landscapes are totally different. The abundance of mutations in epigenetic regulators is a characteristic feature of CH [
      • Xie M
      • Lu C
      • Wang J
      • et al.
      Age-related mutations associated with clonal hematopoietic expansion and malignancies.
      ,
      • Genovese G
      • Kähler AK
      • Handsaker RE
      • et al.
      Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
      ,
      • Jaiswal S
      • Fontanillas P
      • Flannick J
      • et al.
      Age-related clonal hematopoiesis associated with adverse outcomes.
      ], whereas mutations in signal transduction molecules, oncogenes, and tumor suppressors are more frequent in other tissues [
      • Suda K
      • Nakaoka H
      • Yoshihara K
      • et al.
      Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium.
      ,
      • Blokzijl F
      • De Ligt J
      • Jager M
      • et al.
      Tissue-specific mutation accumulation in human adult stem cells during life.
      ,
      • Yizhak K
      • Aguet F
      • Kim J
      • et al.
      RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues.
      ]. The reason for this discrepancy remains elusive. A better understanding of CH and subsequent hematologic malignancies would unveil what this difference means and further help us understand the basic principle of a clonal evolution.

      Conflict of interest disclosure

      The authors declare no conflicts of interest, financial or otherwise.

      Acknowledgments

      Many thanks to all lab members and our collaborators.

      References

        • Oni SB
        • Osunkoya BO
        • L L
        Paroxysmal nocturnal hemoglobinuria: evidence for monoclonal origin of abnormal red cells.
        Blood. 1970; 36: 145-152
        • Fey MF
        • Liechti-Gallati S
        • Rohr A Von
        • et al.
        Clonality and X-inactivation patterns in hematopoietic cell populations detected by the highly informative M27 beta DNA probe.
        Blood. 1994; 83: 931-938
        • Busque L
        • Mio R
        • Mattioli J
        • et al.
        Nonrandom X-inactivation patterns in normal females: lyonization ratios vary with age.
        Blood. 1996; 88: 59-65
        • Xie M
        • Lu C
        • Wang J
        • et al.
        Age-related mutations associated with clonal hematopoietic expansion and malignancies.
        Nat Med. 2014; 20: 1472-1478
        • Genovese G
        • Kähler AK
        • Handsaker RE
        • et al.
        Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence.
        N Engl J Med. 2014; 371: 2477-2487
        • Jaiswal S
        • Fontanillas P
        • Flannick J
        • et al.
        Age-related clonal hematopoiesis associated with adverse outcomes.
        N Engl J Med. 2014; 371: 2488-2498
        • Jaiswal S
        • Natarajan P
        • Silver AJ
        • et al.
        Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease.
        N Engl J Med. 2017; 377: 111-121
        • Wang W
        • Liu W
        • Fidler T
        • et al.
        Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in JAK2V617F mice.
        Circ Res. 2018; 123: E35-E47
        • Fuster Jose J
        • MacLauchlan S
        • Zuriaga MA
        • et al.
        Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice.
        Science. 2017; 847: 842-847
        • Scott BL
        • Deeg HJ
        Myelodysplastic syndromes.
        N Engl J Med. 2009; 361: 1872-1885
        • Bejar R
        • Stevenson K
        • Abdel-Wahab O
        • et al.
        Clinical effect of point mutations in myelodysplastic syndromes.
        N Engl J Med. 2011; 364: 2496-2506
        • Papaemmanuil E
        • Gerstung M
        • Malcovati L
        • et al.
        Clinical and biological implications of driver mutations in myelodysplastic syndromes.
        Blood. 2013; 122: 3616-3627
        • Haferlach T
        • Nagata Y
        • Grossmann V
        • et al.
        Landscape of genetic lesions in 944 patients with myelodysplastic syndromes.
        Leukemia. 2014; 28: 241-247
        • Boultwood J
        • Perry J
        • Pellagatti A
        • et al.
        Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia.
        Leukemia. 2010; 24: 1062-1065
        • Chen TC
        • Hou HA
        • Chou WC
        • et al.
        Dynamics of ASXL1 mutation and other associated genetic alterations during disease progression in patients with primary myelodysplastic syndrome.
        Blood Cancer J. 2014; 4 (e177–e177)
        • Thol F
        • Friesen I
        • Damm F
        • et al.
        Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes.
        J Clin Oncol. 2011; 29: 2499-2506
        • Gelsi-Boyer V
        • Trouplin V
        • Roquain J
        • et al.
        ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia.
        Br J Haematol. 2010; 151: 365-375
        • Itzykson R
        • Kosmider O
        • Renneville A
        • et al.
        Prognostic score including gene mutations in chronic myelomonocytic leukemia.
        J Clin Oncol. 2013; 31: 2428-2436
        • Itzykson R
        • Solary E
        An evolutionary perspective on chronic myelomonocytic leukemia.
        Leukemia. 2013; 27: 1441-1450
        • Vannucchi AM
        • Lasho TL
        • Guglielmelli P
        • et al.
        Mutations and prognosis in primary myelofibrosis.
        Leukemia. 2013; 27: 1861-1869
        • Nangalia J
        • Massie CE
        • Baxter EJ
        • et al.
        Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2.
        N Engl J Med. 2013; 369: 2391-2405
        • Lundberg P
        • Karow A
        Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms.
        Blood. 2014; 123: 2220-2228
        • Schischlik F
        • Kralovics R
        Mutations in myeloproliferative neoplasms—their significance and clinical use.
        Expert Rev Hematol. 2017; 10: 961-973
        • Papaemmanuil E
        • Gerstung M
        • Bullinger L
        • et al.
        Genomic classification and prognosis in acute myeloid leukemia.
        N Engl J Med. 2016; 374: 2209-2221
        • Mengistu G
        • Balcha F
        • Britton S
        Acute myeloid leukemia.
        N Engl J Med. 2015; 373: 1136-1152
        • Meyer SC
        • Levine RL
        Translational implications of somatic genomics in acute myeloid leukaemia.
        Lancet Oncol. 2014; 15: e382-e394
        • The Cancer Genome Atlas Research Network
        Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia.
        N Engl J Med. 2013; 368: 2059-2074
        • Welch JS
        • Ley TJ
        • Link DC
        • et al.
        The origin and evolution of mutations in acute myeloid leukemia.
        Cell. 2012; 150: 264-278
        • Marcucci G
        • Haferlach T
        • Döhner H
        Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications.
        J Clin Oncol. 2011; 29: 475-486
        • Metzeler KH
        • Becker H
        • Maharry K
        • et al.
        ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN favorable genetic category.
        Blood. 2011; 118: 6920-6929
        • Schnittger S
        • Eder C
        • Jeromin S
        • et al.
        ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome.
        Leukemia. 2013; 27: 82-91
        • Tyner JW
        • Tognon CE
        • Bottomly D
        • et al.
        Functional genomic landscape of acute myeloid leukaemia.
        Nature. 2018; 562: 526-531
        • Gerstung M
        • Pellagatti A
        • Malcovati L
        • et al.
        Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes.
        Nat Commun. 2015; 6: 5901
        • Yoshizato T
        • Dumitriu B
        • Hosokawa K
        • et al.
        Somatic mutations and clonal hematopoiesis in aplastic Anemia.
        N Engl J Med. 2015; 373: 35-47
        • Fisher CL
        • Berger J
        • Randazzo F
        • Brock HW
        A human homologue of Additional sex combs, ADDITIONAL SEX COMBS-LIKE 1, maps to chromosome 20q11.
        Gene. 2003; 306: 115-126
        • Chou WC
        • Chou SC
        • Liu CY
        • et al.
        TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics.
        Blood. 2011; 118: 3803-3810
        • Tan M
        • Ng IKS
        • Chen Z
        • et al.
        Clinical implications of DNMT3A mutations in a Southeast Asian cohort of acute myeloid leukaemia patients.
        J Clin Pathol. 2017; 70: 669-676
        • Hou HA
        • Kuo YY
        • Liu CY
        • et al.
        DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications.
        Blood. 2011; 118: 559-569
        • Chou WC
        • Huang HH
        • Hou HA
        • et al.
        Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations.
        Blood. 2010; 116: 4086-4094
        • Balasubramani A
        • Larjo A
        • Bassein JA
        • et al.
        Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex.
        Nat Commun. 2015; 6: 1-15
        • Sanchez R
        • Zhou MM
        The PHD finger: a versatile epigenome reader.
        Trends Biochem Sci. 2011; 36: 364-372
        • Kennison JA
        • Tamkun JW
        Dosage-dependent modifiers of Polycomb and Antennapedia mutations in Drosophila.
        Proc Natl Acad Sci USA. 1988; 85: 8136-8140
        • Simon J
        • Chiang A
        • Bender W
        Ten different Polycomb group genes are required for spatial control of the abdA and AbdB homeotic products.
        Development. 1992; 114: 493-505
        • Kennison JA
        Transcriptional activation of Drosophila homeotic genes form distant regulatory elements.
        Trends Genet. 1993; 9: 75-79
        • Pirrotta V
        PcG complexes and chromatin silencing.
        Curr Opin Genet Dev. 1997; 7: 249-258
        • Schuettengruber B
        • Bourbon HM
        • Di Croce L
        • Cavalli G
        Genome regulation by Polycomb and Trithorax: 70 years and counting.
        Cell. 2017; 171: 34-57
        • Wang H
        • Wang L
        • Erdjument-Bromage H
        • Vidal M
        • Tempst P
        • Jones RS ZY
        Role of histone H2A ubiquitination in Polycomb silencing.
        Nature. 2004; 431: 873-878
        • de Napoles M
        • Mermoud JE
        • Wakao R
        • et al.
        Polycomb group proteins ring1A/B link ubiquitylation of histone H2A to heritable gene silencing and X inactivation.
        Dev Cell. 2004; 7: 663-676
        • Wang L
        • Brown JL
        • Cao R
        • Zhang Y
        • Kassis JA
        • Jones RS
        Hierarchical recruitment of polycomb group silencing complexes.
        Mol Cell. 2004; 14: 637-646
        • Milne TA
        • Sinclair DAR
        • Brock HW
        The Additional sex combs gene of Drosophila is required for activation and repression of homeotic loci, and interacts specifically with Polycomb and super sex combs.
        Mol Gen Genet. 1999; 261: 753-761
        • Abdel-Wahab O
        • Adli M
        • LaFave LM
        • et al.
        ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression.
        Cancer Cell. 2012; 22: 180-193
        • Scheuermann JC
        • De Ayala Alonso AG
        • Oktaba K
        • et al.
        Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB.
        Nature. 2010; 465: 243-247
        • Wang J
        • Li Z
        • He Y
        • et al.
        Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice.
        Blood. 2014; 123: 541-553
        • Inoue D
        • Fujino T
        • Sheridan P
        • et al.
        A novel ASXL1–OGT axis plays roles in H3K4 methylation and tumor suppression in myeloid malignancies.
        Leukemia. 2018; 32: 1327-1337
        • Fisher CL
        • Pineault N
        • Brookes C
        • et al.
        Loss-of-function additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia.
        Blood. 2010; 115: 38-46
        • Abdel-Wahab O
        • Gao J
        • Adli M
        • et al.
        Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo.
        J Exp Med. 2013; 210: 2641-2659
        • Ley TJ
        • Ding L
        • Walter MJ
        • et al.
        DNMT3A mutations in acute myeloid leukemia.
        N Engl J Med. 2010; 363: 2424-2433
        • Inoue D
        • Matsumoto M
        • Nagase R
        • et al.
        Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels.
        Exp Hematol. 2016; 44: 172-176
        • Inoue D
        • Kitaura J
        • Togami K
        • et al.
        Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations.
        J Clin Invest. 2013; 123: 4627-4640
        • Asada S
        • Goyama S
        • Inoue D
        • et al.
        Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis.
        Nat Commun. 2018; 9: 1-18
        • Yang H
        • Kurtenbach S
        • Guo Y
        • et al.
        Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies.
        Blood. 2018; 131: 328-341
        • Hsu YC
        • Chiu YC
        • Lin CC
        • et al.
        The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model.
        J Hematol Oncol. 2017; 10: 1-15
        • Uni M
        • Masamoto Y
        • Sato T
        • Kamikubo Y
        • Arai S
        • Kurokawa M
        Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification.
        Leukemia. 2018; 33: 191-204
        • Nagase R
        • Inoue D
        • Pastore A
        • et al.
        Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.
        J Exp Med. 2018; 215: 1729-1747
        • Challen GA
        • Sun D
        • Jeong M
        • et al.
        Dnmt3a is essential for hematopoietic stem cell differentiation.
        Nat Genet. 2012; 44: 23-31
        • Challen GA
        • Sun D
        • Mayle A
        • et al.
        Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells.
        Cell Stem Cell. 2014; 15: 350-364
        • Quivoron C
        • Couronné L
        • Della Valle V
        • et al.
        TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis.
        Cancer Cell. 2011; 20: 25-38
        • Moran-Crusio K
        • Reavie L
        • Shih A
        • et al.
        Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation.
        Cancer Cell. 2011; 20: 11-24
        • Sun D
        • Luo M
        • Jeong M
        • et al.
        Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal.
        Cell Stem Cell. 2014; 14: 673-688
        • Cheung P
        • Vallania F
        • Warsinske HC
        • et al.
        Single-cell chromatin modification profiling reveals increased epigenetic variations with aging.
        Cell. 2018; 173 (e14): 1385-1397
        • Hannum G
        • Guinney J
        • Zhao L
        • et al.
        Genome-wide methylation profiles reveal quantitative views of human aging rates.
        Mol Cell. 2013; 49: 359-367
        • Fraga MF
        • Ballestar E
        • Paz MF
        • et al.
        Epigenetic differences arise during the lifetime of monozygotic twins.
        Proc Natl Acad Sci USA. 2005; 102: 10604-10609
        • Boks MP
        • Derks EM
        • Weisenberger DJ
        • et al.
        The relationship of DNA methylation with age, gender and genotype in twins and healthy controls.
        PLoS One. 2009; 4: 21-23
        • Coombs CC
        • Zehir A
        • Devlin SM
        • et al.
        Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes.
        Cell Stem Cell. 2017; 21 (e4): 374-382
        • Hsu JI
        • Dayaram T
        • Tovy A
        • et al.
        PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy.
        Cell Stem Cell. 2018; 23 (e6): 700-713
        • Lindsley RC
        • Saber W
        • Mar BG
        • et al.
        Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation.
        N Engl J Med. 2017; 376: 536-547
        • Christiansen DH
        • Andersen MK
        • Pedersen-Bjergaard J
        Mutations with loss of heterozygosity of p53 are common in therapy-related myelodysplasia and acute myeloid leukemia after exposure to alkylating agents and significantly associated with deletion or loss of 5q, a complex karyotype, and a poor prognosis.
        J Clin Oncol. 2001; 19: 1405-1413
        • Kahn JD
        • Miller PG
        • Silver AJ
        • et al.
        PPM1D-truncating mutations confer resistance to chemotherapy and sensitivity to PPM1D inhibition in hematopoietic cells.
        Blood. 2018; 132: 1095-1105
        • Gibson CJ
        • Lindsley RC
        • Tchekmedyian V
        • et al.
        Clonal hematopoiesis associated with adverse outcomes after autologous stem-cell transplantation for lymphoma.
        J Clin Oncol. 2017; 35: 1598-1605
        • Takahashi K
        • Wang F
        • Kantarjian H
        • et al.
        Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case–control study.
        Lancet Oncol. 2017; 18: 100-111
        • Tenedini E
        • Bernardis I
        • Artusi V
        • et al.
        Targeted cancer exome sequencing reveals recurrent mutations in myeloproliferative neoplasms.
        Leukemia. 2014; 28: 1052-1059
        • Youn HS
        • Kim TY
        • Park UH
        • et al.
        Asxl1 deficiency in embryonic fibroblasts leads to cellular senescence via impairment of the AKT-E2F pathway and Ezh2 inactivation.
        Sci Rep. 2017; 7: 1-13
        • Young NS
        Aplastic anemia.
        N Engl J Med. 2018; 379: 1643-1656
        • Abelson S
        • Collord G
        • Ng SWK
        • et al.
        Prediction of acute myeloid leukaemia risk in healthy individuals.
        Nature. 2018; 559: 400-404
        • Jan M
        • Snyder TM
        • Corces-Zimmerman MR
        • et al.
        Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia.
        Sci Transl Med. 2012; 4: 149ra118
        • Shlush LI
        • Zandi S
        • Mitchell A
        • et al.
        Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia.
        Nature. 2014; 506: 328-333
        • Walter MJ
        • Shen D
        • Ding L
        • et al.
        Clonal architecture of secondary acute myeloid leukemia.
        N Engl J Med. 2012; 366: 1090-1098
        • Ding L
        • Ley TJ
        • Larson DE
        • et al.
        Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing.
        Nature. 2012; 481: 506-510
        • Makishima H
        • Yoshizato T
        • Yoshida K
        • et al.
        Dynamics of clonal evolution in myelodysplastic syndromes.
        Nat Genet. 2017; 49: 204-212
        • Desai P
        • Mencia-Trinchant N
        • Savenkov O
        • et al.
        Somatic mutations precede acute myeloid leukemia years before diagnosis.
        Nat Med. 2018; 24: 1015-1023
        • Young AL
        • Challen GA
        • Birmann BM
        • Druley TE
        Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults.
        Nat Commun. 2016; 7: 1-7
        • Suda K
        • Nakaoka H
        • Yoshihara K
        • et al.
        Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium.
        Cell Rep. 2018; 24: 1777-1789
        • Blokzijl F
        • De Ligt J
        • Jager M
        • et al.
        Tissue-specific mutation accumulation in human adult stem cells during life.
        Nature. 2016; 538: 260-264
        • Lee-Six H
        • Øbro NF
        • Shepherd MS
        • et al.
        Population dynamics of normal human blood inferred from somatic mutations.
        Nature. 2018; 561: 473-478
        • Yokoyama A
        • Kakiuchi N
        • Yoshizato T
        • et al.
        Age-related remodelling of oesophageal epithelia by mutated cancer drivers.
        Nature. 2019; 565: 312-317
        • Yizhak K
        • Aguet F
        • Kim J
        • et al.
        RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues.
        Science. 2019; 364
        • Martincorena I
        • Roshan A
        • Gerstung M
        • et al.
        High burden and pervasive positive selection of somatic mutations in normal human skin.
        Science. 2015; 348: 880-886
        • Martincorena I
        • Fowler JC
        • Wabik A
        • et al.
        Somatic mutant clones colonize the human esophagus with age.
        Science. 2018; 362: 911-917