Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2017

Open Access 01.12.2017 | Research

The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model

verfasst von: Yueh-Chwen Hsu, Yu-Chiao Chiu, Chien-Chin Lin, Yuan-Yeh Kuo, Hsin-An Hou, Yi-Shiuan Tzeng, Chein-Jun Kao, Po-Han Chuang, Mei-Hsuan Tseng, Tzu-Hung Hsiao, Wen-Chien Chou, Hwei-Fang Tien

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2017

Abstract

Background

Additional sex combs-like 1 (ASXL1) is frequently mutated in myeloid malignancies. Recent studies showed that hematopoietic-specific deletion of Asxl1 or overexpression of mutant ASXL1 resulted in myelodysplasia-like disease in mice. However, actual effects of a “physiological” dose of mutant ASXL1 remain unexplored.

Methods

We established a knock-in mouse model bearing the most frequent Asxl1 mutation and studied its pathophysiological effects on mouse hematopoietic system.

Results

Heterozygotes (Asxl1 tm/+ ) marrow cells had higher in vitro proliferation capacities as shown by more colonies in cobblestone-area forming assays and by serial re-plating assays. On the other hand, donor hematopoietic cells from Asxl1 tm/+ mice declined faster in recipients during transplantation assays, suggesting compromised long-term in vivo repopulation abilities. There were no obvious blood diseases in mutant mice throughout their life-span, indicating Asxl1 mutation alone was not sufficient for leukemogenesis. However, this mutation facilitated engraftment of bone marrow cell overexpressing MN1. Analyses of global gene expression profiles of ASXL1-mutated versus wild-type human leukemia cells as well as heterozygote versus wild-type mouse marrow precursor cells, with or without MN1 overexpression, highlighted the association of in vivo Asxl1 mutation to the expression of hypoxia, multipotent progenitors, hematopoietic stem cells, KRAS, and MEK gene sets. ChIP-Seq analysis revealed global patterns of Asxl1 mutation-modulated H3K27 tri-methylation in hematopoietic precursors.

Conclusions

We proposed the first Asxl1 mutation knock-in mouse model and showed mutated Asxl1 lowered the threshold of MN1-driven engraftment and exhibited distinct biological functions on physiological and malignant hematopoiesis, although it was insufficient to lead to blood malignancies.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13045-017-0508-x) contains supplementary material, which is available to authorized users.
Abkürzungen
AML
Acute myeloid leukemia
Asxl1 tm/+
Asxl1 G643WfsX12 heterozygous mice
BMC
Bone marrow cells
CAFC
Cobblestone-area-forming cell assay
CFC
Colony-forming cell
ChIP-Seq
Chromatin immunoprecipitation sequencing
CMP
Common myeloid progenitors
CRU
Competitive repopulating unit assay
GMP
Granulocyte-monocytic progenitors
GSEA
Gene Set Enrichment Analysis
H3K27me3
Tri-methylation of Histone 3 at lysine 27
HPC
Hematopoietic progenitors
HSC
Hematopoietic stem cells
HSPC
Hematopoietic stem cells and progenitors
LK
lin-c-Kit+Sca-1- cells
LSK
lin-c-Kit+Sca-1+ cells
LSK-
lin-c-Kit-Sca-1+ cells
LTC-IC
Long-term culture-initiating cells
MDS
Myelodysplastic syndrome
MEP
Megakaryocyte-erythroid progenitors
WT
Wild-type control mice

Background

Additional sex combs-like 1 (ASXL1) is the human homolog of Drosophila additional sex combs (Asx) [1], frequently mutated in acute myeloid leukemia (AML) and other myeloid malignancies [24]. Germline heterozygous nonsense mutation of ASXL1 results in Bohring-Opitz syndrome, a congenital disease with multi-system developmental abnormalities [5]. ASXL1 binds a deubiquitinase BAP1 to form a critical complex for H2A K119 deubiquitination through the catalysis of polycomb repressive complex 1 [6, 7]. The deubiquitination activity is enhanced when BAP1 is complexed with truncated form of ASXL1 [8]. BAP1 deletion produces phenotypes mimicking human chronic myelomonocytic leukemia in mice [9]. Thus, it is likely that ASXL1-BAP1 axis is important to prevent leukemogenesis [9].
We previously analyzed the clinical implications of ASXL1 mutation in a large cohort of patients and found that this mutation occurred in 10.8% (54/501) of de novo AML patients and predicted a shorter survival [10]. Several studies also showed that ASXL1 mutation was a poor prognostic factor in myeloid malignancies [1014].
Since the discovery of ASXL1 mutation in myeloid malignancies in 2009 [15], many studies about its pathophysiology have been reported. However, controversies exist among these reports. For example, in vivo deletion of Asxl1 was shown to result in subtle phenotypes including defects in the frequencies of myeloid and lymphoid cells in blood, marrow or other hematopoietic organs in mice but not myelodysplastic syndrome (MDS) or leukemia [16]. However, in other studies, knockout of Asxl1 led to systemic developmental defects including MDS-like presentation, with alteration of the self-renewal and repopulation capacities of the mutant hematopoietic stem/progenitor cells and global reduction of H3K27 tri-methylation (H3K27me3) [17, 18].
The pathophysiological effect of ASXL1 truncation mutations in human myeloid malignancies is another matter of debate. For example, it was suggested that ASXL1 mutation was a loss-of-function mutation because of failure in detecting mutant protein in human leukemia cells [14]. However, the findings that overexpression of truncating mutation in hematopoietic cells of mice displayed human MDS features with de-repression of Hoxa9 in another study [19] and detectability of truncating proteins in human cell lines bearing ASXL1 truncating mutations argued for gain-of-function or dominant negative effects of ASXL1 mutations [19, 20]. These controversies are likely due to different methods of genetic engineering of the animals or forced overexpression of the mutation. Overall, the pathophysiological alterations in human acute myeloid leukemia (AML) cells bearing ASXL1 mutations have not been explored systematically.
To overcome these problems, we generated and analyzed a mouse model bearing human-like Asxl1 mutation followed by extensive phenotypic and molecular characterizations on this mouse model. In our model, the Asxl1 mutation was knocked in to the endogenous Asxl1 allele, thus the mice have “physiological dose” of mutation, as we see in the patients. For translating to clinical situations, we also investigated the global expression profiles of our large AML cohort to delineate the pathophysiology related to ASXL1 mutations. We found that bone marrow cells from Asxl1 heterozygotes formed more colonies in cobblestone-area-forming assays and the ability to form colonies persisted longer in serial colony-forming cell assays. On the other hand, in vivo transplantation assays showed that donor bone marrow cells from Asxl1 mutant mice declined faster in their recipients than those from the wild-type mice. While forced overexpression of mutant Asxl1 in mouse bone marrow hematopoietic cells could lead to MDS-like disease [19], our mice bearing a “physiological dose” of mutant Asxl1 did not show obvious trend of developing blood diseases throughout their life span. However, with overexpression of MN1, mutant Asxl1 hematopoietic stem cells and progenitors (HSPCs) were more likely to engraft in recipient mice than wild-type HSPCs, suggesting that Asxl1 mutation could lower the threshold of engraftment driven by MN1 overexpression. Global expression profiling in mutant versus wild-type Asxl1 mouse cells as well as in ASXL1-mutated versus wild-type human AML cells, with or without concurrent MN1 overexpression, disclosed pathophysiological pathways involved in Asxl1 mutation. ChIP-Seq experiments showed global Asxl1 mutation-modulated H3K27me3 patterns in HSPCs.

Methods

Generation of Asxl1 mutation knock-in mice

The cognate mouse mutation is predicted to be c.1925dupG; p.G643WfsX12, encoding 654 amino acids mimicking the most common form of human mutant ASXL1 protein, compared to 1514 residues in wild-type Asxl1 protein. Potential chimeras were crossed with wild-type C57BL/6 mice to facilitate the confirmation of germ-line transmission, their offspring who harbored Asxl1 mutation were backcrossed with C57BL/6 to generate inbred strains then maintained at C57BL/6 background. Heterozygous mice were mated with wild-type mice to get heterozygous mice and littermate control mice. Heterozygous mice were mated with each other to get homozygous mice. Mice between 2- to 6-month age were used for experiment except those were assigned to long-term observation cohort. All animals were housed in specific pathogen-free animal facility and all procedures were approved by IACUC of the National Taiwan University College of Medicine.

Chromatin immunoprecipitation sequencing (ChIP-Seq)

We used Lin- bone marrow cells as a surrogate to identify genome-wide histone modification affected by Asxl1 mutations. Chromatin lysate was harvested and sonicated with a sonicator (Bioruptor®Pico) to shear the DNA into a length ~200 bp, then it was hybridized with anti-H3K27me3 (Millipore, Germany). Immunoprecipitated DNA was sent to the National Center for Genome Medicine and sequenced by Illumina HiSeq 2000 sequencer with 100 × 2 bp paired-end sequencing.

ChIP-Seq data analysis

Sequencing reads were aligned to the mm10 mouse reference genome by Burrows-Wheeler Alignment tool (BWA; version 0.7.15). We used the Model-based Analysis of ChIP-Seq tool (MACS2) to detect peaks of reads between sample and input sequences in ASXL1 tm/+ and wild-type bone marrow cells. ASXL1 tm/+ (or wild-type) specific peaks were called by intersecting the identified peaks with BEDTools v2.17.0 [21]. These condition-specific peaks were annotated by Peak Annotation and Visualization (PAVIS) and analyzed for the enrichments in gene regions [22]. To realize the biological functions governed by the interaction of Asxl1 mutation and H3K27me3, we analyzed peaks-associated genes by The Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8 with default settings [23, 24]. Furthermore, we performed motif analysis on sequences around the condition-specific peaks (±250 bps from peak center) by MEME-ChIP web tool included in the MEME Suite [25, 26]. Sequencing reads and identified peaks were visualized with Integrative Genome Viewer (IGV) [27, 28].

Statistical analysis

In vitro and in vivo experiments were performed at least three times independently. Data were processed in Microsoft Excel or GraphPad Prism software. Student’s t test, paired t test, ANOVA or chi-square test were used to compare the differences in values between groups.
For the other experimental procedures, please see the Additional file 1.

Results

Generation of Asxl1 G643WfsX12 gene knock-in mice

In human AML, the most common mutation is c.1934dupG; p.G646WfsX12 (up to 66%) [10]. The mouse and human ASXL1 proteins share 74% identity in amino acid sequence. The changed amino acid G646 is within a stretch of highly conserved region ATTAIGGGGGPGGGG (designated as a bold and underlined G) [10]. An additional guanine was inserted into this 8-G cassette located at mouse Asxl1 exon 13 to mimic this frequent human ASXL1 mutation (Fig. 1a). This insertion causes frame shift in the reading frame and introduces a premature stop codon so that the mutant Asxl1 would be shorter than wild-type form and lack the c-terminal region which contains a plant homeodomain (PHD). Therefore, the cognate mouse mutation c.1925dupG; p.G643WfsX12 is expected to bear similar pathophysiological consequence as human’s. In our mouse model, mutant Asxl1 expression was driven by the endogenous Asxl1 promoter. Therefore, mutant Asxl1 would be expressed identically as the endogenous Asxl1, not restrictive to hematopoietic cells (Fig. 1a).
The additional guanine inserted into the 8-G cassette at exon 13 was confirmed by DNA sequencing (Fig. 1b). Both Asxl1 G643WfsX12 heterozygotes and homozygotes were fertile. The pups’ genotypes fit Mendelian ratio through gestation period till birth (Additional file 1: Figure S1). Homozygous new-born mice suffered from high rate of post-natal death, with only 7% of viability after weaning. Autopsy of the dead new born mice did not show any obvious organ abnormalities (data not shown). Lack of nursing was probably the main reason of these post-natal lethal events, but the exact causes remains to be elucidated. Due to the difficulty in gathering sufficient mice for observation, we hence focused our long-term observation on heterozygous and wild-type mice.

Asxl1 tm/+ hematopoietic cells had higher short-term in vitro proliferation capacities

We initially performed several in vitro assays to evaluate the population frequency and differentiation potencies of HSPCs in the bone marrow of Asxl1 tm/+ and wild-type mice. We first used cobblestone-area forming cell (CAFC) assays to evaluate the frequencies of hematopoietic precursor cells in bone marrows. The cobblestone areas were counted one week after seeding and we found that Asxl1 tm/+ cells formed more cobblestone areas than Asxl1-wild cells (N = 3 each, p = 0.028) (Fig. 2a). Colony-forming cell (CFC) assay were also performed to test the effects of Asxl1 mutant on cell differentiation. Serial plating was performed every 7 days to estimate population frequencies of hematopoietic precursors in the initial plating. In initial plating, Asxl1 tm/+ and wild-type bone marrow cells formed similar numbers of each type of colonies. However, total colony number as well as granulocyte colonies (CFU-G) formed by Asxl1 tm/+ cells were more than wild-type cells in second plating and this trend last to the third plating (Fig. 2b, Additional file 1: Figure S2A to S2C). These results indicate that mutated Asxl1 confers stronger short-term in vitro proliferation capabilities to hematopoietic precursors than the wild-type Asxl1.

Bone marrow cells of Asxl1 G643WfsX12 heterozygotes showed compromised long-term in vivo repopulation and self-renewal capabilities

To evaluate the in vivo influence of Asxl1 mutation on hematopoiesis in a long-term basis, we employed bone marrow transplantation assays. Five hundred Lin-c-Kit+Sca-1+ (LSK) bone marrow cells sorted from Asxl1 mutant and wild-type mice, respectively, together with 200,000 helper cells, were transplanted into wild-type recipient mice for competitive repopulation unit assays. The peripheral blood of the transplanted mice was sampled monthly and evaluated for reconstitution efficiency in a 4-month period. We found that recipient mice transplanted with LSK bone marrow cells from Asxl1 tm/+ donors had less donor-derived cells in peripheral blood and marrow when compared to those receiving wild-type LSK bone marrow cells (Fig. 3a). Interestingly, B cells in the peripheral blood of Asxl1 tm/+ mice were particularly reduced (Fig. 3c). These data suggest that Asxl1 mutant LSK cells have reduced in vivo long-term repopulation capacities compared with wild-type LSK cells.
Next, we performed serial bone marrow transplantation assays to rigorously test the potency of in vivo self-renewal ability of the Asxl1 tm/+ HSPCs. In this setting, whole bone marrow cells were serially transplanted into recipients without helper cells. To evaluate the reconstitution efficiency, peripheral blood of the recipient mice transplanted with either Asxl1 tm/+ bone marrow cells or wild-type bone marrow cells were sampled 2 months after every round of transplantation. We found that the frequencies of total cells and T cells, but not B or myeloid cells, in the recipient mice’s peripheral blood derived from Asxl1 tm/+ mice declined faster compared with those derived from wild-type controls (Fig. 4). The results suggest that Asxl1 mutation renders a compromised long-term in vivo self-renewal capability in a variety of lineages compared to wild-type cells in vivo.

The HSPC components of Asxl1 G643WfsX12 heterozygous mice were largely similar to those of the wild-type littermates

The amount of HSPCs in the bone marrow of Asxl1 tm/+ and wild-type littermates were analyzed and compared by FACS analysis. We noted that bone marrow LSK cells, long-term (Lin-Sca-1+c-Kit+CD150+CD48-) and short-term hematopoietic stem cells (Lin-Sca-1+c-Kit+CD150+CD48+), multipotent progenitors (Lin-Sca-1+c-Kit+CD150-CD48+), common myeloid progenitors (CMP, Lin-Sca-1-c-Kit+CD34+FcγRlo), granulocyte-monocytic progenitors (GMP, Lin-Sca-1-c-Kit+CD34+FcγRhi), and megakaryocyte-erythroid progenitors (MEP, Lin-Sca-1-c-Kit+CD34-FcγRlo) were all not different between the Asxl1 tm/+ heterozygotes and the wild-type mice (Additional file 1: Figure S2D). These data suggest that Asxl1 mutation does not affect the amount of hematopoietic cell components in vivo by surface marker analysis, although both in vitro and in vivo experiments indicate presence of its biological activities in Asxl1 tm/+ bone marrow cells as shown above.

Asxl1 G643WfsX12 alone was not sufficient for development of blood malignancies in mice

A cohort of Asxl1 tm/+ and wild-type control mice were collected to observe the influence of Asxl1 mutation on overall health status in an 18- to 24-month period (N = 33 for Asxl1 tm/+ mice and N = 38 for wild-type controls). Heterozygous mice were significantly lighter than wild-type mice (Fig. 5a). While there were no significant differences in hemograms in the peripheral blood or marrow hematopoietic components in younger mice, there were subtle hematopoietic phenotypes when the mice were old at 18 months. Old male (18-month age) Asxl1 tm/+ mice had higher WBC (p = 0.0091) and RBC counts (p = 0.03893) (Fig. 5b). There were more T cells in bone marrows of Asxl1 tm/+ mice (p = 0.049) than wild-type controls, while both mice had similar frequency of HSPCs (Fig. 5c, Additional file 1: Figure S3). Within the Lin-c-Kit+Sca-1- (LK) bone marrow cells, there was no significant difference in the proportion of CMP, GMP, and MEP between Asxl tm/+ and wild-type mice (Fig. 5d). The autopsy also did not show any difference in the incidence of splenomegaly between the two groups. The only six viable Asxl1 homozygous mice did not exhibit obvious abnormalities in hemogram or in autopsy findings at age of 18 months (Additional file 1: Figure S4). During the life span of the mice, Asxl1 G643WfsX12 showed no tendency to induce any kind of blood malignancies; hence, we concluded that Asxl1 G643WfsX12 alone was not sufficient for leukemogenesis in vivo.

Asxl1 G643WfsX12 lowered the engraftment threshold of MN1-overexpressing cells

Since Asxl1 mutation alone did not produce obvious blood diseases in mice, we sought to determine if this mutation functions as a facilitator for leukemogenesis. In our patients with array data (N = 349, among whom the mutation status of ASXL1 was known in 343) [2931], we noted that those bearing ASXL1 mutation tended to have higher MN1 expression (P = 0.056, Fig. 6a). Moreover, among the 225 patients who received standard chemotherapy, those with higher MN1 expression (≥ median) as well as ASXL1 mutation had shorter overall survival compared to those with higher MN1 expression but without ASXL1 mutation (Fig. 6b), suggesting a possible cooperative effect between these two genetic aberrancies in AML patients. MN1 over-expression is a sufficient driving event for mouse leukemogenesis [32]. Therefore, we were interested to know whether Asxl1 mutation facilitates the engraftment of MN1 over-expression in mice. To this end, we overexpressed MN1 in Asxl1 tm/+ or wild-type Lin- bone marrow cells by retroviral transduction. Proliferation of wild-type and Asxl1 tm/+ cells were not different as examined by BrdU incorporation assays (Fig. 6c). When MN1 was transduced into Asxl1 tm/+ and wild-type bone marrow cells, respectively, we still could not observe significant difference in proliferation rate between these two types of cells (Fig. 6d). However, long-term culture-initiation cell (LTC-IC) assay showed that when MN1 was overexpressed in Asxl1 tm/+ bone marrow cells, there was higher percentage of long-term colony forming cells compared to MN1 overexpressed wild-type bone marrow cells (Fig. 6e), implying that Asxl1 mutation promoted stem cell activities of marrow cells in MN1 overexpression background. To test this hypothesis, we transplanted several different doses of MN1-transduced Lin- bone marrow cells, together with 200,000 helper cells, into lethally irradiated recipients. Bone marrow cells of the recipient mice were harvested between 4 to 5 weeks after transplantation to evaluate the reconstitution efficiency. More than 1% MN1 over-expressing cells in the marrow cells of recipient mice was defined to be successfully reconstituted. At 5000-cell dose, 100% of recipient mice transplanted with either MN1 overexpressed Asxl1 mutant or wild-type bone marrow cells were successfully reconstituted. However, while most recipient mice transplanted with low-dose MN1-transduced cells could be reconstituted in the presence of Asxl1 mutation (9 out of 11 at 1000 test cells and 7 out of 7 at 500 test cells were successfully reconstituted), significantly lower proportion of recipient mice transplanted with the same doses of MN1-transduced cells without Asxl1 mutation were successfully reconstituted (7 out of 12 at 1000 test cells and 1 out of 6 at 500 test cells, p = 0.036 by Chi-square test) (Table 1 and Additional file 1: Figure S5). Our results suggest that Asxl1 G643WfsX12 can lower the threshold of MN1-driven engraftment.
Table 1
The number of mice successfully reconstituted in transplantation assay of MN1 overexpressed cells with WT or mutant Asxl1
Cell dose
WT + MN1
Asxl1 tm/+ +MN1
Number tested
Number reconstituted
Number tested
Number reconstituted
5000
6
6
8
8
1000
12
7
11
9
500
6
1
7
7
p = 0.036 by chi square test
Abbreviations: WT Asxl1 wild type, MN1 MN1 overexpression

Microarray analyses showed the cooperative effects of Asxl1 mutation and MN1 overexpression

Our mouse model provided an ideal platform to investigate the impacts of Asxl1 mutation per se on global gene expression patterns, since the genetic backgrounds of our mice were far less complicated than those in human leukemia cells. In addition, we were interested in the mechanisms underlying the supportive role of Asxl1 mutation in the engraftment of MN1 overexpressing bone marrow cells. To these ends, we collected Lin- marrow cells from Asxl1 tm/+ and wild-type control mice with or without MN1 transduction (wild-type, Asxl1 mutation, wild-type + MN1 overexpression, and Asxl1 mutation + MN1 overexpression) for microarray analyses to explore differential gene expression patterns as well as molecular functions conferred by Asxl1 mutation per se and/or its interplay with MN1 overexpression. Of note, only 4.6% genes were significantly differentially expressed between Asxl1-mutated and wild-type cells (Fig. 7a; left bar); the number of perturbed functional gene sets was also very modest (3.2%, comparing Asxl1 mutation vs. wild-type cells, Fig. 7b; left bar). However, the differences became obvious when comparing Asxl1-mutated cells overexpressing MN1 vs. wild-type cells overexpressing MN1 cells: up to 12.2% differentially expressed genes among all genes (Fig. 7a, second bar from the left), higher than that achieved by randomly shuffling the microarray dataset for 100 times (empirical p < 0.01), with correspondingly large scale of perturbed gene sets, up to 23.9% (Fig. 7b, second left bar). These data were consistent with our findings that Asxl1 alone did not render obvious blood diseases in the mice but it might play a cooperative role with MN1.
To compare our mouse model with human disease, we profiled gene expression of leukemia cells from a total of 343 AML patients and compared the expression patterns between samples with (N = 50) and without (N = 293) ASXL1 mutation. For 172 AML patients with higher MN1 expression (above the median level), we also compared the expression patterns between those with (N = 29) and without (N = 143) ASXL1 mutation. The disturbance of global gene expression profiles and gene sets related to Asxl1 mutation were quite comparable and obvious in total cohort (Fig. 7a, right two bars) and in the subgroup of patients with higher MN1 expression (Fig. 7b, right two bars).

Gene set enrichment analysis revealed oncogenic functions perturbed by the interaction between Asxl1 mutation and MN1 overexpression

A deeper look into the lists of significantly differential gene sets derived by GSEA revealed a handful of crucial oncogenic functions perturbed by the interaction between Asxl1 mutation and MN1 overexpression. Hypoxia-related genes were implied to be relevant factors of leukemogenesis [33, 34]. In our data, while the expression of genes of a hypoxia signature did not show an overall change in mice with Asxl1 mutation vs. wild-type littermates (GSEA p = 0.272; Fig. 7c), they were significantly co-upregulated in Asxl1 tm/+ , compared to wild-type mice, in the presence of MN1-overexpression (p = 0.007; Fig. 7d). Similar enrichments were seen in signatures representing multipotent progenitors and hematopoietic stem cells, as well as genes related to oncogenic KRAS and MEK, in an MN1-dependent manner (all p values <0.0005 in Asxl1 tm/+ vs. wild-type mice transduced with MN1, Fig. 7d; compared with p > 0.05 in Asxl1 mutation vs. wild-type mice without MN1 overexpression, except for genes related to MEK, Fig. 7c, p value 0.012). Such positive enrichment toward Asxl1 mutation was corroborated in AML patients, regardless of the abundance of MN1 (all p values <0.0005; Fig. 7e and f). In aggregate, the logistic relationship between Asxl1 mutation and MN1 overexpression is summarized as (1) Asxl1 mutation promoted engraftment of bone marrow cells in MN1 overexpression background (Fig. 6e and Table 1); (2) AML patients with both ASXL1 mutation and high MN1 expression had inferior survival when compared with ASXL1-wild-type and high MN1 expression (Fig. 6b); (3) In the background of MN1 overexpression, Asxl1 mutation in mice and in human AML patients was associated with upregulation of signatures of hematopoietic stem/progenitor cells and related to hypoxia, KRAS, and MEK pathways.

ChIP-Seq analysis revealed Asxl1 mutation-modulated binding profiles of H3K27me3

Several studies have linked functions of Asxl1 mutation to H3K27me3, an inactive mark associated with transcriptional repression [19]. In order to investigate their interactions in our mouse model, we performed histone extraction followed by western hybridization to evaluate the global H3K27me3 levels in bone marrow cells. There was no significant difference in global H3K27me3 levels between Asxl1 tm/+ bone marrow cells and wild-type bone marrow cells (Additional file 1: Figure S6). Then, we analyzed if there was different global H3K27me3 pattern between Asxl1 tm/+ and wild-type Lin- bone marrow cells via ChIP-Seq analysis. Comparing sequencing reads of ChIP products and input controls, we identified ~70 k H3K27me3-binding peaks in each of the Asxl1 tm/+ and wild-type samples. Of note, considerable proportions of them were Asxl1 tm/+ cells-specific (25,695; 37.0%) or wild-type (26,850; 37.7%) cells-specific. These peaks are highly concordant with gene loci in the mouse genome (Fig. 8a). Mn1 harbored three H3K27me3-binding sites, of which one was Asxl1 tm/+ -specific (Fig. 8a, left lower panel). We then analyzed the distribution of the condition-specific peaks in gene regions. Significant enrichment of peaks was found in upstream (within 5 k bps; 7.2 and 6.8% of Asxl1 tm/+ - and wild-type-specific peaks, respectively; both p < 0.001) and downstream regions of gene bodies (within 1 k bps; 1.4%, p = 0.031; and 1.3%, p = 0.023, respectively) compared to randomly distributed peaks across the genome (Fig. 8b). Other genomic categories, such as 5’ and 3’ untranslated regions (UTRs) and exons, were not enriched (all p values >0.05), suggesting the preference of Asxl1 mutation-associated H3K27me3 occupancy in gene regulatory regions.
To further investigate Asxl1 mutation-modulated targets of H3K27me3, we analyzed enriched motifs on the peaks by the MEME-ChIP web tool, which performs motif discovery, enrichment, and visualization from DNA sequences of interest. Interestingly, while Asxl1 tm/+ and wild-type specific peaks do not overlap with each other, they carried very similar motifs: RGRAA, TVTGTR, and TTTAWW (all E values <0.001; Fig. 8c), indicating that the modulation of Asxl1 mutation in H3K27me3 occupancy is independent of the binding motifs.
In order to investigate the effects of such selective targeting on gene expression and biological functions, we linked the peaks with gene expression microarrays. Lists of H3K27me peaks-associated genes with concordant significant downregulation are provided in Additional file 1: Table S1. Notably, the down-regulated genes in Asxl1 wild-type cells were significantly associated with concordant Asxl1 wild-type- specific H3K27m3 peaks (15.69% of the Asxl1 wild-type-specific downregulated genes harbored concordant Asxl1 wild-type-specific H3K27m3 peaks, compared to 11.69% of non-downregulated genes; Fisher’s exact test p = 0.001; Fig. 8d, upper panel). However, Asxl1 tm/+ cells-specific peaks were not significantly associated with down-regulated genes in Asxl1 tm/+ cells (Fisher’s exact test p = 0.52; Fig. 8d, lower panel), implying that in Asxl1 mutated cells, the association between H3K27m3 and gene downregulation is disrupted when compared with Asxl1 wild-type condition. Taken together, our ChIP-Seq data demonstrated distinct Asxl1 mutation-modulated binding profiles of H3K27me3.

Discussion

For the first time, we have demonstrated the pathophysiological functions of a “physiological” dose of Asxl1 mutations in vivo and in vitro. In contrast to the previous studies with enforced overexpression of mutant ASXL1 protein in a background of two wild-type alleles of endogenous Asxl1 [18, 19], our model facilitated investigation of a more clinically relevant Asxl1 mutation.
In our study, we noted while Asxl1 mutation promoted engraftment of MN1-overexpressing cells and showed increased colony formation and cobblestone area formation, the LSK cells bearing Asxl1 mutation had inferior repopulation capacities when compared with wild-type cells in vivo. This counterintuitive observation could be explained by two possibilities: (1) in our in vivo transplantation assays (Figs. 3 and 4), we assessed the activities of HSCs. But MN1 overexpression targeted committed progenitor cells, not HSCs [35]. This may explain the discrepancies between these experimental results; (2) in serial transplantation, the marrow stem cells were taken and expanded in a previously irradiated microenvironment, not normal hematopoietic niche. Spangrude et al. have shown a vastly inferior repopulation capacity of LSK cells repeatedly exposed to such perturbed microenvironment [36]. Such radiation perturbation on microenvironment was absent in colony formation and cobblestone area formation assays, and less severe in MN1 overexpressing cell transplantation assays. We could not rule out the possibility that Asxl1 mutant cells were particularly susceptible to this factor, thus showing decreased repopulation capacity in serial transplantation assays, while similar phenomenon was not shown in the other assays without serial irradiation. Moreover, Kamminga et al. showed that although a gradual decrease of the percentage of LSK cells was observed when LSK cells were used as donor cells in serial transplantation, only minor decrease was observed for the clonogenic CAFC activity of the purified cells [37]. These results suggested that in vivo repopulation ability of LSK cells might be affected by residue host cells or competitor cells. They also highlighted the limitation of current animal assays to detect the “real” in vivo hematopoietic stem cell activities.
In our model, Asxl1 G643WfsX12 mutation did not lead to leukemia or other blood malignancies in a 18-24-month observation period, indicating that a physiological dose of Asxl1 G643WfsX12 was not sufficient for leukemogenesis. Nevertheless, the mutation could enhance engraftment of MN1 overexpressing cells, suggesting that Asxl1 mutation could function as a cooperative hit of MN1 overexpression to promote the engraftment of bone marrow cells. This is consistent with the clinical observation that ASXL1 mutant burden often increases in disease progression and mutations in ASXL1 [3841], as well as other genes encoding epigenetic modifiers, were often acquired early in the disease and were almost never found in isolation [42].
Gene expression microarray and GSEA showed limited difference between Asxl1 tm/+ and wild-type control bone marrow cells under steady state, consistent with our observation that Asxl1 tm/+ mice did not develop obvious blood diseases. In MN1 overexpression background, the expression patterns and physiological pathways between Asxl1 mutation and wild-type became distinctive (Fig. 7), implying the promoting effects of Asxl1 mutation on MN1 overexpression-induced engraftment of bone marrow cells. The high number of differentially expressed genes and perturbed biological pathways in human ASXL1-mutated versus wild-type AML cells demonstrated a far more complicated milieu in human AML cells compared with mice HSPCs with Asxl1 mutation per se (Fig. 7a, b).
From our microarray studies, we found that Asxl1 mutation alone in mice had little effects on both gene expression profiles and biological pathways, while the perturbation became obvious in the presence of MN1 overexpression. How MN1 overexpression augments the genomic effects of Asxl1 mutation is not completely defined in our study, but we found that Asxl1 mutation plus MN1 overexpression, but not Asxl1 mutation alone, was associated with enrichment of signatures representing multipotent progenitors and hematopoietic stem cells, as well as genes related to oncogenic MEK.
Hypoxia-related genes are considered critical for the survival of leukemia initiation cells [34, 43]. The enrichment in hematopoietic stem cell and multipotent progenitor gene sets further implies the supporting function of ASXL1 mutation in blood malignancies. KRAS is considered relevant in leukemia formation [4446]; the enrichment in KRAS gene set confers the possibility that mutant ASXL1 act as a cofactor in disease development. MAPK/ERK pathway is crucial for hematopoiesis and aberrant MAPK/ERK pathway is associated with cancer formation [47]. RAS signaling are also considered to be involved in AML transformation at both genetic and epigenetic levels [48]. Bone marrow cells of our mouse model were supposed to have Asxl1 mutation alone, but in ASXL1-mutated human AML cells, we expected there were additional genetic perturbations. One of the advantages of our mouse model was that it enabled us to interrogate the functions of Asxl1 mutation per se, in a “simpler” genetic background. This was probably why we saw different biological effects of MN1 overexpression between BM cells in our mouse model and human AML cells with more complex genetic background.
Since Asxl1 has been considered to be associated with the regulation of H3K27me3, we performed ChIP-Seq to investigate the alteration of global H3K27me3 pattern in Asxl1 tm/+ bone marrow cells. Considerable numbers of H3K27me3 peaks specific to Asxl1 tm/+ and to wild-type bone marrow cells were noted and preponderantly located within 5 k upstream and 1 k downstream of gene bodies. These results indicate that Asxl1 mutation can modulate the global pattern of histone methylation in a non-random manner, preferentially immediate to the gene bodies. In addition, in Asxl1 mutated cells, the correlation between H3K27m3 and gene down-regulation appears attenuated when compared with Asxl1 wild-type context, suggesting functional implications of Asxl1 functions in H3K27me3 modulation. Taken together, our systematic analyses unveiled crucial oncogenic functions perturbed by the interplay between Asxl1 mutation and MN1 overexpression that may partially account for the cooperative role of Asxl1 mutations in MN1-associated leukemia in human and mouse settings and the functional impacts of ASXL1 mutation in human AML.

Conclusions

Taken together, for the first time, our study reveals the in vitro and in vivo effects of a “physiological” dose of Asxl1 mutation. Although mutant Asxl1 does not act as a sufficient driver in blood malignancies, it facilitates engraftment of cells overexpressing MN1. Our study also enlightens the effects on global H3K27m3 profiles by Asxl1 mutation and several potential biological pathways underlying mutant ASXL1.

Acknowledgements

We thank the technical services provided by the Transgenic Mouse Model Core Facility of the National Core Facility Program for Biotechnology, Ministry of Science and Technology, Taiwan, the Gene Knockout Mouse Core Laboratory of National Taiwan University Center of Genomic Medicine, and the National Center for Genome Medicine. MN1 expression construct was a kind gift from Dr. R. Keith Humphries. We would like to acknowledge the service provided by the Flow Cytometric Analyzing and Sorting Core Facilities at National Taiwan University Hospital and First Core Laboratory of National Taiwan University College of Medicine. We thank the Taiwan Mouse Clinic (MOST 105-2325-B-001-010) which is funded by the National Research Program for Biopharmaceuticals (NRPB) at the Ministry of Science and Technology of Taiwan for technical support in complete blood count and tissue section experiment. We thank the Drs. Hsing-Chen Tsai, Dr. Tai-Chung Huang, and Yen-Wei Chen for the technical support in ChIP-Seq sample preparation and analysis. We also thank the National Center for Genome Medicine for the technical and bioinformatics service in ChIP-Seq analysis. We appreciated the staff of the Eighth Core Lab, Department of Medical Research, National Taiwan University Hospital for technical support during the study.

Funding

The study was supported by a National Taiwan University Hospital − National Taiwan University joint research grant (UN103-051), Ministry of Science and Technology of Taiwan (MOST102-2325-B-002-028, 103-2314-B-002-130-MY3, 103-2314-B-002-131MY3 and 104-2923-B-002-001), Far Eastern Hospital and NTUH joint grant 105-FTN24, and Ministry of Health and Welfare of Taiwan (MOHW106-TDU-B-211-144005).

Availability of data and materials

All data generated or analyzed during this study are available from the corresponding author upon reasonable request.

Authors’ contributions

YCH wrote the paper, performed the experiments, and analyzed data. YCC wrote the paper and analyzed the data. WCC and HFT planned, designed and coordinated the research, and wrote the manuscript. CCL, YYK, HAH, YST, CJK, PHC, MHT, and THH provided important materials and help in the study. All authors read and approved the final manuscript.
The collection of patients’ leukemia cells for microarray studies was approved by the Institutional Review Board of the National Taiwan University Hospital. Animals used in this study were housed in a specific pathogen-free animal facility and all procedures were approved by IACUC of National Taiwan University College of Medicine (IACUC approval number: 20120346).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Fisher CL, Lee I, Bloyer S, Bozza S, Chevalier J, Dahl A, Bodner C, Helgason CD, Hess JL, Humphries RK, et al. Additional sex combs-like 1 belongs to the enhancer of trithorax and polycomb group and genetically interacts with Cbx2 in mice. Dev Biol. 2010;337(1):9–15.CrossRefPubMed Fisher CL, Lee I, Bloyer S, Bozza S, Chevalier J, Dahl A, Bodner C, Helgason CD, Hess JL, Humphries RK, et al. Additional sex combs-like 1 belongs to the enhancer of trithorax and polycomb group and genetically interacts with Cbx2 in mice. Dev Biol. 2010;337(1):9–15.CrossRefPubMed
2.
Zurück zum Zitat Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, Heguy A, Bueso-Ramos C, Kantarjian H, Levine RL, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70(2):447–52.CrossRefPubMedPubMedCentral Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, Heguy A, Bueso-Ramos C, Kantarjian H, Levine RL, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70(2):447–52.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Boultwood J, Perry J, Pellagatti A, Fernandez-Mercado M, Fernandez-Santamaria C, Calasanz MJ, Larrayoz MJ, Garcia-Delgado M, Giagounidis A, Malcovati L, et al. Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia. Leukemia. 2010;24(5):1062–5.CrossRefPubMed Boultwood J, Perry J, Pellagatti A, Fernandez-Mercado M, Fernandez-Santamaria C, Calasanz MJ, Larrayoz MJ, Garcia-Delgado M, Giagounidis A, Malcovati L, et al. Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia. Leukemia. 2010;24(5):1062–5.CrossRefPubMed
4.
Zurück zum Zitat Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adelaide J, Rey J, Vainchenker W, Bernard OA, Chaffanet M, Vey N, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183–6.CrossRefPubMed Carbuccia N, Murati A, Trouplin V, Brecqueville M, Adelaide J, Rey J, Vainchenker W, Bernard OA, Chaffanet M, Vey N, et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia. 2009;23(11):2183–6.CrossRefPubMed
5.
Zurück zum Zitat Hoischen A, van Bon BW, Rodriguez-Santiago B, Gilissen C, Vissers LE, de Vries P, Janssen I, van Lier B, Hastings R, Smithson SF, et al. De novo nonsense mutations in ASXL1 cause Bohring-Opitz syndrome. Nat Genet. 2011;43(8):729–31.CrossRefPubMed Hoischen A, van Bon BW, Rodriguez-Santiago B, Gilissen C, Vissers LE, de Vries P, Janssen I, van Lier B, Hastings R, Smithson SF, et al. De novo nonsense mutations in ASXL1 cause Bohring-Opitz syndrome. Nat Genet. 2011;43(8):729–31.CrossRefPubMed
6.
7.
Zurück zum Zitat Scheuermann JC, de Ayala Alonso AG, Oktaba K, Ly-Hartig N, McGinty RK, Fraterman S, Wilm M, Muir TW, Muller J. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature. 2010;465(7295):243–7.CrossRefPubMedPubMedCentral Scheuermann JC, de Ayala Alonso AG, Oktaba K, Ly-Hartig N, McGinty RK, Fraterman S, Wilm M, Muir TW, Muller J. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature. 2010;465(7295):243–7.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Balasubramani A, Larjo A, Bassein JA, Chang X, Hastie RB, Togher SM, Lahdesmaki H, Rao A. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex. Nat Commun. 2015;6:7307.CrossRefPubMedPubMedCentral Balasubramani A, Larjo A, Bassein JA, Chang X, Hastie RB, Togher SM, Lahdesmaki H, Rao A. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex. Nat Commun. 2015;6:7307.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, Kirkpatrick DS, Pham VC, Lill JR, Bakalarski CE, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012;337:6.CrossRef Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, Kirkpatrick DS, Pham VC, Lill JR, Bakalarski CE, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012;337:6.CrossRef
10.
Zurück zum Zitat Chou W-C, Huang H-H, Hou H-A, Chen C-Y, Tang J-L, Yao M, Tsay W, Ko B-S, Wu S-J, Huang S-Y, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94.CrossRefPubMed Chou W-C, Huang H-H, Hou H-A, Chen C-Y, Tang J-L, Yao M, Tsay W, Ko B-S, Wu S-J, Huang S-Y, et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood. 2010;116(20):4086–94.CrossRefPubMed
11.
Zurück zum Zitat Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, Wagner K, Chaturvedi A, Sharma A, Wichmann M, et al. Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol. 2011;29(18):2499–506.CrossRefPubMed Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, Wagner K, Chaturvedi A, Sharma A, Wichmann M, et al. Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol. 2011;29(18):2499–506.CrossRefPubMed
12.
Zurück zum Zitat Schnittger S, Eder C, Jeromin S, Alpermann T, Fasan A, Grossmann V, Kohlmann A, Illig T, Klopp N, Wichmann HE, 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(1):82–91.CrossRefPubMed Schnittger S, Eder C, Jeromin S, Alpermann T, Fasan A, Grossmann V, Kohlmann A, Illig T, Klopp N, Wichmann HE, 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(1):82–91.CrossRefPubMed
13.
Zurück zum Zitat Pratcorona M, Abbas S, Sanders MA, Koenders JE, Kavelaars FG, Erpelinck-Verschueren CAJ, Zeilemakers A, Löwenberg B, Valk PJM. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388–92.CrossRefPubMedPubMedCentral Pratcorona M, Abbas S, Sanders MA, Koenders JE, Kavelaars FG, Erpelinck-Verschueren CAJ, Zeilemakers A, Löwenberg B, Valk PJM. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388–92.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, Pandey J, Patel JP, Chung YR, Koche R, et al. ASXL1 mutation promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012;22:14.CrossRef Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, Pandey J, Patel JP, Chung YR, Koche R, et al. ASXL1 mutation promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012;22:14.CrossRef
15.
Zurück zum Zitat Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N, Carbuccia N, Lagarde A, Prebet T, Nezri M, Sainty D, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800.CrossRefPubMed Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N, Carbuccia N, Lagarde A, Prebet T, Nezri M, Sainty D, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800.CrossRefPubMed
16.
Zurück zum Zitat Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bodner C, Hess JL, Humphries RK, Brock HW. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia. Blood. 2010;115(1):38–46.CrossRefPubMedPubMedCentral Fisher CL, Pineault N, Brookes C, Helgason CD, Ohta H, Bodner C, Hess JL, Humphries RK, Brock HW. Loss-of-function Additional sex combs like 1 mutations disrupt hematopoiesis but do not cause severe myelodysplasia or leukemia. Blood. 2010;115(1):38–46.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Wang J, Li Z, He Y, Pan F, Chen S, Rhodes S, Nguyen L, Yuan J, Jiang L, Yang X, et al. Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice. Blood. 2014;123(4):541–53.CrossRefPubMedPubMedCentral Wang J, Li Z, He Y, Pan F, Chen S, Rhodes S, Nguyen L, Yuan J, Jiang L, Yang X, et al. Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice. Blood. 2014;123(4):541–53.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, Kuscu C, Hricik T, Ndiaye-Lobry D, Lafave LM, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210(12):2641–59.CrossRefPubMedPubMedCentral Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, Kuscu C, Hricik T, Ndiaye-Lobry D, Lafave LM, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210(12):2641–59.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Inoue D, Kitaura J, Togami K, Nishimura K, Enomoto Y, Uchida T, Kagiyama Y, Kawabata KC, Nakahara F, Izawa I, et al. Myelodysplastic syndromes are induced by histone methylation-altering ASXL1 mutations. J Clin Invest. 2013;123(11):14.CrossRef Inoue D, Kitaura J, Togami K, Nishimura K, Enomoto Y, Uchida T, Kagiyama Y, Kawabata KC, Nakahara F, Izawa I, et al. Myelodysplastic syndromes are induced by histone methylation-altering ASXL1 mutations. J Clin Invest. 2013;123(11):14.CrossRef
20.
Zurück zum Zitat Inoue D, Matsumoto M, Nagase R, Saika M, Fujino T, Nakayama KI, Kitamura T. Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels. Exp Hematol. 2016;44(3):172–176.e171.CrossRefPubMed Inoue D, Matsumoto M, Nagase R, Saika M, Fujino T, Nakayama KI, Kitamura T. Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels. Exp Hematol. 2016;44(3):172–176.e171.CrossRefPubMed
22.
23.
Zurück zum Zitat da Huang W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13.CrossRef da Huang W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13.CrossRef
24.
Zurück zum Zitat da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.CrossRef da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.CrossRef
26.
Zurück zum Zitat Bailey TL, Boden M, Buske FA, Frith M, Grant CE, Clementi L, Ren J, Li WW, Noble WS. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 2009;37(Web Server issue):W202–208.CrossRefPubMedPubMedCentral Bailey TL, Boden M, Buske FA, Frith M, Grant CE, Clementi L, Ren J, Li WW, Noble WS. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 2009;37(Web Server issue):W202–208.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6.CrossRefPubMedPubMedCentral Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178–92.CrossRefPubMed Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013;14(2):178–92.CrossRefPubMed
29.
Zurück zum Zitat Chuang MK, Chiu YC, Chou WC, Hou HA, Chuang EY, Tien HF. A 3-microRNA scoring system for prognostication in de novo acute myeloid leukemia patients. Leukemia. 2015;29(5):1051–9.CrossRefPubMed Chuang MK, Chiu YC, Chou WC, Hou HA, Chuang EY, Tien HF. A 3-microRNA scoring system for prognostication in de novo acute myeloid leukemia patients. Leukemia. 2015;29(5):1051–9.CrossRefPubMed
30.
Zurück zum Zitat Chuang MK, Chiu YC, Chou WC, Hou HA, Tseng MH, Kuo YY, Chen Y, Chuang EY, Tien HF. An mRNA expression signature for prognostication in de novo acute myeloid leukemia patients with normal karyotype. Oncotarget. 2015;6(36):39098–110.PubMedPubMedCentral Chuang MK, Chiu YC, Chou WC, Hou HA, Tseng MH, Kuo YY, Chen Y, Chuang EY, Tien HF. An mRNA expression signature for prognostication in de novo acute myeloid leukemia patients with normal karyotype. Oncotarget. 2015;6(36):39098–110.PubMedPubMedCentral
31.
Zurück zum Zitat Chiu YC, Tsai MH, Chou WC, Liu YC, Kuo YY, Hou HA, Lu TP, Lai LC, Chen Y, Tien HF, et al. Prognostic significance of NPM1 mutation-modulated microRNA-mRNA regulation in acute myeloid leukemia. Leukemia. 2016;30(2):274–84.PubMed Chiu YC, Tsai MH, Chou WC, Liu YC, Kuo YY, Hou HA, Lu TP, Lai LC, Chen Y, Tien HF, et al. Prognostic significance of NPM1 mutation-modulated microRNA-mRNA regulation in acute myeloid leukemia. Leukemia. 2016;30(2):274–84.PubMed
32.
Zurück zum Zitat Heuser M, Argiropoulos B, Kuchenbauer F, Yung E, Piper J, Fung S, Schlenk RF, Dohner K, Hinrichsen T, Rudolph C, et al. MN1 overexpression induces acute myeloid leukemia in mice and predicts ATRA resistance in patients with AML. Blood. 2007;110(5):1639–47.CrossRefPubMed Heuser M, Argiropoulos B, Kuchenbauer F, Yung E, Piper J, Fung S, Schlenk RF, Dohner K, Hinrichsen T, Rudolph C, et al. MN1 overexpression induces acute myeloid leukemia in mice and predicts ATRA resistance in patients with AML. Blood. 2007;110(5):1639–47.CrossRefPubMed
33.
Zurück zum Zitat Losman JA, Looper RE, Koivunen P, Lee S, Schneider RK, McMahon C, Cowley GS, Root DE, Ebert BL, Kaelin Jr WG. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science (New York, NY). 2013;339(6127):1621–5.CrossRef Losman JA, Looper RE, Koivunen P, Lee S, Schneider RK, McMahon C, Cowley GS, Root DE, Ebert BL, Kaelin Jr WG. (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science (New York, NY). 2013;339(6127):1621–5.CrossRef
35.
Zurück zum Zitat Heuser M, Yun H, Berg T, Yung E, Argiropoulos B, Kuchenbauer F, Park G, Hamwi I, Palmqvist L, Lai Courteney K, et al. Cell of origin in AML: susceptibility to MN1-induced transformation is regulated by the MEIS1/AbdB-like HOX protein complex. Cancer Cell. 2011;20(1):39–52.CrossRefPubMedPubMedCentral Heuser M, Yun H, Berg T, Yung E, Argiropoulos B, Kuchenbauer F, Park G, Hamwi I, Palmqvist L, Lai Courteney K, et al. Cell of origin in AML: susceptibility to MN1-induced transformation is regulated by the MEIS1/AbdB-like HOX protein complex. Cancer Cell. 2011;20(1):39–52.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Spangrude G, Brooks D, Tumas D. Long-term repopulation of irradiated mice with limiting numbers of purified hematopoietic stem cells: in vivo expansion of stem cell phenotype but not function. Blood. 1995;85(4):1006–16.PubMed Spangrude G, Brooks D, Tumas D. Long-term repopulation of irradiated mice with limiting numbers of purified hematopoietic stem cells: in vivo expansion of stem cell phenotype but not function. Blood. 1995;85(4):1006–16.PubMed
37.
Zurück zum Zitat Kamminga LM, van Os R, Ausema A, Noach EJK, Weersing E, Dontje B, Vellenga E, de Haan G. Impaired hematopoietic stem cell functioning after serial transplantation and during normal aging. Stem Cells. 2005;23(1):82–92.CrossRefPubMed Kamminga LM, van Os R, Ausema A, Noach EJK, Weersing E, Dontje B, Vellenga E, de Haan G. Impaired hematopoietic stem cell functioning after serial transplantation and during normal aging. Stem Cells. 2005;23(1):82–92.CrossRefPubMed
38.
Zurück zum Zitat Beekman R, Valkhof MG, Sanders MA, van Strien PMH, Haanstra JR, Broeders L, Geertsma-Kleinekoort WM, Veerman AJP, Valk PJM, Verhaak RG, et al. Sequential gain of mutations in severe congenital neutropenia progressing to acute myeloid leukemia. Blood. 2012;119(22):5071–7.CrossRefPubMed Beekman R, Valkhof MG, Sanders MA, van Strien PMH, Haanstra JR, Broeders L, Geertsma-Kleinekoort WM, Veerman AJP, Valk PJM, Verhaak RG, et al. Sequential gain of mutations in severe congenital neutropenia progressing to acute myeloid leukemia. Blood. 2012;119(22):5071–7.CrossRefPubMed
39.
40.
Zurück zum Zitat Engle EK, Fisher DAC, Miller CA, McLellan MD, Fulton RS, Moore DM, Wilson RK, Ley TJ, Oh ST. Clonal evolution revealed by whole genome sequencing in a case of primary myelofibrosis transformed to secondary acute myeloid leukemia. Leukemia. 2015;29(4):869–76.CrossRefPubMed Engle EK, Fisher DAC, Miller CA, McLellan MD, Fulton RS, Moore DM, Wilson RK, Ley TJ, Oh ST. Clonal evolution revealed by whole genome sequencing in a case of primary myelofibrosis transformed to secondary acute myeloid leukemia. Leukemia. 2015;29(4):869–76.CrossRefPubMed
41.
Zurück zum Zitat Makishima H, Jankowska AM, McDevitt MA, O'Keefe C, Dujardin S, Cazzolli H, Przychodzen B, Prince C, Nicoll J, Siddaiah H, et al. CBL, CBLB, TET2, ASXL1, and IDH1/2 mutations and additional chromosomal aberrations constitute molecular events in chronic myelogenous leukemia. Blood. 2011;117(21):e198–206.CrossRefPubMedPubMedCentral Makishima H, Jankowska AM, McDevitt MA, O'Keefe C, Dujardin S, Cazzolli H, Przychodzen B, Prince C, Nicoll J, Siddaiah H, et al. CBL, CBLB, TET2, ASXL1, and IDH1/2 mutations and additional chromosomal aberrations constitute molecular events in chronic myelogenous leukemia. Blood. 2011;117(21):e198–206.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21.CrossRefPubMedPubMedCentral Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Testa U, Labbaye C, Castelli G, Pelosi E. Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol. 2016;44(7):540–60.CrossRefPubMed Testa U, Labbaye C, Castelli G, Pelosi E. Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol. 2016;44(7):540–60.CrossRefPubMed
44.
Zurück zum Zitat Braun BS, Shannon K. Targeting Ras in myeloid leukemias. Clin Cancer Res. 2008;14(8):2249–52.CrossRefPubMed Braun BS, Shannon K. Targeting Ras in myeloid leukemias. Clin Cancer Res. 2008;14(8):2249–52.CrossRefPubMed
45.
Zurück zum Zitat Grossmann V, Schnittger S, Poetzinger F, Kohlmann A, Stiel A, Eder C, Fasan A, Kern W, Haferlach T, Haferlach C. High incidence of RAS signalling pathway mutations in MLL-rearranged acute myeloid leukemia. Leukemia. 2013;27(9):1933–6.CrossRefPubMed Grossmann V, Schnittger S, Poetzinger F, Kohlmann A, Stiel A, Eder C, Fasan A, Kern W, Haferlach T, Haferlach C. High incidence of RAS signalling pathway mutations in MLL-rearranged acute myeloid leukemia. Leukemia. 2013;27(9):1933–6.CrossRefPubMed
46.
Zurück zum Zitat Kong G, Chang YI, Damnernsawad A, You X, Du J, Ranheim EA, Lee W, Ryu MJ, Zhou Y, Xing Y, et al. Loss of wild-type Kras promotes activation of all Ras isoforms in oncogenic Kras-induced leukemogenesis. Leukemia. 2016;30(7):1542–51.CrossRefPubMedPubMedCentral Kong G, Chang YI, Damnernsawad A, You X, Du J, Ranheim EA, Lee W, Ryu MJ, Zhou Y, Xing Y, et al. Loss of wild-type Kras promotes activation of all Ras isoforms in oncogenic Kras-induced leukemogenesis. Leukemia. 2016;30(7):1542–51.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Geest CR, Coffer PJ. MAPK signaling pathways in the regulation of hematopoiesis. J Leukoc Biol. 2009;86(2):237–50.CrossRefPubMed Geest CR, Coffer PJ. MAPK signaling pathways in the regulation of hematopoiesis. J Leukoc Biol. 2009;86(2):237–50.CrossRefPubMed
48.
Zurück zum Zitat Caye A, Strullu M, Guidez F, Cassinat B, Gazal S, Fenneteau O, Lainey E, Nouri K, Nakhaei-Rad S, Dvorsky R, et al. Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network. Nat Genet. 2015;47(11):1334–40.CrossRefPubMed Caye A, Strullu M, Guidez F, Cassinat B, Gazal S, Fenneteau O, Lainey E, Nouri K, Nakhaei-Rad S, Dvorsky R, et al. Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network. Nat Genet. 2015;47(11):1334–40.CrossRefPubMed
Metadaten
Titel
The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model
verfasst von
Yueh-Chwen Hsu
Yu-Chiao Chiu
Chien-Chin Lin
Yuan-Yeh Kuo
Hsin-An Hou
Yi-Shiuan Tzeng
Chein-Jun Kao
Po-Han Chuang
Mei-Hsuan Tseng
Tzu-Hung Hsiao
Wen-Chien Chou
Hwei-Fang Tien
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2017
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-017-0508-x

Weitere Artikel der Ausgabe 1/2017

Journal of Hematology & Oncology 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.