Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Nov 2.
Published in final edited form as: Leukemia. 2016 Nov 24;31(6):1382–1390. doi: 10.1038/leu.2016.347

Loss of p300 Accelerates MDS-associated Leukemogenesis

Guoyan Cheng 1, Fan Liu 1, Takashi Asai 1, Fan Lai 2, Na Man 1, Haiming Xu 4, Shi Chen 1, Sarah Greenblatt 1, Pierre-Jacques Hamard 1, Koji Ando 1, Conception Martinez 1, Madhavi Tadi 1, Lan Wang 1, Mingjiang Xu 1, Feng-Chun Yang 1, Ramin Shiekhattar 2, Stephen D Nimer 1,3,*
PMCID: PMC5667352  NIHMSID: NIHMS837948  PMID: 27881875

Abstract

The role that changes in DNA methylation and histone modifications play in human malignancies is poorly understood. p300 and CBP, two distinct but highly homologous lysine acetyltransferases (KATs), are mutated in several cancers, suggesting their role as tumor suppressors. In the current study, we found that deletion of p300, but not CBP, markedly accelerated the leukemogenesis of Nup98-HoxD13 (NHD13) transgenic mice, an animal model that phenotypically copies human myelodysplastic syndrome (MDS). p300 deletion restored the ability of NHD13 expressing hematopoietic stem and progenitor cells (HSPCs) to self-renew in vitro, and to expand in vivo, with an increase in stem cell symmetric self-renewal divisions and a decrease in apoptosis. Furthermore, loss of p300, but not CBP, promoted cytokine signaling, including enhanced activation of the MAPK and JAK/STAT pathways in the HSPC compartment. Altogether, our data indicate that p300 plays a pivotal role in blocking the transformation of MDS to acute myeloid leukemia (AML), a role distinct from that of CBP.

Introduction

Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoiesis, characterized by inefficient hematopoiesis with bone marrow dysplasia, peripheral blood cytopenias, and a predisposition to progress to acute myeloid leukemia (AML)1. Considering the therapeutic response of MDS to DNA hypomethylating agents, and the recurrent somatic mutations found in epigenetic modifiers, including ASXL1, TET2 and DNMT3A2, 3, many cases of MDS appear to represent an epigenetic disease.

The histone lysine acetyltransferases (KATs), p300 and its homologue CBP (CREB binding protein), function primarily as transcriptional co-factors for a number of nuclear proteins, including MYC, p53, and E2F4. Both p300 and CBP have been implicated in tumorigenesis. For example, germline mutations that disrupt p300 and CBP are responsible for some cases of Rubinstein–Taybi syndrome (RTS), a genetic disorder with increased predisposition to develop tumors of neural and hematopoietic origins5. Somatic, loss-of-function mutations in p300 and CBP are found in lymphomas and some solid tumors, including colorectal cancer and gastric cancer6. Although rare, chromosomal translocations, t(8;16) and t(8;22), that fuse amino terminus of CREBBP (the gene encoding CBP) or EP300 (the gene encoding p300) to MOZ (the gene encoding monocytic leukemia zinc-finger protein) are found in the M4/M5 subtype of AML7, 8. These translocations generate haploinsufficiency for CBP and p300, in addition to the gain-of-functions intrinsic to the fusion proteins they generate.

The function of p300/CBP in normal and malignant hematopoiesis has been studied in genetic mouse models. Both p300−/−and CBP−/−mice exhibit embryonic lethality due to similar defects in growth and neural tube closure9. p300 or CBP deficiency impairs the self-renewal and the differentiation of hematopoietic stem cells in mice10, 11. In addition, heterozygous CBP knockout mice, but not heterozygous p300 knockout mice, develop hematopoietic malignancies12, indicating the non-redundant function of these two KATs in leukemogenesis.

While the above evidence suggests a tumor suppressor role of p300/CBP, we previously reported that p300 acetylates the AML1-ETO leukemogenic fusion protein and is required for AML1-ETO-driven AML development13, suggesting an oncogene-specific and cell context-dependent function of p300 in tumorigenesis. Genetic studies on MDS and MDS-related AML patients revealed various mutations, deletions and translocations in Ep300 gene1417. The mutation rate of Ep300 ranges from 2 to 7% in different cohorts of patients in these studies. Here, we examined the role of p300 and CBP in the pathogenesis of MDS-derived AML; using Mx1Cre induced deletion of Ep300 or CREBBP in Nup98-HoxD13 (NHD13) transgenic mice. We found that loss of p300, but not CBP, significantly accelerated the onset of AML and triggered hematopoietic stem and progenitor cell (HSPC) expansion. The early onset of transformation in p300 deleted NHD13 mice is associated with an enhanced “cytokine signaling profile”, affecting both MAPK and JAK/STAT pathways. Overall, our results indicate that p300, but not CBP, functions as a powerful tumor suppressor in the progression of NHD13-driven MDS to AML.

Materials and Methods

Mice

CD45.1 B6SJL mice and Mx1Cre mice were purchased from Jackson laboratory. NHD13 transgenic mice (kindly provided by Perter Aplan, National Institute of Health), p300flox/flox mice and CBPflox/flox mice (kindly provided by Paul K. Brindle, St. Jude Children's Research Hospital) were previously described18. All mice used in the experiments are in pure C57BL/6 genetic background. Mice were maintained in the University of Miami animal facility under virus-antibody-free (VAF) conditions. Animal studies were approved by the Institutional Animal Care and Use Committee at the University of Miami.

Flow analysis and cell sorting

Single cell suspensions were prepared from bone marrow and spleen cells as previously described19. APC-c-kit, PE-Cy7-Sca-1 and Pacblue-Ki67 antibodies were purchased from eBioscience. APC-Cy7-Streptavidin, FITC-CD45.1 and APC-CD45.2 antibodies were purchased from Biolegend. The bone marrow biotin lineage panel and 7AAD/annexin-V staining kit were purchased from BD Pharmingen. All flow samples were analyzed by Canto-II or Fortessa flow cytometer and sorted by Aria-II cell sorter (Becton Dickinson).

For phospho-Flow, bone marrow cells were stimulated with or without a cytokine cocktail, containing murine IL-3 (PeproTech) 10ng/mL, murine IL-6 (PeproTech) 10ng/mL, murine SCF (PeproTech) 100ng/mL, and murine GM-CSF (BioLegend) 200 ng/mL for 15 minutes. Cells were then fixed in 1.5% paraformaldehyde (Electron Microscopy Sciences) at 37 °C for 10 minutes and permeabilized on ice with Methanol for 15 minutes. Subsequently, cells were stained with antibodies to cell surface markers as well as to pERK1/2 (T202/Y204), pSTAT3 (Y705), pSTAT5 (Y694), pAKT (S473) or IgG isotope control for 30 minutes at room temperature, before being subjected to FACS analysis. All phospho-specific antibodies were purchased from eBiosciences and are PE-conjugated.

Bone marrow transplantation and induced deletion of p300/CBP

8-week old B6SJL recipient mice were lethally irradiated (total body irradiation at 950 rads), and 6 hours later, 2×106 donor bone marrow cells were injected intravenously (i.v.) into each recipient mouse. Four weeks after transplantation, Mx1Cre-directed deletion of p300/CBP was induced by three intraperitoneal (i.p.) injections of poly(I:C) (10ug/G body weight, Invivogen), given every other day.

Paired daughter cell assay

CD34 LSK cells were sorted individually into 96-well plates with 150μl of StemSpan Serum-Free Expansion Medium (Stem Cell Technology) per well, supplemented with 100ng/ml murine SCF and TPO. After overnight culture, if the single cell divided into 2 cells, the medium was vigorously mixed to separate the daughter cells. IMDM containing 10% FBS plus murine SCF (100ng/ml), TPO (100ng/ml), G-CSF (20ng/ml each), IL3 (10ng/ml) and EPO (2U/ml) was added to these wells to make the final volume to 200μl/well. Plates were incubated for another 2 weeks (37°C/5% CO2). Cytospins were prepared from the clones formed by each daughter pair. After Diff-Quick staining, symmetric self-renewal divisions (SS) were determined by the presence of all 4 lineages (granulocyte, erythroid, megakaryocyte, monocyte) from each of the paired daughter cells. Symmetric commitment divisions (SD) were determined by the presence of 3 or less lineages from each of the paired daughter cells. Asymmetric self-renewal divisions (AS) were determined by the presence of 3 or less lineages from one daughter cell but 4 lineages from the other daughter cell.

Serial replating assay

1×104 BM Linc-kit+ HSPCs were plated as duplicates in methylcellulose media (MethoCult™ GF M3434, Stem Cell Technologies). Colonies were scored 7 days after seeding; replating was conducted weekly.

RNA-sequencing and data analyses

Sorted Linc-kit+ HSPCs were lysed in Trizol (Invitrogen) and RNA was isolated and treated with DNase as manufacturer’s instruction. The quality of RNA was evaluated on an Agilent Bioanalyzer, and samples with RNA integrity numbers > 8.0 were prepared for sequencing. Poly(A)-tailed RNA was prepared by using mRNA Sequencing Sample Preparation kit (Illumina) and libraries for the deep sequencing studies were generated by the Genomic Core Facility at the University of Miami. Sequencing and analysis were performed with three individual samples per group. Reads were aligned to the GRCm38/mm10 build of the Mus musculus genome with Subread aligner. We tested differentially expressed genes for enrichment in KEGG gene sets20 in the Molecular Signature Database (MSigDB http://www.broadinstitute.org/gsea/msigdb/index.jsp; version 5.0). Gene set enrichment analysis (GSEA) was conducted using the pre-ranked tool in GSEA software21.

RT-PCR

RNA was extracted from bone marrow cells and spleen cells using the RNeasy Mini or RNeasy Micro kit (Qiagen). cDNA was synthesized using the iScript cDNA synthesis kit (Bio-Rad). Mouse Ep300 primers (Mm01310115_m1), Crebbp primers (Mm01342452_m1), Il3ra primers (Mm00434273_m1), Il4ra primers (Mm01275139_m1), Il6ra primers (Mm00439653_m1), Csf2rb primers (Mm00655745_m1) and Gapdh PCR primers (Mm99999915_g1) were purchased from Invitrogen. qPCR was performed on an ABI 7500 real-time cycler. Data were analyzed by comparative CT method as 2 ΔΔCT.

Statistical Analysis

All data were analyzed by GraphPad Prism 6. For survival analysis, we plotted Kaplan-Meir curves and performed the log-rank (Mantel-Cox) test to assess the significance. The comparison of two groups or multiple groups was performed by unpaired t-test or ANOVA with Tukey’s multiple comparisons test. Significant differences are designated as *p<0.05, **p<0.01, ***p<0.001, and ****p<0.0001.

Results

Loss of p300 greatly accelerates the leukemogenesis of NHD13 BM cells

To evaluate the role of p300 and CBP in the progression of MDS disease, we utilized the well-studied, NHD13 transgenic mouse model of MDS22. Initially identified in therapy-related MDS, the NHD13 fusion protein induces murine MDS that recapitulates many key clinical features of human MDS, including peripheral blood cytopenias, bone marrow dysplasia with increased apoptosis, and a significant risk of transformation to acute leukemia.

We crossed NHD13 transgenic mice with p300- or CBP-floxed mice; these mice were further crossed with Mx1Cre transgenic mice to generate NHD13-positive; p300 or CBP conditional KO mice. To determine p300/CBP function in MDS, we first performed BM transplantation using 2-month old mice with the following genotypes as donors: NHD13;p300flox/flox;Mx1Cre (referred to as NHD13 in figures), NHD13;p300flox/+;Mx1Cre+, NHD13;p300flox/flox;Mx1Cre+, NHD13;CBPflox/flox;Mx1Cre+ and p300flox/flox;Mx1Cre+. Deletion of p300 or CBP was induced in the transplant recipient mice 4 weeks later by three i.p. injections of poly(I:C), and qPCR analysis confirmed the efficient deletion of these genes (Fig S1A). To our surprise, the mice that received NHD13;p300flox/flox;Mx1Cre+ BM cells developed increased WBC counts and decreased platelet counts as early as 4 weeks post poly(I:C) injections (Fig S1B). While all of the NHD13 mice developed MDS, with one-third of them died from MDS and one-third of them died from MDS-derived AML by week 40, all of the NHD13;p300Δ/Δ recipient mice died from AML by week 20, without a typical period of cytopenias (Fig 1A and Table). In contrast, loss of CBP in NHD13-expressing BM cells did not affect disease progression in the recipient mice. Furthermore, p300 deletion in wild-type (WT) BM cells had no obvious effect on the survival of recipient mice (Fig 1A and Table), as all mice were disease-free 40 weeks post poly(I:C) injection.

Figure 1. Lack of p300 accelerates the leukemogenesis of NHD13 BM cells in a transplantation model.

Figure 1

(A) BM cells from p300flox/flox;Mx1Cre+, NHD13;p300flox/flox;Mx1Cre- (NHD13), NHD13;p300flox/+;Mx1Cre+, NHD13;p300flox/flox;Mx1Cre+ and NHD13;CBPflox/flox;Mx1Cre+ mice were injected into 8-week old lethally irradiated B6SJL recipient mice (n=12–18). Poly(I:C) administrations were performed 4 weeks post transplantation. Shown is the Kaplan-Meier survival curve of recipient mice of each genotype post poly(I:C) injections.

(B) Top panels show BM and spleen cellularity of leukemic NHD13;p300Δ/Δ mice, compared to that of age-matched NHD13 mice. Bottom panels show WBC, RBC and platelet counts from peripheral blood of leukemic NHD13;p300Δ/Δ and age-matched NHD13 mice (n=4–5).

(C) HSPCs were isolated from recipient mice 2 weeks post poly(I:C) injections. Shown are the representative FACS profiles of Lin- (upper panel), LK and LSK cells (middle panel) of each genotype indicated in the plots. The absolute number of Lin-, LK and LSK BM cells (from 2 femurs and 2 tibias of each mouse) is plotted in the bottom panel.

Table.

The cause of death by 40 weeks post poly(I:C) injections for each genotype in the transplanted experiment.

Cause of death NHD13 NHD13 p300 Δ/+ NHD13 p300 Δ/Δ NHD13 CBP Δ/Δ p300 Δ/Δ
AML 4 (33.3%) 6 (50.0%) 18 (100%) 4 (33.3%) 0
MDS 4 (33.3%) 4 (33.3%) 0 4 (33.3%) 0
Others 0 0 0 1 (16.6%) 0
# of mice alive 4 (33.3%) 2 (16.6%) 0 3 (25.0%) 12 (100%)

The leukemic NHD13;p300Δ/Δ mice had markedly enlarged spleens, increased white blood cell (WBC) counts, and decreased red blood cell (RBC) and platelet (PLT) counts compared to age-matched NHD13 mice (Fig 1B). Immunostaining and flow cytometry showed an expanded c-kit+ population, while Giemsa staining of BM cytospin showed a significant increase in leukemic blasts (Fig S1C).

p300 loss expands the HSPC population in NHD13 mice

To better understand the acceleration of leukemogenesis observed in p300 deleted NHD13 mice, we analyzed the BM HSPC compartment in all recipient mice two weeks after the poly(I:C) injection, when there was no obvious sign of MDS or leukemia. Loss of p300 rapidly altered the BM compartment of the NHD13 mice with a marked expansion of both Lin c-kit+ Sca-1 (LK) cells and Lin c-kit+ Sca-1+ (LSK) cells at this early time point (Fig 1C). In contrast, deletion of p300 in WT BM cells had only modest effect on the HSPC compartment, identifying the context-dependent regulation of hematopoiesis by p300. Furthermore, deletion of CBP in NHD13 mice did not significantly change the LK/LSK cell profile compared to CBP intact NHD13 mice (Fig S1D).

To exclude any potential influence of the BM ablation that follows lethal irradiation on HSPC behavior and disease progression, we performed similar experiments using primary mice, and the same genotypes as used for the transplantation experiments. Mice were injected with poly(I:C) at the pre-MDS stage (2 months of age) and followed for 40 weeks. We confirmed the survival pattern seen in the BM transplantation model, as loss of p300 accelerated leukemogenesis and impaired survival, while the loss of CBP did not alter disease progression or the survival of the NHD13 mice (Fig S2A). The expansion of both LK and LSK population was similar to that seen in the transplanted models (Fig S2B), including the absence of any change in the HSPC compartment when CBP was deleted in the primary NHD13 mice (Fig S2C). In addition, we have also performed the bone marrow transplantation experiments using leukemia cells isolated from primary NHD13, NHD13;p300Δ/Δ and NHD13;CBPΔ/Δ mice, and found that leukemias from all 3 genotypes are all transplantable to secondary recipient mice (data not shown).

Loss of p300 enhances NHD13 HSPC self-renewal

To understand the basis for the p300 deleted NHD13 HSPC expansion, we performed serial replating assays using HSPCs isolated from transplant recipient mice (Fig 2A) or primary mice (Fig 2B) 2 weeks after poly (I:C) injection. In both cases, the NHD13;p300Δ/Δ HSPCs showed increased self-renewal, which was particularly evident after the third replating. As expected, HSPCs from the NHD13 mice had little ability to form colonies in semi-solid culture media. Loss of p300 only minimally affected colony formation of WT cells, confirming the unique role of p300 in NHD13-expressing HSPCs.

Figure 2. Increased stem cell self-renewal and reduced cell death are intrinsic to p300 deleted NHD13 BM cells.

Figure 2

(A) HSPCs were sorted from the indicated recipient mice 2 weeks post poly(I:C) injection. Serial replating assays were performed and the resulting colonies were counted weekly before being re-plated. Shown is the data from 2 independent experiments.

(B) Serial replating assays were performed using HSPCs isolated from the indicated primary mice 2 weeks post poly(I:C) injection. Data plotted here is from 2 separate experiments.

(C) Two weeks post poly(I:C) injection, CD34- LSK cells were isolated from each indicate primary mice and sorted individually into 96-well plate for the paired daughter cell assays. Data are presented as mean frequency from one representative assay out of 2 independent experiments in total.

(D) Shown are the representative FACS profiles of Annexin-V and 7AAD staining of LK (upper panel) and LSK cells (bottom panel) from the indicated mice.

To further our understanding of the improved self-renewal of the NHD13;p300Δ/Δ HSPCs, we performed paired daughter cell assays in vitro using sorted CD34 LSK cells obtained 2 weeks after the poly(I:C) injection. Cytospins were prepared from the clones formed by each daughter pair, and the frequency of symmetric vs. asymmetric division was quantified (Fig 2C). Symmetric commitment divisions (SD) were dominant in NHD13 CD34 LSK cells (50%) compared to WT (28%), p300Δ/Δ (22%) or NHD13;p300Δ/Δ (14%) CD34 LSK cells, while symmetric self-renewal divisions (SS) were over-represented in the NHD13;p300Δ/Δ HSCs (68%) comparing to the WT (31%), p300Δ/Δ (48%) or NHD13 (28%) cells. Altogether, these data indicate a crucial role for p300 in controlling the balance of symmetric stem cell self-renewing divisions vs. stem cell depleting divisions in NHD13 mice.

Deletion of p300 decreases apoptosis in NHD13 BM cells

To further clarify the effect of p300 deletion on NHD13 expressing BM cells, we determined their proliferation and apoptosis 2 weeks after poly(I:C) injection. Using BM cells isolated from primary mice, we found that loss of p300 reduced the elevated apoptotic rate in NHD13-transduced BM cells, especially in the stem cell enriched LSK cells (Fig 2D). In contrast, p300 deletion had little if any effect on the proliferation of WT or NHD13-positive BM cells (Fig S3). Thus, loss of p300 led to unique alterations in the survival of NHD13-expressing BM cells, which would be expected to contribute to leukemia progression.

RNA-sequencing analysis reveals a unique gene signature in p300-null NHD13 HSPCs

To further understand how loss of p300 promotes leukemogenesis, we also purified HSPCs from WT, NHD13, p300Δ/Δ and NHD13;p300Δ/Δ primary mice two weeks after the poly(I:C) injection and from NHD13;p300Δ/Δ mice at the leukemia stage, and prepared RNA samples for comparative RNA-sequencing analysis. We initially normalized the expression profiles of the top 100 genes with the largest standard deviations across all samples by comparing each to the WT HSPCs and noticed many similarities between the WT, p300Δ/Δ and NHD13 samples (Fig 3A). In contrast, the NHD13;p300Δ/Δ HSPCs exhibited a unique profile at the pre-AML, but also at the leukemic stage, indicating a specific interaction between p300 and NHD13 in HSPCs. Consistent with this notion, we found that p300 deletion affected significantly more genes in the NHD13 HSPCs than in the WT HSPCs (2612 vs. 545), with only 148 genes being commonly affected by p300 deletion in both genotypes (Fig 3B). Using unbiased, gene set enrichment analysis (GSEA), we identified three up-regulated pathways that were directly related to hematopoiesis, including the cytokine/cytokine receptor interaction pathway (Fig 3C). Down-regulated pathways included several pathways thought to be involved in malignant transformation, such as RNA splicing, oxidative phosphorylation, and mismatch repair2325.

Figure 3. Differentially expressed genes in p300-null NHD13 HSPCs.

Figure 3

HSPCs were isolated from the indicated primary mice and sorted 2 weeks after poly(I:C) injection. Lin- and ckit+ cells were also isolated from NHD13;p300Δ/Δ mice at leukemia stage. RNA samples were then prepared for RNA-sequencing analyses.

(A) Normalized expression profiles of the top 100 genes with the largest standard deviations across all samples, including samples from the leukemic NHD13;p300Δ/Δ mice. Data represented are variance-stabilizing transformation (VST) counts from the DESeq2 package. The dendrogram shows samples in all groups clustered as expected.

(B) Venn diagram shows the overlap of differentially expressed genes in p300 deleted WT vs NHD13 mice. Genes were considered to be differentially expressed if they had a P-value of less than 0.05 and absolute log fold change of more than 1.5.

(C) Up- and down-regulated pathways identified using Gene Set Enrichment Analysis (GSEA) after p300 deletion in NHD13 HSPCs.

(D) qPCR validation for several differentially expressed genes. Data were normalized to the expression of GAPDH.

Given the flurry of papers reporting the abnormal expression of cytokine receptors on MDS/AML initiating stem cells2629, we performed qPCR assays for several genes, including Il3ra, Il4ra, Il6ra and Csf2rb to validate the RNA-seq data, and we observed highly correlated data showing the up-regulation of these cytokine receptors (Fig 3D and S4A), which could help drive leukemogenesis in the p300-deleted NHD13 mice. In contrast, loss of CBP had no obvious impact on the expression of these cytokine receptors, except for Il4ra (Fig S4B).

p300 deletion enhances cytokine signaling in NHD13 HSPCs

Aberrant cytokine signaling occurs in a variety of malignancies30, with activation of JAK/STAT signaling commonly underlying hematopoietic cell transformation31. To further explore cytokine signaling, we performed the phospho-Flow analyses, using BM cells isolated from mice 2 weeks post poly(I:C) injection. As shown in Fig 4A, B, and C, loss of p300, but not CBP, enhanced phosphorylation of ERK1/2 and STAT5 in response to cytokine stimulation in both WT and NHD13-transformed LK and LSK cells. This suggests that p300 can function to repress cytokine signaling in HSPCs. Given the importance of STAT3 signaling in HSC self-renewal32, it is intriguing to see that p300 deletion markedly up-regulated the STAT3 signaling pathway specifically in NHD13-transformed, but not WT, LSK cells. In contrast, we failed to see any change in AKT phosphorylation after p300 deletion in HSPCs of all genotypes we tested (Fig S5).

Figure 4. Activated MAPK and JAK/STAT signaling in response to cytokine stimulation in NHD13;p300Δ/Δ HSPCs.

Figure 4

BM cells isolated from mice 2 weeks post poly(I:C) injection from wt, NHD13, p300Δ/Δ, NHD13;p300Δ/Δ, CBPΔ/Δ, and NHD13;CBPΔ/Δ were cultured in DMEM supplemented with 20% FBS. Phosphorylation of (A) ERK1/2, (B) STAT3 and (C) STAT5 in LK and LSK cells were determined by flow cytometry after cells were cytokine staved for 2 hours, and stimulated or un-stimulated with a cytokine cocktail (GM-CSF 200ng/ml, SCF 100ng/ml, IL-3 10ng/ml and IL-6 10ng/ml) for 15 minutes. Left panels show representative FACS profiles. Right panels plot the frequency of pERK1/2, pSTAT3, and pSTAT5 positive cells in LK and LSK population (n=4). 1-Wt; 2-NHD13; 3-p300Δ/Δ; 4-NHD13;p300Δ/Δ; 5-CBPΔ/Δ and 6- NHD13;CBPΔ/Δ.

Discussion

We report here that loss of p300 significantly accelerated the transformation of NHD13 BM cells, identifying p300 as a tumor suppressor in NHD13 driven leukemogenesis, which is quite distinct from its role in promoting AML1-ETO associated leukemogenesis 33. Mechanistically, loss of p300 enhanced self-renewal and reduced apoptosis in NHD13 BM cells; in contrast, loss of p300 did not significantly alter WT hematopoiesis, indicating the cell context-dependent role of p300. An earlier study of NUP98-HOXA9-associated blast crisis chronic myeloid leukemia (CML) and AML reported a shift from asymmetric stem cell divisions to symmetric self-renewing divisions and proposed that this shift could lead to disease progression34. Similarly, the increase in symmetric self-renewal divisions (68% vs. 28%) that follows the deletion of p300 in NHD13-expressing HSCs may underlie the profoundly increased risk for leukemogenesis in these mice.

Loss of p300 triggered a unique gene signature in both NHD13-expressing and NHD13-transformed HSPCs, including the up-regulation of the cytokine/cytokine receptor interaction pathway. While this pathway is crucial for normal HSPC biology, it is co-opted in myeloid leukemia cells with inappropriate activation of cytokine signaling resulting from the overexpression or constitutive activation of receptor tyrosine kinases, such as c-KIT, FLT3, and IL-3Rα35. The NHD13;p300Δ/Δ HSPCs have increased expression of several cytokine receptors, including IL-3Rα, IL-4Rα, IL-6Rα and CSFRβ, which activate the MAPK, PI3K/AKT, and JAK/STAT pathways in response to cytokines36. We found significantly increased MAPK and JAK/STAT activation in NHD13;p300Δ/Δ BM cells. Although PI3K/AKT pathway is also downstream of these receptors, it is not affected by p300 deletion, indicating that other mechanism(s) play a more important role in regulating this signaling pathway. It appears that the elevated cytokine signaling that follows the loss of p300 in NHD13 BM confers a growth advantage to pre-leukemic blasts.

Both p300 and CBP were recently shown to facilitate leukemogenesis in cooperation with MOZ-TIF2 and NUP98-HOXA9 37. In the NHD13 MDS model, p300 acts to prevent the transformation to AML, while loss of CBP has no effect on disease progression. Even though these two proteins are highly homologous, p300 and CBP appear to have non-redundant functions in NHD13-mediated leukemogenesis, and it will likely take a combined proteomics and expression profiling approach to better understand how p300 differs from CBP in modifying the epigenetic landscape and controlling this transition.

Acknowledgments

We thank the members of the Nimer lab for their assistance and thoughtful input. We thank the Sylvester Comprehensive Cancer Center Flow Cytometry and Oncogenomics Core Facilities helping carry out this work. We also thank Delphine Prou for her assistance in preparing this manuscript. This work was supported by National Cancer Institute Grant R01 CA166835-01 (SN). Shiekhattar’s laboratory was supported by R01 GM078455 and R01 GM105754 from the National Institute of Health and Sylvester Comprehensive Cancer Center.

Footnotes

Authorship contributions:

S.D.N. supervised the project. G.C. designed and performed most of the experiments, analyzed the data and wrote the manuscript; F.Liu analyzed the data and revised the manuscript; T.A. , M. N., and F.Lai, performed some of the experiments and analyzed the data; H.X. generated the mouse colonies. S.C., S.G., PJ.H., K.A., C.M., M.T. and L.W. helped with some of the experiments.

Disclosure of conflicts of interest: the authors declare no competing financial interest.

Conflict of interest: The authors declare no conflict of interest.

Publisher's Disclaimer: This is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication. NPG are providing this early version of the manuscript as a service to our customers. The manuscript will undergo copyediting, typesetting and a proof review before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers apply

References

  • 1.Nimer SD. Myelodysplastic syndromes. Blood. 2008 May 15;111(10):4841–4851. doi: 10.1182/blood-2007-08-078139. [DOI] [PubMed] [Google Scholar]
  • 2.Maegawa S, Gough SM, Watanabe-Okochi N, Lu Y, Zhang NX, Castoro RJ, et al. Age-related epigenetic drift in the pathogenesis of MDS and AML. Genome Research. 2014 Apr;24(4):580–591. doi: 10.1101/gr.157529.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011 Jun 30;364(26):2496–2506. doi: 10.1056/NEJMoa1013343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Goodman RH, Smolik S. CBP/p300 in cell growth, transformation, and development. Genes Dev. 2000 Jul 1;14(13):1553–1577. [PubMed] [Google Scholar]
  • 5.Roelfsema JH, White SJ, Ariyurek Y, Bartholdi D, Niedrist D, Papadia F, et al. Genetic heterogeneity in Rubinstein-Taybi syndrome: mutations in both the CBP and EP300 genes cause disease. Am J Hum Genet. 2005 Apr;76(4):572–580. doi: 10.1086/429130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Iyer NG, Ozdag H, Caldas C. p300/CBP and cancer. Oncogene. 2004 May 24;23(24):4225–4231. doi: 10.1038/sj.onc.1207118. [DOI] [PubMed] [Google Scholar]
  • 7.Borrow J, Stanton VP, Jr, Andresen JM, Becher R, Behm FG, Chaganti RS, et al. The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nat Genet. 1996 Sep;14(1):33–41. doi: 10.1038/ng0996-33. [DOI] [PubMed] [Google Scholar]
  • 8.Kitabayashi I, Aikawa Y, Yokoyama A, Hosoda F, Nagai M, Kakazu N, et al. Fusion of MOZ and p300 histone acetyltransferases in acute monocytic leukemia with a t(8;22)(p11;q13) chromosome translocation. Leukemia. 2001 Jan;15(1):89–94. doi: 10.1038/sj.leu.2401983. [DOI] [PubMed] [Google Scholar]
  • 9.Yao TP, Oh SP, Fuchs M, Zhou ND, Ch'ng LE, Newsome D, et al. Gene dosage-dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell. 1998 May 1;93(3):361–372. doi: 10.1016/s0092-8674(00)81165-4. [DOI] [PubMed] [Google Scholar]
  • 10.Rebel VI, Kung AL, Tanner EA, Yang H, Bronson RT, Livingston DM. Distinct roles for CREB-binding protein and p300 in hematopoietic stem cell self-renewal. Proc Natl Acad Sci U S A. 2002 Nov 12;99(23):14789–14794. doi: 10.1073/pnas.232568499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chan WI, Hannah RL, Dawson MA, Pridans C, Foster D, Joshi A, et al. The Transcriptional Coactivator Cbp Regulates Self-Renewal and Differentiation in Adult Hematopoietic Stem Cells. Mol Cell Biol. 2011 Dec;31(24):5046–5060. doi: 10.1128/MCB.05830-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kung AL, Rebel VI, Bronson RT, Ch'ng LE, Sieff CA, Livingston DM, et al. Gene dose-dependent control of hematopoiesis and hematologic tumor suppression by CBP. Genes Dev. 2000 Feb 1;14(3):272–277. [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang L, Gural A, Sun XJ, Zhao X, Perna F, Huang G, et al. The leukemogenicity of AML1-ETO is dependent on site-specific lysine acetylation. Science. 2011 Aug 5;333(6043):765–769. doi: 10.1126/science.1201662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Shigeno K, Yoshida H, Pan L, Luo JM, Fujisawa S, Naito K, et al. Disease-related potential of mutations in transcriptional cofactors CREB-binding protein and p300 in leukemias. Cancer Lett. 2004 Sep 15;213(1):11–20. doi: 10.1016/S0304-3835(03)00442-7. [DOI] [PubMed] [Google Scholar]
  • 15.Ramos F, Robledo C, Izquierdo-Garcia FM, Suarez-Vilela D, Benito R, Fuertes M, et al. Bone marrow fibrosis in myelodysplastic syndromes: a prospective evaluation including mutational analysis. Oncotarget. 2016 May 24;7(21):30492–30503. doi: 10.18632/oncotarget.9026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013 Nov 21;122(22):3616–3627. doi: 10.1182/blood-2013-08-518886. quiz 3699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Duhoux FP, De Wilde S, Ameye G, Bahloula K, Medves S, Lege G, et al. Novel variant form of t(11;22)(q23;q13)/MLL-EP300 fusion transcript in the evolution of an acute myeloid leukemia with myelodysplasia-related changes. Leuk Res. 2011 Mar;35(3):e18–20. doi: 10.1016/j.leukres.2010.09.024. [DOI] [PubMed] [Google Scholar]
  • 18.Kasper LH, Fukuyama T, Biesen MA, Boussouar F, Tong CL, de Pauw A, et al. Conditional knockout mice reveal distinct functions for the global transcriptional coactivators CBP and p300 in T-cell development. Mol Cell Biol. 2006 Feb;26(3):789–809. doi: 10.1128/MCB.26.3.789-809.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xu HM, Menendez S, Schlegelberger B, Bae N, Aplan PD, Gohring G, et al. Loss of p53 accelerates the complications of myelodysplastic syndrome in a NUP98-HOXD13-driven mouse model. Blood. 2012 Oct 11;120(15):3089–3097. doi: 10.1182/blood-2012-01-405332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Research. 2000 Jan 1;28(1):27–30. doi: 10.1093/nar/28.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005 Oct 25;102(43):15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lin YW, Slape C, Zhang Z, Aplan PD. NUP98-HOXD13 transgenic mice develop a highly penetrant, severe myelodysplastic syndrome that progresses to acute leukemia. Blood. 2005 Jul 1;106(1):287–295. doi: 10.1182/blood-2004-12-4794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Graubert TA, Shen D, Ding L, Okeyo-Owuor T, Lunn CL, Shao J, et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat Genet. 2012 Jan;44(1):53–U77. doi: 10.1038/ng.1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pan J, Zhang Q, Wang YA, You M. 26S Proteasome Activity Is Down-Regulated in Lung Cancer Stem-Like Cells Propagated In Vitro. Plos One. 2010 Oct 11;5(10) doi: 10.1371/journal.pone.0013298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Offman J, Opelz G, Doehler B, Cummins D, Halil O, Banner NR, et al. Defective DNA mismatch repair in acute myeloid leukemia/myelodysplastic syndrome after organ transplantation. Blood. 2004 Aug 1;104(3):822–828. doi: 10.1182/blood-2003-11-3938. [DOI] [PubMed] [Google Scholar]
  • 26.Schinke C, Giricz O, Li WJ, Shastri A, Gordon S, Barreryo L, et al. IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood. 2015 May 14;125(20):3144–3152. doi: 10.1182/blood-2015-01-621631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Testa U, Riccioni R, Diverio D, Rossini A, Lo Coco F, Peschle C. Interleukin-3 receptor in acute leukemia. Leukemia. 2004 Feb;18(2):219–226. doi: 10.1038/sj.leu.2403224. [DOI] [PubMed] [Google Scholar]
  • 28.Gits J, van Leeuwen D, Carroll HP, Touw IP, Ward AC. Multiple pathways contribute to the hyperproliferative responses from truncated granulocyte colony-stimulating factor receptors. Leukemia. 2006 Dec;20(12):2111–2118. doi: 10.1038/sj.leu.2404448. [DOI] [PubMed] [Google Scholar]
  • 29.Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, Pettigrew AL, et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000 Oct;14(10):1777–1784. doi: 10.1038/sj.leu.2401903. [DOI] [PubMed] [Google Scholar]
  • 30.O'Sullivan LA, Liongue C, Lewis RS, Stephenson SEM, Ward AC. Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease. Mol Immunol. 2007 Apr;44(10):2497–2506. doi: 10.1016/j.molimm.2006.11.025. [DOI] [PubMed] [Google Scholar]
  • 31.Vainchenker W, Constantinescu SN. JAK/STAT signaling in hematological malignancies. Oncogene. 2013 May 23;32(21):2601–2613. doi: 10.1038/onc.2012.347. [DOI] [PubMed] [Google Scholar]
  • 32.Chung YJ, Park BB, Kang YJ, Kim TM, Eaves CJ, Oh IH. Unique effects of Stat3 on the early phase of hematopoietic stem cell regeneration. Blood. 2006 Aug 15;108(4):1208–1215. doi: 10.1182/blood-2006-01-010199. [DOI] [PubMed] [Google Scholar]
  • 33.Wang L, Gural A, Sun XJ, Zhao XY, Perna F, Huang G, et al. The Leukemogenicity of AML1-ETO Is Dependent on Site-Specific Lysine Acetylation. Science. 2011 Aug 5;333(6043):765–769. doi: 10.1126/science.1201662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wu M, Kwon HY, Rattis F, Blum J, Zhao C, Ashkenazi R, et al. Imaging hematopoietic precursor division in real time. Cell Stem Cell. 2007 Nov;1(5):541–554. doi: 10.1016/j.stem.2007.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Van Etten RA. Aberrant cytokine signaling in leukemia. Oncogene. 2007 Oct 15;26(47):6738–6749. doi: 10.1038/sj.onc.1210758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Baker SJ, Rane SG, Reddy EP. Hematopoietic cytokine receptor signaling. Oncogene. 2007 Oct;26(47):6724–6737. doi: 10.1038/sj.onc.1210757. [DOI] [PubMed] [Google Scholar]
  • 37.Giotopoulos G, Chan WI, Horton SJ, Ruau D, Gallipoli P, Fowler A, et al. The epigenetic regulators CBP and p300 facilitate leukemogenesis and represent therapeutic targets in acute myeloid leukemia. Oncogene. 2015 Apr 20; doi: 10.1038/onc.2015.92. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES