U.S. flag

An official website of the United States government

Display Settings:

Items per page

PMC Full-Text Search Results

Items: 9

1.
Figure 4.

Figure 4. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

MF-initiating cells are present in the BM and spleen of JAK2V617F/Ezh2Δ/Δ mice. (a) Levels of TPO protein in the serum of WT (n = 6), Ezh2Δ/Δ (n = 4), JAK2V617F (n = 4), and JAK2V617F/Ezh2Δ/Δ (n = 4) mice 1–2 mo after the deletion of Ezh2. (b) qRT-PCR analysis of Il6 and Il1β in MEPs from WT (n = 6), Ezh2Δ/Δ (n = 8), JAK2V617F (n = 8), and JAK2V617F/Ezh2Δ/Δ (n = 8) mice 1–2 mo after the deletion of Ezh2. (c and d) Chimerism of CD45.2+ donor cells in Mac1+ myeloid cells, B220+ B cells, and CD4+/CD8+ T cells in the PB (c) and BM LSK cells (d) of CD45.1+ recipients (n = 5 each) at 3 mo after transplantation of BM and spleen LSKs isolated from WT and JAK2V617F/Ezh2Δ/Δ mice. (a–d) Bars and asterisks show the mean ± SEM and *, P < 0.05; **, P < 0.01; and ***, P < 0.001 by the Student’s t test; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
2.
Figure 3.

Figure 3. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

The loss of Ezh2 promotes the development of EMH. (a) Spleen weight of WT (n = 5), Ezh2Δ/Δ (n = 5), JAK2V617F (n = 5), and JAK2V617F/Ezh2Δ/Δ (n = 9) mice 4 wk after the deletion of Ezh2. (b) Proportions of Gr-1+ and/or Mac-1+ myeloid, B220+ B cells, CD4+ or CD8+ T cells, and Ter119+ erythroid cells among CD45+ hematopoietic cells in the spleen. (c) Histology of the spleen from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice observed by hematoxylin-eosin staining (top) and silver staining (bottom). Bars, 50 µm. (d) Proportions of LSKs and MkPs (LinSca-1c-Kit+CD150+CD41+) in the spleen of WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (n = 5–9) 4 wk after the deletion of Ezh2. (a, b, and d) Bars and asterisks show the mean ± SEM and *, P < 0.05; **, P < 0.01; and ***, P < 0.001 by the Student’s t test; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
3.
Figure 5.

Figure 5. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

Megakaryocyte-restricted deletion of Ezh2 contributes to the progression of JAK2V617F-induced fibrosis. (a) qRT-PCR analysis of Ezh2 in LSKs, MkPs, and GMPs from WT and JAK2V617F/Ezh2MkΔ/Δ cells at 2–3 mo after transplantation. (b) CBC of WT (n = 5), Ezh2MkΔ/Δ (n = 4), JAK2V617F (n = 6), and JAK2V617F/Ezh2MkΔ/Δ (n = 10) mice 5 mo after transplantation. (c) Spleen weight of WT (n = 5), Ezh2MkΔ/Δ (n = 4), JAK2V617F (n = 4), and JAK2V617F/Ezh2MkΔ/Δ (n = 5) mice 5 mo after transplantation. (d) Proportions of LSK cells and MkPs in the spleen of WT and JAK2V617F/Ezh2MkΔ/Δ mice (n = 3–5) 5 mo after transplantation. (e) Histology of the BM and spleen from WT, Ezh2MkΔ/Δ, JAK2V617F, and JAK2V617F/Ezh2MkΔ/Δ mice observed by hematoxylin-eosin staining (middle) and silver staining (top and bottom). Bars, 100 µm. (f) BM fibrosis grading (grades 0–3; ) of WT, Ezh2MkΔ/Δ, JAK2V617F, and JAK2V617F/Ezh2MkΔ/Δ (n = 3–5) mice 5 mo after transplantation. (a–d) Bars and asterisks show the mean ± SEM and *, P < 0.05; and **, P < 0.01 by the Student’s t test. (f) Asterisk shows *, P < 0.05 by the Mann–Whitney U test; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
4.
Figure 2.

Figure 2. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

The loss of Ezh2 promotes the development of JAK2V617F-induced MF. (a) BM cell counts (two femurs and two tibias) of WT (n = 5), Ezh2Δ/Δ (n = 5), JAK2V617F (n = 5), and JAK2V617F/Ezh2Δ/Δ (n = 9) mice 4 wk after the deletion of Ezh2. (b) Proportions of Gr-1+Mac-1+ neutrophils, Mac-1+ monocytes, B220+ B cells, and CD4+/CD8+ T cells among CD45+ hematopoietic cells in the BM 4 wk after the deletion of Ezh2. (c) Histology of the BM from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice observed by hematoxylin-eosin staining (top) and silver staining (bottom). (d) Histology of the BM from WT and JAK2V617F/Ezh2Δ/Δ mice observed by hematoxylin-eosin staining (an arrow indicates a megakaryocyte with emperipolesis). (e) Proportions of CD150+CD34LSK LT-HSCs in the BM of WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (n = 3 each) 4 wk after the deletion of Ezh2. (f and g) Proportions of LSKs (f) and GMPs (LinSca-1c-Kit+CD34+FcγR+; g) in the BM of WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (n = 3 each). (h) Proportions of CD71+Ter119+ double-positive erythroblasts in the BM of WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (n = 5 each) 4 wk after the deletion of Ezh2. (i) Proportions of Annexin V+ apoptotic cells in CD71+Ter119+ erythroblasts in the BM of WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (n = 3 each). (a, b, and e–i) Bars and asterisks show the mean ± SEM and *, P < 0.05; **, P < 0.01; and ***, P < 0.001 by the Student’s t test; two independent experiments. Bars: (c) 50 µm; (d) 10 µm.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
5.
Figure 1.

Figure 1. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

The loss of Ezh2 severely compromises hematopoiesis in the presence of the JAK2V617F mutant. (a) Experimental scheme of our model mouse using JAK2V617F transgenic and Ezh2 conditional KO BM cells. (b) qRT-PCR analysis of Ezh2 in LSKs and MEPs from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice 4 wk after the Cre-mediated deletion of Ezh2. (c) Verification of elimination of Ezh2 protein and levels of H3K27me3 in LK cells detected by Western blotting. (d) CBC of WT (n = 5), Ezh2Δ/Δ (n = 5), JAK2V617F (n = 5), and JAK2V617F/Ezh2Δ/Δ (n = 10) mice 4 wk after the deletion of Ezh2 and moribund JAK2V617F/Ezh2Δ/Δ mice (n = 10). (e) Proportions of myeloid (Gr-1+ and/or Mac-1+), B220+ B cells, and CD4+ or CD8+ T cells among CD45.2+ donor–derived hematopoietic cells in the PB (JAK2V617F/Ezh2Δ/Δ n = 10, others n = 5). (f) Dysplastic red blood cells in JAK2V617F/Ezh2Δ/Δ mice observed by May-Grünewald-Giemsa staining. Bars, 10 µm. (g) Kaplan-Meier survival curves of WT (n = 6), Ezh2Δ/Δ (n = 6), JAK2V617F (n = 8), JAK2V617F/Ezh2Δ/+ (n = 9), and JAK2V617F/Ezh2Δ/Δ (n = 10) mice; three independent experiments were performed. ***, P < 0.0001 by the log-rank test. (h) CBC of WT (n = 5), Ezh2Δ/Δ (n = 5), JAK2V617F (n = 6), and JAK2V617F/Ezh2Δ/+ (n = 9) mice 4 mo after the deletion of Ezh2. (a, d, e, and h) Bars and asterisks show the mean ± SEM and *, P < 0.05; **, P < 0.01; and ***, P < 0.001 by the Student’s t test; two independent experiments. (b and c) Data are shown as mean ± SD; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
6.
Figure 8.

Figure 8. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

Brd4 inhibition abrogates the MF-initiating capacity of JAK2V617F/Ezh2Δ/Δ cells. (a) Experimental scheme of the JQ1 treatment of chimeric mice reconstituted with WT and mutant cells. (b) Fold changes in chimerism of CD45.2+ mutant cells in Mac1+ myeloid cells in the PB of recipients (n = 5 each) 8 wk after JQ1 administration compared with that of pre-JQ1 administration (red bars show the mean). (c) Experimental scheme of the JQ1 treatment of WT mice and JAK2V617F/Ezh2Δ/Δ mice. (d) Hemoglobin levels of WT (open circles) and JAK2V617F/Ezh2Δ/Δ (closed circles) mice pretreatment and 7 d after the completion of the JQ1 treatment. (e) Histology and fibrosis grading (grade 0–3) of the BM from DMSO-treated (n = 7) and JQ1-treated (n = 6) JAK2V617F/Ezh2Δ/Δ mice observed by silver staining. Bars, 50 µm. (f) Spleen weight of DMSO-treated (n = 5) or JQ1-treated (n = 4) JAK2V617F/Ezh2Δ/Δ mice 4 wk after the end of the treatment. (g) Proportions of LSKs and MkPs in the spleen of DMSO-treated and JQ1-treated JAK2V617F/Ezh2Δ/Δ mice 4 wk after the completion of the treatment. (h and i) Chimerism of CD45.2+ cells in the PB (h) and BM LSKs, spleen LSKs, and spleen MkPs (i) of CD45.1+ recipients (n = 5 each) at 4 mo after transplantation of 106 spleen cells isolated from DMSO-treated or JQ1-treated WT and JAK2V617F/Ezh2Δ/Δ mice. (d and f–i) Bars and asterisks show the mean ± SEM and *, P < 0.05 by the Student’s t test (d and f–h) or *, P < 0.05 and **, P < 0.01 by the Mann–Whitney U test (i); two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
7.
Figure 6.

Figure 6. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

JAK2V617F and the loss of Ezh2 cooperatively alter transcriptional programs of hematopoiesis. (a) Venn diagrams showing overlaps of up- and down-regulated genes (left and right, respectively) between Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ LSKs and MEPs isolated 4 wk after the deletion of Ezh2 relative to their WT counterparts. (b) Hierarchical clustering based on total gene expression in LSKs and MEPs isolated from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (linkage scores are indicated on the right). (c) A principal component (PC) analysis based on total gene expression in LSKs (open circles) and MEPs (closed circles) isolated from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice. (d) GSEA plots for Stat5 up-regulated genes defined in human HSPCs comparing mouse LSKs isolated from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice. (e) Levels of the mean fluorescence intensity (MFI) of phospho-Stat5 (p-Y694) in LSKs from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ (n = 3 each) mice after serum starvation (“(−)”) and stimulation of IL-3 (“IL-3”). Bars show the mean ± SEM; three independent experiments. (f) GSEA plots for canonical PRC2 targets defined in LSK HSPCs comparing LSKs isolated from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice. (g) GSEA for gene expression signatures of HSCs and MkPs comparing LSKs isolated from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ mice (the NES, p-value, and FDR q-value [top and bottom rows, respectively] relative to WT LSKs are shown in each cell). (h) GSEA plots for the HSC signature in LSKs isolated from JAK2V617F/Ezh2Δ/Δ compared with JAK2V617F mice. (a–d and f–h) Experiments used cells from two to four mice individual mice per genotype. (d and f–h) The normalized enrichment score (NES), nominal p-value, and false discovery rate (FDR) q-value are indicated.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
8.
Figure 7.

Figure 7. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

Alterations in H3K27me3 upon the loss of Ezh2 results in the activation of specific oncogenes. (a) Fold enrichment (ChIP/input) values of H3K27me3 (TSS ± 2.0 kb of RefSeq genes) in WT and JAK2V617F/Ezh2Δ/Δ LSKs 4 wk after the deletion of Ezh2. (b) A scatter plot showing the relationship of fold enrichment (ChIP/input) values of H3K27me3 (TSS ± 2.0 kb of RefSeq genes) between WT and JAK2V617F/Ezh2Δ/Δ LSKs 4 wk after the deletion of Ezh2. The light diagonal line represents the borderlines for twofold changes in H3K27me3 levels. (c) Fold enrichment (ChIP/input) values of H3K27me3 at TSS ± 2.0 kb of MkP signature genes in WT and JAK2V617F/Ezh2Δ/Δ LSKs. (d) A scatter plot showing the relationship of expression of RefSeq genes in JAK2V617F/Ezh2Δ/Δ LSKs relative to WT LSKs and H3K27me3 levels of RefSeq genes in JAK2V617F/Ezh2Δ/Δ LSKs relative to WT LSKs and potential oncogenes activated in JAK2V617F/Ezh2Δ/Δ LSKs (see also Table S2). (e) ChIP-seq view of H3K27me3 levels at the promoter region of Hmga2 in WT and JAK2V617F/Ezh2Δ/Δ LSKs 4 wk after the deletion of Ezh2. (f) qRT-PCR analysis of Hmga2 in LSKs and MEPs from WT, Ezh2Δ/Δ, JAK2V617F, and JAK2V617F/Ezh2Δ/Δ (n = 3–4) mice 1–2 mo after the deletion of Ezh2. (g) The total cell counts of WT (black lines) and JAK2V617F (red lines) HSCs transduced with either a control (straight lines) or an Hmga2 (broken lines) retrovirus were monitored for 8 d. (h) Representative pictures of Hmga2-transduced or control vector–transduced WT and JAK2V617F HSCs on day 8 of the culture observed by May-Grünewald-Giemsa staining. Bars: 10 µm (high magnitude); 100 µm (low magnitude). (i) Proportions of megakaryocytes in Hmga2-transduced or control vector–transduced WT and JAK2V617F HSCs on day 8 of the culture. (a and c) Boxes and whiskers show the mean and minimum to maximum, and asterisks show ***, P < 0.001 by the Student’s t test; experiments used cells from two to four individual mice per genotype. (f, g, and i) Bars and asterisks show the mean ± SEM and *, P < 0.05; **, P < 0.01; and ***, P < 0.001 by the Student’s t test; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.
9.
Figure 9.

Figure 9. From: The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition.

Brd4 inhibition restores de-repression of PRC2 target genes. (a) Fold enrichment (ChIP/input) values of H3K27ac at the enhancer and the promoter regions in JAK2V617F/Ezh2Δ/Δ LK cells 2 wk after the completion of the JQ1 treatment. (b) A scatter plot showing the relationship of fold enrichment (ChIP/input) values of H3K27ac (TSS ± 2.0 kb of RefSeq genes) between DMSO- and JQ1-treated JAK2V617F/Ezh2Δ/Δ LK cells (the light diagonal line represents the borderlines for twofold changes in H3K27ac levels). (c) GSEA plot for the canonical PRC2 targets defined in comparing LSKs in JQ1-treated with DMSO-treated JAK2V617F/Ezh2Δ/Δ mice. (d) Venn diagrams showing overlaps between PRC2 target genes and JQ1 target genes, which were defined in b and are shown in Table S1. (e) Fold enrichment (ChIP/input) values of H3K27ac at the promoter regions of the canonical PRC2 target genes (defined in ) in JAK2V617F/Ezh2Δ/Δ LK cells 2 wk after the completion of the JQ1 treatment. (f) ChIP-seq view of H3K27ac levels at the promoter region of Hmga2 in JQ1-treated or nontreated JAK2V617F/Ezh2Δ/Δ LK cells. (g) qRT-PCR analysis of the expression of Hmga2 in LSKs and MEPs isolated from DMSO-treated or JQ1-treated WT mice (n = 3) and JAK2V617F/Ezh2Δ/Δ mice (n = 3) 2 wk after the completion of the treatment. (h) GSEA plot for the MkPs signature genes described in comparing LSK cells from JQ1-treated and DMSO-treated JAK2V617F/Ezh2Δ/Δ mice. (i) GO biological process gene sets that were enriched more in JQ-treated LSK cells than in DMSO-treated LSK cells as determined by GSEA (gene sizes and FDR q-values are shown in the top and bottom x axes). (a and e) Boxes and whiskers show the mean and minimum to maximum. The asterisks show ***, P < 0.001 by the Student’s t test. (c, h, and i) The normalized enrichment score (NES), nominal p-value, and false discovery rate (FDR) q-value are indicated. (a–f, h, and i) Experiments used cells from two individual mice per group. (g) Bars and asterisks show the mean ± SEM and *, P < 0.05 by the Student’s t test; two independent experiments.

Goro Sashida, et al. J Exp Med. 2016 Jul 25;213(8):1459-1477.

Display Settings:

Items per page

Supplemental Content

Recent activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...
Support Center