Prognostic relevance of disordered epigenetic regulation in juvenile myelomonocytic leukemia

2013 ◽  
Vol 225 (03) ◽  
Author(s):  
S Fluhr ◽  
T Witte ◽  
CF Krombholz ◽  
C Plass ◽  
CM Niemeyer ◽  
...  
2010 ◽  
Vol 28 (11) ◽  
pp. 1919-1927 ◽  
Author(s):  
Silvia Bresolin ◽  
Marco Zecca ◽  
Christian Flotho ◽  
Luca Trentin ◽  
Andrea Zangrando ◽  
...  

Purpose Juvenile myelomonocytic leukemia (JMML) is a rare early childhood myelodysplastic/myeloproliferative disorder characterized by an aggressive clinical course. Age and hemoglobin F percentage at diagnosis have been reported to predict both survival and outcome after hematopoietic stem cell transplantation (HSCT). However, no genetic markers with prognostic relevance have been identified so far. We applied gene expression–based classification to JMML samples in order to identify prognostic categories related to clinical outcome. Patients and Methods Samples of 44 patients with JMML were available for microarray gene expression analysis. A diagnostic classification (DC) model developed for leukemia and myelodysplastic syndrome classification was used to classify the specimens and identify prognostically relevant categories. Statistical analysis was performed to determine the prognostic value of the classification and the genes identifying prognostic categories were further analyzed through R software. Results The samples could be divided into two major groups: 20 specimens were classified as acute myeloid leukemia (AML) –like and 20 samples as nonAML-like. Four patients could not be assigned to a unique class. The 10-year probability of survival after diagnosis of AML-like and nonAML-like patients was significantly different (7% v 74%; P = .0005). Similarly, the 10-year event-free survival after HSCT was 6% for AML-like and 63% for nonAML-like patients (P = .0010). Conclusion Gene expression–based classification identifies two groups of patients with JMML with distinct prognosis outperforming all known clinical parameters in terms of prognostic relevance. Gene expression–based classification could thus be prospectively used to guide clinical/therapeutic decisions.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4890-4890 ◽  
Author(s):  
Y. Lucy Liu ◽  
Shelly Y. Lensing ◽  
Yan Yan ◽  
Cody Webster ◽  
Peter D. Emanuel

Abstract Juvenile myelomonocytic leukemia (JMML) is a mixed myelodysplastic /myeloproliferative disorder (MDS/MPD). It occurs in infancy and young children with a progressive course leading to death within one year after diagnosis. This disease is characterized by monocytosis, leukocytosis, elevated fetal hemoglobin, hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF), a low percentage of myeloblasts in the bone marrow, and absence of the Philadelphia chromosome or the BCR/ABL fusion gene. Mutations or other abnormalities in RAS, NF1, PTPN11, and CBL have been linked to be responsible for the pathogenesis of JMML in up to 85% of cases. Treatment is very difficult in JMML, and only allogeneic stem cell transplantation (SCT) can extend survival. However, the relapse rate from allogeneic SCT is inordinately high in JMML (28-55%), with 5-year disease-free survival rates of 25-40%. JMML occurs in an age-range when genes are actively being turned on or off in children in adaption to the oxygenized environment after birth. Epigenetics plays a key role in this developmental plasticity. We previously reported hypermethylation on the promoter of PTEN in 77% of JMML patients, and decitabine, a DNA-hypomethylating reagent, significantly inhibited colony formation (CFU-GM) in JMML cells in vitro. In addition, other groups found that aberrant DNA methylation on promoters of BMP4, CALCA, CDKN2B, and RARB is significantly associated with poor prognosis in JMML. Taking together, these data suggest that epigenetic mechanisms may contribute to the pathogenesis of JMML. MicroRNAs (miRNAs) have been reported to play an important role in myeloid differentiation and activation. miRNA function is highly dependent on the cell type. Recently, we reported that miR-183 is overexpressed in JMML. Other groups have reported aberrant expression of miR-29a in acute myeloid leukemia and other cancers. Both miR-183 and miR-29a are located on chromosome 7q32 in humans, which is frequently disrupted in JMML. We hypothesized that miR-29a may be deregulated in JMML, and contribute to the aberrant epigenetic regulation in JMML. In order to test our hypothesis, we collected peripheral blood or bone marrow from 41 JMML patients and 14 normal individuals. Total RNAs were extracted from mononuclear cells (MNCs) using Trizol. We first evaluated the expression levels of miR-29a by using relative-quantitative real-time RT-PCR (qRT-PCR). We found that the expression levels (RQ) of miR-29a in patients are significantly lower than that in normal individuals (median 0.45 vs. 1.11, p< 0.001). By analysis of the expression levels of miR-29a together with previous data on miR-183 in these JMML patients and normal controls, we found that the RQ of miR-29a is inversely correlated with RQ of miR-183 (Spearman’s r=-51, p<0.001). This suggests that the expression of miR-29a is segregated from that of miR-183 in MNCs, although they are located only 1.1 million base-pairs apart on chromosome 7q32. When further evaluating the mRNA expression levels of miR-29a targeting genes by using qRT-PCR, we found that RQ of DNA methyltransferases 3A and 3B (DNMT3a and DNMT3b) were significantly increased in JMML patients’ samples in comparison with normal controls (median 5.16 vs. 1.00, p=0.001 for DNMT3a; median 3.45 vs. 1.08, p=0.001). Strikingly, the RQ of miR-29a was inversely correlated with the RQs of both DNMTs (Spearman’s r=-0.62 for DNMT3a; Spearman’s r=-0.79 for DNMT3b, p<0.001), which suggests that miR-29a plays a significant role in the regulation of DNMT 3a and DNMT3b in MNCs. In conclusion, we found that overexpression of DNMT3a and DNMT3b, the two key molecules in DNA methylation of epigenetic modification in developmental plasticity, is related to downregulation of miR-29a in JMML. In addition to our previous report on overexpression of miR-183 in JMML, this finding provides new insights into the role of miRNAs in the aberrant epigenetic regulation in JMML, and it may apply to other pediatric malignancies. Further investigation is ongoing to uncover the mechanism of down-regulated miR-29a in JMML. If the underexpression of miR-29a in JMML is due to the hypermethylation on miR-29a promoter, as reported in prostate cancer cells, demethylating agents, such as decitabine, or soybean products, isoflavone, could be potential drugs to treat JMML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 438-438
Author(s):  
Silvia Fluhr ◽  
Christopher Felix Krombholz ◽  
Angelina Meier ◽  
Christoph Plass ◽  
Charlotte Niemeyer ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative disorder of early childhood characterized by massive proliferation of not only the monocytic and granulocytic lineage but also of erythropoietic precursors. Elevated levels of fetal hemoglobin (HbF) are found at time of diagnosis in more than half of JMML cases and are associated with poor outcome. We and others previously found that a key molecular feature of many JMML cases is DNA hypermethylation of distinct target genes, which also has adverse prognostic relevance. It is long known that epigenetic processes are involved in hemoglobin regulation. We therefore hypothesized that epigenetic dysregulation would also be involved in aberrant hemoglobin expression in JMML erythroblasts. ɣ-globin silencing, which is completed one year after birth under physiological conditions, is associated with dense CpG methylation of the ɣ-globin promoter. Consistent with re-expression of ɣ-globin in JMML cases with elevated HbF, CpG methylation at the ɣ-globin promoter was decreased in purified erythroblasts from JMML patients with elevated HbF compared to JMML with normal HbF (average methylation 40.0% ±4.2%, N=6 vs. 72.0% ±3.5%, N=4) (P<0.001). CpG methylation at the ɣ-globin promoter inversely correlated with the relative amount of ɣ-globin expression (ɣ/(β+ɣ)) as determined by RT-qPCR in JMML erythroblasts. Correspondingly, the β-globin promoter is normally unmethylated in erythroblasts from healthy adults, and was also found to be unmethylated in erythroblasts from a JMML patient with normal HbF, whereas it was hypermethylated in JMML with elevated HbF (average methylation 67.0% ±26.0%, N=2). We also assessed CpG methylation at the KLF1 gene which encodes an erythroid-specific transcription factor and strong β-globin activator. We found that the KLF1 enhancer (-593 to -312 relative to the transcription start site of KLF1) and promoter (-182 to +203) regions were unmethylated in healthy erythroblasts irrespective of neonatal or adult origin (N=11; enhancer, 4.5% ±0.9%; promoter, 2.2% ±0.6%), whereas both regions were densely methylated (>80%) in healthy non-erythroid cell populations. By contrast, the KLF1 enhancer and promoter were aberrantly methylated in JMML erythroblasts (N=11; enhancer, 21.2% ±4.7%, P=0.002; promoter, 11.5% ±2.9%, P=0.004). This suggested a regulatory role of KLF1 methylation in aberrant globin expression in JMML. To explore this functionally, we used a dual luciferase reporter assay, ligating the KLF1 enhancer into a CpG-free reporter vector and comparing the reporter activity of unmethylated vector with that of in vitro methylated vector after transfection into K562 cells. CpG methylation at the KLF1 enhancer abolished its activity, supporting methylation-associated silencing of KLF1 in JMML erythroblasts. Compatible with this, KLF1 transcript levels were reduced 2.1-fold in JMML erythroblasts (N=12) compared to healthy erythroblasts (N=9) as assessed by RT-qPCR (P=0.018). We additionally assessed the protein expression of the direct KLF1 target CD44 on the surface of JMML erythroblasts by FACS analysis. CD44 expression was 2.4-fold lower in JMML erythroblasts with high KLF1 enhancer methylation compared to JMML erythroblasts with normal KLF1 enhancer methylation. In summary, we show that multiple layers of hemoglobin regulation are affected by epigenetic changes in JMML erythroblasts. The globin genes themselves are targets of aberrant DNA methylation in JMML erythroblasts with elevated HbF. Moreover, the β-globin activator KLF1 is aberrantly methylated and repressed in JMML erythroblasts. These findings provide for the first time a mechanistic explanation for the strong correlation and uniform prognostic relevance of elevated HbF and gene hypermethylation in JMML. Disclosures No relevant conflicts of interest to declare.


Author(s):  
William A. Lambert ◽  
Joseph A. DiGiuseppe ◽  
Tatiana Lara-Ospina ◽  
Markus J. Bookland ◽  
Jonathan E. Martin ◽  
...  

Leukemia ◽  
2021 ◽  
Author(s):  
Astrid Behnert ◽  
Julia Meyer ◽  
Jahan-Yar Parsa ◽  
Aaron Hechmer ◽  
Mignon L. Loh ◽  
...  

Author(s):  
Shrinidhi Nathany ◽  
Gaurav Chatterjee ◽  
Shruti Ghai ◽  
Nirmalya Roy Moulik ◽  
Dhanalaxmi Shetty ◽  
...  

2013 ◽  
Vol 37 ◽  
pp. S93-S94
Author(s):  
A. Eischen ◽  
C. Paillard ◽  
M. Besse ◽  
A. Spiegel ◽  
P. Lutz ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document