AML1/RUNX1 point mutation possibly promotes leukemic transformation in myeloproliferative neoplasms

Blood ◽  
2009 ◽  
Vol 114 (25) ◽  
pp. 5201-5205 ◽  
Author(s):  
Ye Ding ◽  
Yuka Harada ◽  
Jun Imagawa ◽  
Akiro Kimura ◽  
Hironori Harada

Abstract Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by proliferation of one or more myeloid cell lineages. Some patients exhibit leukemic transformation (LT) by unknown mechanisms, and chemotherapy may increase the risk of LT. To clarify the molecular mechanisms of LT, gene alterations involved in LT from patients in the chronic phase (CP) of MPNs were identified. Among 18 patients who progressed to leukemia, AML1/RUNX1 mutations were detected in 5 patients at the LT but in none at the CP. To investigate the leukemogenic effect of AML1/RUNX1 mutants, the AML1D171N mutant was transduced into CD34+ cells from patients in the CP of MPNs. The D171N transduction resulted in proliferation of immature myeloid cells, enhanced self-renewal capacity, and proliferation of primitive progenitors. Taken together, these results indicate that AML1/RUNX1 point mutations may have a leukemogenic potential in MPN stem cells, and they may promote leukemic transformation in MPN.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1902-1902
Author(s):  
Jun Imagawa ◽  
Yuka Harada ◽  
Ye Ding ◽  
Akiro Kimura ◽  
Hironori Harada

Abstract Abstract 1902 Poster Board I-925 Myeloproliferative disorders (MPD), including polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF), are clonal hematopoietic stem cell disorders characterized by proliferation of one or more myeloid cell lineages; they are associated with a high frequency of the Janus kinase 2 (JAK2) mutation V617F (JAK2V617F). Some patients with MPD exhibit leukemic transformation (LT) after several years of disease, but the mechanism of LT has been a matter of some controversy due to insufficient insight into the underlying molecular pathogenesis. LT may be a natural sequela of these diseases, whereas treatment with alkylating agents, hydroxycarbamide or their combination may increase the risk of LT. Previous studies have reported that chromosomal abnormalities including -5/5q- and -7/7q- were frequently detected in patients with MPD at the time of LT, suggesting that the development of cytogenetic abnormalities may be associated with the LT of patients with MPD. However, the genes involved in LT still remain obscure. Therefore, we attempted to identify gene alterations involved in LT from patients in the chronic phase (CP) of MPD. Patients with MPD were diagnosed and classified according to the WHO criteria between 1985 and 2007. Patients with PV, ET, PMF, MPD unclassified (MPD-U) and LT of these diseases were examined as approved by the institutional review board at Hiroshima University. Patients gave written informed consent, according to the Declaration of Helsinki. Mutations of JAK2V617F, AML1, CEPBA, FLT3, N-RAS, c-KIT, PTPN11 and TP53 were screened and were identified. Among 417 patients with MPD, 18 (4.3 %) patients progressed to leukemia. At LT, 13 of the 18 patients showed additional cytogenetic abnormalities, including -7/7q- with trisomy 21, inv(3)(q21q26), i(17)(q10) and t(11;19)(q23;p13.3), which are known to be associated with therapy-related leukemia. JAK2V617F was detected in 10 of 14 patients at LT. No patients had a JAK2 mutation pattern that changed during CP to LT. We analyzed gene mutations that may play an important role in leukemogenesis and found five AML1 mutations, one PTPN11 mutation and one CEBPA mutation in patients at the LT, whereas no mutation was detected in patients at CP. The gene alterations were detected at LT in both JAK2V617F-positive and −negative MPD patients, raising the possibility that the hematopoietic stem cells (HSCs) may have been transformed into leukemic blasts as a result of gene abnormalities. Among these gene abnormalities in the MPN patients, we focused on AML1 mutations. To clarify the leukemogenic effect of AML1 mutants, the AML1D171N mutant was transduced into CD34+ cells from eight patients in the CP of MPD using retrovirus transduction methods. The effect of this mutant on cell differentiation/proliferation was assessed by CFC re-plating assays. The D171N plates contained fewer erythroid colonies and more myeloid colonies than the controls. After re-plating, new colonies were detected on all D171N plates but on only few control plates. The D171N-transduced cells contained more CD34+ cells and proliferated more strongly than the controls. These results indicate that the D171N mutant has the potential to increase the myeloid immature cells and to enhance their self-renewal capacity. Furthermore, D171N-transduced cells retained more CD34+ cells than the controls after long-term culture on MS5 stroma cells, and showed significantly more colonies in long-term culture-initiating cells experiments. In this study, AML1 point mutations were detected with high frequency in patients at the LT from both JAK2V617F-positive and -negative MPD. Furthermore, the AML1D171N mutant transduced into CD34+ cells from MPD patients promoted proliferation of primitive progenitors, i.e. leukemic stem cells. These results indicate that AML1 point mutations may have a leukemogenic potential in JAK2V617F-positive stem cells or in pre-JAK2 stem cells, and they may promote leukemic transformation in MPD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1280-1280
Author(s):  
Nils Heinrich Thoennissen ◽  
Utz O. Krug ◽  
Dhong Hyun Lee ◽  
Norohiko Kawamata ◽  
Terra L Lasho ◽  
...  

Abstract Abstract 1280 Poster Board I-302 Philadelphia-chromosome negative myeloproliferative neoplasms (MPNs) including polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF) are defined as clonal hematopoietic stem cell disorders. These disorders show an inherent tendency for transformation into leukemia (MPN-blast phase) which is hypothesized to be accompanied by acquisition of additional genomic lesions. We, therefore, obtained a comprehensive profile of genomic alterations associated with leukemic transformation by using single-nucleotide polymorphism (SNP) array in 88 MPN patients, as well as 71 cases with MPN-blast phase, and correlated these findings with their clinical parameters. A relatively high number of genomic alterations was found in MPN after leukemic transformation with 4.6 ± 0.6 abnormalities per sample compared to only 1.4 ± 0.2 changes per patient in chronic phase (p<0.001). Compared to the cytogenetic data, SNP-chip analysis detected about 47% additional chromosomal changes in the MPN samples, and 31% more in the MPN-blast phase cases, whereas SNP-array allelokaryotyping practically captured all cytogenetic abnormalities in our study population. Several additionally altered regions were detected in patients with MPN-blast phase compared to chronic phase, including both deletion and copy-number neutral-loss of heterozygosity (CNN-LOH) on chromosome 12p (9%) and 21q (9%), involving ETV6 and RUNX1. Notably, deletion and CNN-LOH on 17p involving TP53 were diagnosed in 18% of MPN-blast phase samples, which was highly associated with preceding treatment with alkylating agents (p=0.016). Moreover, trisomy 8, as well as amplification of 8q24.21 involving the MYC gene, were detected in 13% of patients with MPN-blast phase who were almost exclusively negative for the JAK2V617F mutation. Genome-wide inspection of further critical regions with promising new candidate genes involved in the evolution to the MPN-leukemic phase included deletion and CNN-LOH on 7q22.1 (SH2B2) in 18%, duplication/amplification on 19p13.2 (PIN1, ICAM1, CDC37) in 13% and 21q22.2 (ERG) in 9% of MPN patients with blast crisis. In contrast, we detected a decreased frequency of JAK2V617F in MPN-blast phase samples (52%) compared to chronic phase (71%). Also, the percentage of patients with homozygous mutant JAK2 as a result of CNN-LOH was lower in the MPN-blast phase (43%) compared to the chronic phase (53%). Taken together, the data suggest that gain-of-function mutation of JAK2 is not a perquisite for leukemic transformation. Remarkably, CNN-LOH on either 7q or 9p was related to decreased survival after leukemic transformation (p=0.02 and p=0.012, respectively). Given the variety of allelic imbalances, our data suggest that MPN-blast phase appears to be a heterogeneous disease prone to have evolved multiple mechanisms to provide a proliferative advantage to the abnormal leukemic clone. Our analysis of MPN genomes in the chronic compared to the leukemic stage provided new prognostic insights, as well as novel causative genes which might be involved in the transformation to MPN-blast phase. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 907-907
Author(s):  
Yoshihiro Hayashi ◽  
Hideyo Hirai ◽  
Hisayuki Yao ◽  
Satoshi Yoshioka ◽  
Akihiro Tamura ◽  
...  

Abstract Abstract 907 Enhanced proliferation and differentiation of myeloid cells are features common to emergency granulopoiesis and chronic phase of chronic myeloid leukemia (CP-CML). We have previously shown that C/EBPβ, a transcription factor regulating emergency granulopoiesis, is upregulated in hematopoietic stem/progenitors in CP-CML and that C/EBPβ promotes BCR-ABL-mediated myeloid cell expansion (ASH annual meeting abstract. 2011; 118: 3747). However, the molecular mechanisms involved in the upregulation of C/EBPβ and the effects of C/EBPβ on the CML stem cells remained to be elucidated. Here we show that STAT5 is involved in the BCR-ABL-mediated C/EBPβ upregulation and that C/EBPβ accelerates the exhaustion of CML stem cells. In order to investigate the regulation of C/EBPβ in CP-CML, BCR-ABL gene was retrovirally introduced into EML cells, a mouse hematopoietic stem cell line, to make EML-BCR-ABL. The expression of C/EBPβ was upregulated at mRNA and protein level in EML-BCR-ABL when compared to EML cells transduced with a control vector. The upregulation of C/EBPβ was significantly repressed by adding imatinib mesylate, suggesting that the downstream signaling pathway of BCR-ABL is directly involved in the process. Neither a PI3K inhibitor nor a MEK inhibitor affected the levels of C/EBPβ in EML-BCR-ABL, but a STAT5 inhibitor reversed the upregulation of C/EBPβ in EML-BCR-ABL. Retroviral transduction of dominant-negative STAT5 mutant also impaired the upregulation of C/EBPβ in EML-BCR-ABL. Transduction of constitutively-active STAT5 mutant into parental EML cells significantly upregulated C/EBPβ expression. These results suggest that BCR-ABL upregulates C/EBPβ expression at least in part through activation of STAT5. To clarify the role of C/EBPβ in the regulation of CML stem cells, BCR-ABL-transduced bone marrow (BM) cells from C/EBPβ knockout (KO) mice or wild type (WT) mice were serially transplanted into irradiated recipient mice. In the first round of transplantation, all the recipient mice developed myeloproliferative status and the mice transplanted with BCR-ABL-transduced C/EBPβ KO BM cells survived longer than the mice transplanted with BCR-ABL-transduced WT BM cells. Interestingly, the frequency of c-kit+ cells within BCR-ABL+ cells was higher in the BM of the mice transplanted with BCR-ABL-transduced C/EBPβ KO cells than in the BM transplanted with BCR-ABL-transduced WT cells (Figure). When 2 × 106 BCR-ABL+ BM cells from primary recipient mice were transplanted into sublethally irradiated secondary recipients, all the mice developed a myeloproliferative status. Transplantation of 1 × 106 BCR-ABL+ BM cells from primary recipient mice, four out of the five recipient mice that transplanted with BCR-ABL-transduced C/EBPβ KO BM cells developed a myeloproliferative status and only one out of the four recipient mice that transplanted with BCR-ABL-transduced WT BM cells developed a myeloproliferative status. Transplantation of less than 0.5 × 106 BCR-ABL+ BM cells engrafted none of the secondary recipient mice. The frequencies of leukemia-initiating cells in BM of primary recipient mice that transplanted with BCR-ABL-transduced WT cells or BCR-ABL-transduced C/EBPβ KO cells were 1 in 1,404,129 and 1 in 683,773, respectively. These findings suggest that C/EBPβ promotes exhaustion of CML stem cells. In conclusion, these results suggest that C/EBPβ is upregulated by BCR-ABL at least in part through STAT5 and that C/EBPβ has the ability to exhaust CML stem cells. We are currently investigating the molecular mechanisms which protect CML stem cells from C/EBPβ-mediated exhaustion for complete eradication of CML stem cells in the future. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 4 (19) ◽  
pp. 4887-4897 ◽  
Author(s):  
Damien Luque Paz ◽  
Rébecca Jouanneau-Courville ◽  
Jérémie Riou ◽  
Jean-Christophe Ianotto ◽  
Françoise Boyer ◽  
...  

Abstract Among myeloproliferative neoplasms, polycythemia vera (PV) and essential thrombocythemia (ET) are the 2 entities associated with the most chronic disease course. Leukemic evolution occurs rarely but has a grim prognosis. The interval between diagnosis and leukemic evolution is highly variable, from a few years to &gt;20 years. We performed a molecular evaluation of 49 leukemic transformations of PV and ET by targeted next-generation sequencing. Using a hierarchical classification, we identified 3 molecular groups associated with a distinct time to leukemic transformation. Short-term transformations were mostly characterized by a complex molecular landscape and mutations in IDH1/2, RUNX1, and U2AF1 genes, whereas long-term transformations were associated with mutations in TP53, NRAS, and BCORL1 genes. Studying paired samples from chronic phase and transformation, we detected some mutations already present during the chronic phase, either with a significant allele burden (short-term transformation) or with a very low allele burden (especially TP53 mutations). However, other mutations were not detected even 1 year before leukemic transformation. Our results suggest that the leukemic transformation of PV and ET may be driven by distinct time-dependent molecular mechanisms.


Blood ◽  
2010 ◽  
Vol 116 (15) ◽  
pp. 2812-2821 ◽  
Author(s):  
Fabiana Perna ◽  
Nadia Gurvich ◽  
Ruben Hoya-Arias ◽  
Omar Abdel-Wahab ◽  
Ross L. Levine ◽  
...  

Abstract L3MBTL1, the human homolog of the Drosophila L(3)MBT polycomb group tumor suppressor gene, is located on chromosome 20q12, within the common deleted region identified in patients with 20q deletion-associated polycythemia vera, myelodysplastic syndrome, and acute myeloid leukemia. L3MBTL1 is expressed within hematopoietic CD34+ cells; thus, it may contribute to the pathogenesis of these disorders. To define its role in hematopoiesis, we knocked down L3MBTL1 expression in primary hematopoietic stem/progenitor (ie, CD34+) cells isolated from human cord blood (using short hairpin RNAs) and observed an enhanced commitment to and acceleration of erythroid differentiation. Consistent with this effect, overexpression of L3MBTL1 in primary hematopoietic CD34+ cells as well as in 20q− cell lines restricted erythroid differentiation. Furthermore, L3MBTL1 levels decrease during hemin-induced erythroid differentiation or erythropoietin exposure, suggesting a specific role for L3MBTL1 down-regulation in enforcing cell fate decisions toward the erythroid lineage. Indeed, L3MBTL1 knockdown enhanced the sensitivity of hematopoietic stem/progenitor cells to erythropoietin (Epo), with increased Epo-induced phosphorylation of STAT5, AKT, and MAPK as well as detectable phosphorylation in the absence of Epo. Our data suggest that haploinsufficiency of L3MBTL1 contributes to some (20q−) myeloproliferative neoplasms, especially polycythemia vera, by promoting erythroid differentiation.


2006 ◽  
Vol 63 (4) ◽  
pp. 364-369 ◽  
Author(s):  
Milica Strnad ◽  
Goran Brajuskovic ◽  
Natasa Strelic ◽  
Biljana Zivanovic-Todoric ◽  
Ljiljana Tukic ◽  
...  

Background/Aim. Chronic myeloid leukemia (CML) represents a malignant myeloproliferative disease developed out of pluripotent hematopoietic stem cell that contains the fusion bcr-abl gene. Disorders that occur in the process of apoptosis represent one of the possible molecular mechanisms that bring about the disease progress. The aim of our study was to carry out the analysis of the presence of the amplification of the cmyc oncogene, as well as the analysis of the changes in the expression of Bcl-2 in the patients with CML. Methods. Our study included 25 patients with CML (18 in chronic phase, 7 in blast transformation). Using an immunohistochemical alkaline phosphatase-anti-alkaline phosphatase (APAAP) method, we analyzed the expression of cell death protein in the mononuclear bone marrow cells of 25 CML patients. By a differential PCR (polymerase chain reaction) method, we followed the presence of amplified c-myc gene in mononuclear peripheral blood cells. Results. The level of the expression of Bcl-2 protein was considerably higher in the bone marrow samples of the patients undergoing blast transformation of the disease. The amplification of c-myc gene was detected in 30% of the patients in blast transformation of the disease. Conclusion. The expression of Bcl-2 protein and the amplification of c-myc gene are in correlation with the disease progression.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 193-193
Author(s):  
Yun Zhao ◽  
Allen Delaney ◽  
Afshin Raouf ◽  
Kamini Raghuram ◽  
Haiyan I Li ◽  
...  

Abstract The chronic phase of CML is sustained by rare BCR-ABL+ stem cells. These cells share many properties with normal pluripotent hematopoietic stem cells, but also differ in critical ways that alter their growth, drug responsiveness and genome stability. Understanding the molecular mechanisms underlying the biological differences between normal and CML stem cells is key to the development of more effective CML therapies. To obtain new insights into these mechanisms, we generated Long Serial Analysis of Gene Expression (SAGE) libraries from paired isolates of highly purified lin-CD34+CD45RA-CD36- CD71-CD7-CD38+ and lin-CD34+CD45RA-CD36-CD71-CD7-CD38- cells from 3 chronic phase CML patients (all with predominantly Ph+/BCR-ABL+ cells in both subsets) and from 3 control samples: a pool of 10 normal bone marrows (BMs), a single normal BM and a pool of G-CSF-mobilized blood cells from 9 donors. In vitro bioassays showed the CD34+CD38+ cells were enriched in CFCs (CML: 3–20% pure; normal: 4–19% pure) and the CD34+CD38- cells were enriched in LTC-ICs (CML: 0.2–26% pure; normal: 12–52% pure). Each of the 12 libraries was then sequenced to a depth of ~200,000 tags and tags from libraries prepared from like phenotypes were compared between genotypes using DiscoverySpace software and hierarchical clustering. 1687 (355 with clustering) and 1258 (316 with clustering) transcripts were thus identified as differentially expressed in the CML vs control CD34+CD38− and CD34+CD38+ subsets, respectively. 266 of these transcripts (11 with clustering) were differentially expressed in both subsets. The differential expression of 5 genes (GAS2, IGF2BP2, IL1R1, DUSP1 & SELL) was confirmed by real-time PCR analysis of lin-CD34+ cells isolated from an additional 5 normal BMs and 11 CMLs, and lin-CD34+CD38− cells from an additional 2 normal BMs and 2 CMLs (with dominant Ph+ cells). GAS2 and IL1R1 transcript levels were correlated with BCR-ABL transcript levels in both primitive subsets, and predicted differences in expression of IL1R1 and SELL were apparent within 3 days in CD34+ cord blood cells transduced with a lenti-BCR-ABL-IRES-GFP vs a control lenti-GFP vector (n=3). These findings support a direct role of BCR-ABL in perturbing the expression of these 3 genes. Further comparison of the meta CD34+CD38− and CD34+CD38+ CML cell libraries with most publicly accessible SAGE data revealed 69 novel tags in the CD34+ CML cells that correspond to unique but conserved genomic sequences. Nine of these were recovered by 5′- and 3′- RACE applied to cDNAs pooled from several human leukemic cell lines. These results illustrate the power of SAGE to reveal key components of the transcriptomes of rare human CML stem cell populations including transcripts of genes not previously known to exist. Continuing investigation of their biological roles in primary CML cells and primitive BCR-ABL-transduced human cells offer important strategies for delineating their potential as therapeutic targets.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 827-827
Author(s):  
Hiroto Araki ◽  
Sudhakar Baluchamy ◽  
Benjamin Petro ◽  
Mirza Saqib Baig ◽  
Montha Suhangul ◽  
...  

Abstract Abstract 827 Epigenetic modifications are considered to be important in determining the fate of hematopoietic stem cells (HSC). We previously demonstrated that the sequential addition of the chromatin-modifying agents (CMA) 5-aza-2′-deoxycytidine (5azaD) and trichostatin A (TSA) expands transplantable HSC (Araki et al. Blood 2007, Exp Hematol 2009). Others have shown that valproic acid (VPA), an HDAC inhibitor, also expands HSC (DeFelice et al. Cancer Res 2005). We thus compared the efficacy of 5azaD/TSA and VPA in promoting the ex vivo expansion of human cord blood (CB) HSC. Cells were incubated with cytokines alone (SCF, Flt3 ligand, TPO and IL-3) or with cytokines and either 5azaD/TSA or VPA, resulting in 2.2-fold, 10.7-fold or 65-fold expansion, respectively, of primitive CD34+CD90+ cells after 9 days (n=3, Cytokine alone vs. VPA p=0.004; Cytokine alone vs. 5azaD/TSA p=0.03; VPA vs. 5azaD/TSA p=0.003). Interestingly, the 10.7-fold expansion of CD34+CD90+ cells following 5azaD/TSA treatment correlated with a 10- and 10.5-fold expansion of short-term colony-forming cells (CFC) and long-term cobblestone area-forming cells (CAFC), respectively. However, the 65-fold expansion of CD34+CD90+ cells achieved with VPA treatment yielded only a 25.6- and 8.4-fold expansion of CFC and CAFC, respectively. These results suggest a marked discordance between the phenotype and function of CD34+CD90+ cells when they are expanded in VPA, but not in 5azaD/TSA. Thus, we examined the in vivo hematopoietic repopulation potential of CMA-expanded CB HSC by quantitating SCID mouse repopulating cells (SRC) using limiting dilution assays. The frequency of SRC was 1 in 22,000 in primary CB cells (n=29 mice), 1 in 123,315, in (cytokine) controls (n=16 mice), 1 in 21,720 with VPA-treatment (n=27 mice), and 1 in 3,147, in 5azaD/TSA-treated CD34+CD90+ cell cultures (n=22 mice). Unlike control, treatment with VPA prevents loss of SRC but only results in SRC maintenance, whereas 5azaD/TSA treatment leads to a 7-fold expansion of SRC. Furthermore, serial transplantation of bone marrow (BM) from primary recipients engrafted with unmanipulated CB cells resulted in engraftment in 2 of 5 secondary mice, while BM from mice engrafted with VPA-treated cells failed to display secondary engraftment (n=5 mice), whereas BM from mice engrafted with 5azaD/TSA-treated cells resulted in engraftment in 5 of 6 secondary mice. Hence, we conclude that treatment of CB CD34+ cells with 5azaD/TSA or VPA results in distinct SRC outcomes-expansion or maintenance, respectively. To dissect the molecular mechanisms that may mediate these distinct SRC fates, we examined genes implicated in HSC self-renewal including HoxB4, Bmi1, STAT3, Ezh2 and PU.1. These gene transcript levels were increased in CD34+ cells treated with either 5azaD/TSA or VPA when compared to control cultures as measured by real time quantitative PCR. In accordance with these studies, CHIP assays using antibody against acetylated histone H4 indicate increased acetylation of the promoters of HoxB4 and Bmi1 genes in both VPA- and 5azaD/TSA-treated cells. In addition, higher levels of HoxB4, Ezh2 and PU.1 proteins were observed in VPA- and 5azaD/TSA-expanded cells, compared to control cultures. Since VPA treatment does not result in SRC expansion, these observations raise questions as to the importance of the upregulation of these genes for HSC expansion. Since the pharmacologic activity of CMAs is short (hours) we hypothesize that temporal effects, including early epigenetic modifications, lead to changes in transcription factor expression, which directly or indirectly promote symmetric or asymmetric divisions ultimately resulting in expansion or maintenance of HSC. Importantly, our global microarray data (n=3) using a human genome affymetrix chip (U133 plus 2.0) revealed a set of differentially expressed genes present in 5azaD/TSA- but not in VPA-expanded CD34+ cells, thus uncovering a potential molecular signature for HSC expansion. Currently, we are examining the molecular interactions of these signature genes and the effects of silencing of these genes on HSC expansion or maintenance which should allow us to begin to unravel the molecular mechanisms involved. In summary our data indicate that treatment of HSCs with different CMAs results in distinct fates: expansion or maintenance of HSC, an observation of potential therapeutic importance. Disclosures: No relevant conflicts of interest to declare.


Haematologica ◽  
2010 ◽  
Vol 95 (12) ◽  
pp. 2153-2156 ◽  
Author(s):  
P. A. Beer ◽  
C. A. Ortmann ◽  
F. Stegelmann ◽  
P. Guglielmelli ◽  
J. T. Reilly ◽  
...  

Hematology ◽  
2005 ◽  
Vol 2005 (1) ◽  
pp. 156-160 ◽  
Author(s):  
A. Thomas Look

Abstract Clonal disorders of hematopoiesis, such as myelodysplastic syndromes (MDS) and myeloproliferative diseases (MPD), affect both hematopoietic stem cells and progenitor cells within the erythroid, platelet and granulocytic lineages and can have devastating consequences in children and adults. The genetic features of these diseases often include clonal, nonrandom chromosomal deletions (e.g., 7q–, 5q–, 20q–, 6q–, 11q– and 13q–) that appear to inactivate tumor suppressor genes required for the normal development of myeloid cells (reviewed in Bench1 and Fenaux2). These putative tumor suppressors have proved to be much more difficult to identify than oncogenes activated by chromosomal translocations, the other major class of chromosomal lesions in MDS and MPD.3 Although MDS and MPD are almost certainly caused by mutations in stem/progenitor cells,4 the role of inactivated tumor suppressor genes in this process remains poorly understood. In a small portion of myeloid diseases, mutations have been identified in genes encoding factors known to be required for normal hematopoiesis, such as PU.1, RUNX1, CTNNA1 (α-catenin) and c/EBPα, and implicating these genes as tumor suppressors.5–7 Nonetheless, the identities of most deletion-associated tumor suppressors in these diseases remains elusive, despite complete sequencing of the human genome. The deleted regions detected by cytogenetic methods are generally very large, containing many hundreds of genes, thus making it hard to locate the critical affected gene or genes. It is also unclear whether dysfunctional myelopoiesis results from haploinsufficiency, associated with the deletion of one allele, or from homozygous inactivation due to additional point mutations or microdeletions of the retained wild-type allele. In general MDS have proved surprisingly resistant to conventional treatments. Targeted therapeutic advances in MDS will likely depend on a full comprehension of underlying molecular mechanisms, in particular the tumor suppressor genes lost through clonal, nonrandom chromosomal deletions, such as the 7q– and (del)5q.


Sign in / Sign up

Export Citation Format

Share Document