A Novel t(3;17) Variant of Acute Promyelocytic Leukemia with Rearrangement of the RARA Locus.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4428-4428
Author(s):  
Robert L. Redner ◽  
Lydia C. Contis ◽  
Carol Evans ◽  
Maureen E. Sherer ◽  
Sofia Shekhter-Levin

Abstract The vast majority of patients with Acute Promyelocytic Leukemia (APL, FAB M3) have the t(15;17)(q12;q21) chromosomal translocation. This introduces the gene for PML into the retinoic acid receptor alpha (RARA) locus, which leads to expression of a PML-RARA fusion. There is convincing evidence that expression of PML-RARA underlies the APL phenotype. Yet, there have been identified rare cases of APL that do not manifest t(15;17). Many of these cases exhibit cryptic rearrangements of PML and RARA. However, in a number of cases it has clearly been shown that a fusion protein different than PML-RARA is expressed. These include the t(11;17)(q23;q21) that expresses a PLZF-RARA fusion; t(5;17)(q35;q21) that encodes NPM-RARA; t(11;17)(q13;q21) that encodes NUMA-RARA; and der(17) with duplication of 17q21.3-q23 that fuses STAT5b to RARA. We report here a novel case of APL with t(3;17) with rearrangement of RARA, but not PML. A 72 year old man presented with leukocytosis, anemia, and thrombocytopenia: wbc 20.4 X10E+9/L; hgb 10.3 g/L; PLT 22 x10E+9/L. The wbc differential showed 20% polys, 4% bands, 15% lymphocytes, 19% monocytes, 34% blasts, 1% promyelocyte, 6% myelocyte, 1% metamyelocytes. Auer rods were seen. The bone marrow was hypercellular (approximately 80%), with 88% blasts, 1.7% promyelocytes, 0.3% myelocyte, 0.3% polys, 0.3% eosinophile, 3% monocytes, 0.3% pronormoblasts, 3.7% normoblasts, and 2.3% lymphocytes. The blasts demonstrated prominent cytoplasmic granulation, Flow cytometric analysis showed the blasts to be CD117 positive, myeloperoxidase positive, CD13/33 positive, but lacking CD34 or HLA-DR expression, consistent with a diagnosis of APL. Cytogenetic studies indicated a mosaic abnormal analysis with an apparent normal cell line and one that demonstrated a 47,XY,t(3;17)(p25;q12-21), +8 karyotype. Analysis for PML-RARA expression by RT-PCR was indeterminate, owing to poor quality of the extracted RNA. Fluorescence In Situ Hybridization (FISH) was therefore performed on two hundred unstimulated cells, primarily in interphase, using the Vysis t(15;17) dual color DNA probe. 98.5% of the cells were negative for PML-RARA rearrangement (the value of 1.5% positivity is within the laboratory’s control range for false positives). To confirm that the t(3;17) rearrangement involved the RARA locus, we scored 203 unstimulated cells using the LSI RARA dual color DNA probe. 100% were positive for the RARA gene rearrangement (split signal). Four metaphase cells each showed one fused red/green signal on the normal chromosome 17, one red signal on der (17), and one green signal on the distal arm of chromsome 3. The FISH analysis therefore indicated rearrangement of the RARA, but not the PML locus. The patient expired before treatment could begin. To determine whether the t(3;17) blasts could differentiate (a hallmark of t(15;17) APL), we cultured the bone marrow cells in RPMI 1640 with 10% FCS and 10E-6 M ATRA. At 10 days 58% of the cells resembled metamyelocytes, bands, or mature polys, compared with none in the control culture. This indicates that t(3;17) retains its ability to differentiate in the presence of ATRA, consistent with its classification as a novel variant of APL.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4977-4977
Author(s):  
Mary Kay Vaske ◽  
Adeola Tomi-Olugbodi ◽  
Indumathy Varadarajan ◽  
J. Steve Hou

Abstract Introduction Five to eight percent of acute promyelocytic leukemia (APL) occurs in mid-life with disseminated intravascular coagulation (DIC) being a classic presentation. Laboratory investigation usually reveals high white cell count (WBC), promyelocytes and early myeloid series with abundant auer rods, with the exception of the microgranular variant. The bone marrow in APL is usually hypercellular with positive myeloperoxidase stains and the characteristic CD33, CD13, CD15, and CD117 positivity on immunohistochemistry and flow cytometry. The presence of a reciprocal translocation t(15;17)(q22;q12) on chromosome 17 causing a fusion of the promyelocytic - and retinoic acid receptor-alpha genes (PML-RARA) is the hallmark of APL, which is found in 95% of cases. The diagnosis of APL becomes challenging when it presents with uncharacteristic features, causing a delay in early commencement of treatment. A wide index of suspicion arises from being aware of unusual presentations. Case presentation We therefore present a 52 year old diabetic male who while been treated for hyperosmolar non ketotic hyperglycemia (HONK) was found with persistent isolated leucopenia with severe neutropenia (WBC: 0.7, absolute neutropenia: 100) and weight loss. This was initially reckoned as an adverse reaction to medication. He was treated with granulocyte colony stimulating factor (GCSF) and he responded with an unsustained increase in WBC and declining platelet counts. His medical history was insignificant for malignancy, splenomegaly, adenopathy nor sepsis. His medications comprised of antihypertensives, antidiabetics and the statins. Bone marrow findings included patchy cellularity, marked myeloid hyperplasia, few cells with auer rods, and myeloid maturation (1% blasts, 26% promyelocytes, and 22% myelocytes). The maturation and other listed bone marrow findings were initially attributed to the effect of the GCSF, but were also highly suspicious for APL. The diagnosis of APL was not given until a molecular analysis was positive for PML-RARA fusion protein. Following a diagnosis of APL with translocation, induction chemotherapy was commenced with all-trans retinoic acid (ATRA) and arsenic trioxide, during which the patient achieved molecular remission with no evidence of residual leukemia in the bone marrow (0% blasts, 1% promyelocytes, and 6% myelocytes) and a negative flow cytometric analysis. The consolidation phase is ongoing and the patient has consistently maintained remission and normal complete blood count (WBC: 1.1, absolute neutrophil count: 300). Discussion This case highlights the importance of having a high index of suspicion for APL. In such clinical settings, an early bone marrow examination is imperative and can be the first pointer to the diagnosis of APL. The prior use of GCSF created a distraction by adding an increased myelocyte population to the patient's differential. Fortunately, molecular testing for PML-RARA detected the fusion protein and treatment was then initiated. Any increased promyelocyte population, regardless of the remaining differential, should always prompt a discussion regarding the high value of ruling out APL by molecular testing. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1995 ◽  
Vol 85 (2) ◽  
pp. 495-499 ◽  
Author(s):  
L Zhao ◽  
KS Chang ◽  
EH Estey ◽  
K Hayes ◽  
AB Deisseroth ◽  
...  

Abstract The translocation between chromosomes 15 and 17, t(15;17)(q22–24;q11– 21), is present in the bone marrow cells of most patients with acute promyelocytic leukemia (APL). Although conventional cytogenetic methods are useful for diagnosing this disease, difficulties are experienced in detecting residual disease among those patients who have achieved remission. In this study, we used the fluorescence in situ hybridization (FISH) method to attempt to detect residual leukemic cells in 10 APL patients in clinical remission. The duration of remission ranged from 2 to 93 months at the time of study. Multiple bone marrow samples were analyzed by FISH in most patients. In 6 patients, no cell with t(15;17) was found. These patients remain in complete remission at present (approximately 25 to 33 months since first studied by FISH). In 4 patients, low frequencies of cells with t(15;17) were observed in at least one bone marrow sample examined. All of these patients relapsed within 1 to 14 months. No cell with t(15;17) was identified by the conventional G-banding method in any sample. The FISH results correlated well with that of a two-round nested reverse transcription polymerase chain reaction assay that was performed on the same samples. Thus, our study suggests that FISH is potentially a useful tool for detecting residual APL cells and for identifying patients at high risk of relapse.


Blood ◽  
1995 ◽  
Vol 85 (2) ◽  
pp. 495-499
Author(s):  
L Zhao ◽  
KS Chang ◽  
EH Estey ◽  
K Hayes ◽  
AB Deisseroth ◽  
...  

The translocation between chromosomes 15 and 17, t(15;17)(q22–24;q11– 21), is present in the bone marrow cells of most patients with acute promyelocytic leukemia (APL). Although conventional cytogenetic methods are useful for diagnosing this disease, difficulties are experienced in detecting residual disease among those patients who have achieved remission. In this study, we used the fluorescence in situ hybridization (FISH) method to attempt to detect residual leukemic cells in 10 APL patients in clinical remission. The duration of remission ranged from 2 to 93 months at the time of study. Multiple bone marrow samples were analyzed by FISH in most patients. In 6 patients, no cell with t(15;17) was found. These patients remain in complete remission at present (approximately 25 to 33 months since first studied by FISH). In 4 patients, low frequencies of cells with t(15;17) were observed in at least one bone marrow sample examined. All of these patients relapsed within 1 to 14 months. No cell with t(15;17) was identified by the conventional G-banding method in any sample. The FISH results correlated well with that of a two-round nested reverse transcription polymerase chain reaction assay that was performed on the same samples. Thus, our study suggests that FISH is potentially a useful tool for detecting residual APL cells and for identifying patients at high risk of relapse.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1267-1267
Author(s):  
Katharina Korf ◽  
Harald Wodrich ◽  
Alexander Haschke ◽  
Ron M. Evans ◽  
Thomas M. Sternsdorf

Abstract Although Acute Promyelocytic Leukemia (APL) has become a curable disease due to in-depth understanding of the underlying molecular processes, its investigation has provided unique and valuable insights into the processes involved in leukemogenesis. Therefore we use it as a model disease. 99% of APL-patients express a PML-RAR fusion protein. While involvement of RAR has proven indispensable for oncogenicity, the role of the PML domain is far less clear. In our previous study (Sternsdorf et al., Cancer Cell, 2006) we found that substitution of PML with heterologous self-interaction domains suffices to induce leukemias, but drastically decreases oncogenic potency of the resulting fusion proteins. In this study, we have chosen the inverse strategy: we have modified the PML domain to create a more active artificial model oncoprotein by adapting PR to its biological environment: As the typical model organism for APL studies is the mouse, we have replaced the human PML domain with the murine PML domain. This oncoprotein (mPR) creates APL-type leukemias in mice with higher penetrance and shorter latency than its human counterpart, hPR. We have used this system to study immediate early effects of expression of the model oncoprotein. While proliferating murine bone marrow cells go into senescence ex vivo, expression of mPR prevents this and robustly immortalizes murine bone marrow from every mouse strain tested so far. Senescence-associated upregulation of the cell-cycle regulators p21 and p19 was efficiently blocked by mPR expression. In mouse cells, mPR exhibits higher potency in disrupting the PML-associated Daxx/ATRX complex than hPR. Knockdown of ATRX, but not Daxx ameliorated ATRA-induced growth suppression and p21 upregulation in the human APL model cell line NB4. These data suggest, that PML-RAR promotes leukemogenesis by disrupting the Daxx/ATRX complex, which assembles at PML nuclear bodies during the onset of senescence. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 76 (9) ◽  
pp. 960-967
Author(s):  
Vesna Djordjevic ◽  
Marija Dencic-Fekete ◽  
Jelica Jovanovic ◽  
Marijana Virijevic ◽  
Nada Kraguljac-Kurtovic ◽  
...  

Introduction. The isochromosome of the long arm of derivative chromosome 17, that originates from the translocation t(15;17) [ider(17)(q10)t(15;17), or ider(17q)] in acute promyelocytic leukemia (APL), is a rare chromosome aberration associated with a poor prognosis. Case report. We report the clinical and laboratory data associated with ider(17q) for two APL patients. Cytogenetic analysis of bone marrow cells in both cases showed a mosaic karyotype with the ider(17q); reverse transcription polymerase chain reaction (RT-PCR) was positive for the long (L) isoform of the retionic acid receptor alpha (PML-RARA) fusion transcript in each patient. Fluorescence in situ hybridization (FISH) analysis with the DNA probes for the PML gene on 15q24.1, and the RARA gene on 17q21.2, confirmed the extra copy of the RARA-PML fusion gene or ider(17q). Additionally, the FISH analysis with a DNA probe for the p53 gene on 17p13.1 confirmed loss of one copy of the universal tumor suppressor p53 in both patients. Conclusion. Both reported APL patients with ider(17q) had predominance of the clone with ider(17q) compared to those with t(15;17) and/or the normal karyotype, indicating that duplication of der(17) may provide a growth advantage allowing the relevant clone to become dominant. Moreover, as an important oncogenic event and poor prognostic factor in leukemia, loss of one gene copy of the tumor suppressor p53, may also contribute to this growth advantage. Although the clinical and prognostic significance for the patients with an ider(17q) remains unclear, cytogenetic and molecular-genetic analysis should be combined to reveal more details about this complex and rare chromosomal abnormality.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4273-4273
Author(s):  
Rodrigo S. Abreu e Lima ◽  
Marcelo R. Baruffi ◽  
Ana Silvia G. Lima ◽  
Lorena L. Figueiredo ◽  
Roberto P. Falcao ◽  
...  

Abstract Acute promyelocytic leukemia (APL) associated with the t(15;17)/PML/RARα and AML with t(8;21)/AML1/ETO are AML subtypes characterized by distinct cytomorphological and clinical features. The coexistence of both genetic abnormalities in a single leukemic clone is extremely rare, and whether it affects PML subcellular distribution or the response to the treatment with all trans retinoic acid (ATRA) has not been previously analyzed. Here we report an AML case refractory to treatment in which the PML/RARα or AML1/ETO expression was analyzed by conventional cytogenetics, FISH, RT-PCR, Southern blot and Spectral Karyotyping (SKY), in addition PML distribution was analyzed by immunofluorescence staining. A 47-year-old female patient complaining of dyspnea for 1 month and presenting hematomas and pallor was referred to the University Hospital of Ribeirao Preto. Adenopathy and splenomegaly were absent. The hemoglobin level was 6g/dl, platelet count was 14x103/ml and leukocyte count was 10.2x103/ml with 22% blasts. The coagulation profile was normal, except for the D-dimers level wich was between 4,000 – 8,000 ng/ml. The differential counts of bone marrow (B.M.) aspirates revealed the presence of 21% blasts and 24% promyelocytes. Immunofluorescence staining of B.M. cytospin preparations using the PG-M3 antibody revealed that PML was delocalizated from the nuclear bodies, a feature suggestive of the diagnosis of APL. The immunophenotypic analysis identified two cell subsets: one CD33+ CD13+ HLA-DR− CD34− CD15+ with high Sideward Scatter (SSC) and another one CD33+ CD13+ HLA-DR+ CD34+ CD15− with low SSC values. The karyotype after G-banding was as follows: 46X, iso(X)(q11), t(8;21)(q22;q12) in 18/18 metaphases.SKY analysis confirmed the chromosomal abnormalities detected by G-banding and identified a cryptic insertion of chromosome 15 material into chromosome 17 in 5/5 metaphases.The expression of AML1/ETO and PML/RARα genes was demonstrated by RT-PCR. FISH analysis were performed using Vysis PML and RARα probes did not detect PML/RARα rearrangements in 300 interphases. On the contrary, FISH assays using Vysis AML1and ETO probes confirmed the presence of t(8;21) in 15% of 300 interphases. Finally, RARα rearrangement was detected by Southern blot analysis performed on B.M. cells genomic DNA using the H18 and K3 RARα genomic probes.The patient was treated with ATRA 45 mg/m2/d for 30 days associated with standard 3+7 AML induction regimen but did not achieve remission. ATRA dose was increased to 90 mg/m2/d and a second identical course of chemotherapy was administered from Day +35. B.M. aspirate obtained on Day +63 presented 2% of blasts/promyelocytes, but the PML/RARαand AML1/ETO transcripts were still detectable by RT-PCR. The patient died of sepsis on Day +67. The lack of response to ATRA observed in this patient contrasts with the favorable outcome observed in the majority of APL patients.Since both PML/RARα and AML1/ETO oncoproteins affect transcription by forming repressor complexes containing histone deacetylase, it is formally possible that their coexpression could lead to irreversible chromatin remodeling. Despite its rarity, the present case is informative because it suggests that PML/RARα and AML1/ETO may synergize and thus render the leukemic cells resistant to treatment.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3741-3741
Author(s):  
Saravanan Ganesan ◽  
Ansu Abu Alex ◽  
Ezhilarasi Chendamarai ◽  
Nithya Balasundaram ◽  
Hamenth Kumar Palani ◽  
...  

Abstract Degradation of PML-RARA upon treatment with ATO is predominantly mediated by the proteasome complex. Reports suggest that in relapsed APL patients who were treated upfront with ATO, mutations in the B2 domain of PML in PML-RARA gene are involved in resistance to ATO. These mutations predict a poor clinical outcome in spite of subsequent combination of ATO with chemotherapy (NEJM 2014). We had previously reported that Bortezomib (Bo) was able to synergize with ATO by inducing apoptosis through increased levels of ROS and up regulation of the UPR pathway (Blood. 2012;120, 3552) and we had also noted PML-RARA degradation and nuclear body formation with this combination. We further evaluated the mechanism of degradation of PML-RARA when ATO was combined with Bo and the effect of this combination on resistant cell lines, a mouse model and in relapsed patients. We generated in-house ATO resistant NB4 cell lines (NB4EV-ASR1, ASR2 and ASR3). In NB4EV-ASR1 we confirmed the presence of A216V mutation in the PML B2 domain (previously reported to be involved in ATO resistance) while the other 2 clones did not have this or any other mutation in PML-RARA. We noted an increase in the baseline proteasomal activity in all the resistant cell lines when compared to naïve NB4 cells (n=3; data not shown). The combination of ATO and Bo induced a significant apoptosis in all the resistant cells similar to naïve NB4 cells (Figure 1A: n=4; Combination Index = 0.02).The mechanism of inducing apoptosis in the resistant cell lines was similar to naïve NB4 cells, as previously reported by us, and involved an increased level of ROS, decreased mitochondrial membrane potential, induction of UPR and activation of caspase-3 (Figure 1B). We next evaluated PML-RARA degradation in NB4 naive cells treated with a combination of ATO+Bo. At 24 hours, there was an evidence of induction in autophagy as shown by LC3II formation using western blot technique which increased at 48 hours; this time point coincides with time at which maximum PML-RARA degradation occurred (Figure1C). Similar results were seen in the resistant cell lines (with and without mutation A216V). Blocking autophagy by 3-methyl adenine showed a partial inhibition in the degradation of PML-RARA. We have also observed that there is an accumulation of p62 (ubiquitin binding protein) at 24 hours and this was degraded by 48 hours suggests that accumulated ubiquitinated products were cleared by autophagy via p62 (Figure1D). In a co-immunoprecipitation experiment, p62 and LC3II proteins precipitated along with PML-RARA (figure 1E). Knock down of p62 transcript by siRNA followed by ATO+Bo treatment showed an accumulation of PML-RARA in the treated cells in comparison to the scrambled and control. In an APL transplanted mice model, combination of ATO and Bo prolonged the life span of the mice as illustrated in Figure1F. In this group there was a significant decrease in the leukemia burden evidenced by decreased leukemic cells in bone marrow, peripheral blood and spleen by flow cytometry, RQ-PCR and decreased spleen size on day +20. A reduction in the LIC was demonstrated by secondary transplants. We also observed that transplantation of bone marrow cells from the long term surviving mice post ATO+Bo therapy did not induce leukemia (Figure1G) and no transcripts of PML-RARA were detected in the recipients. A phase II clinical study combining Bo with ATO and chemotherapy has been initiated for patients with relapsed APL (NCT01950611). In this ongoing study 11 patients have been enrolled. The median age was 32 years. 7 were males. All patients achieved hematological remission and the median time to complete molecular remission was 42 days (one patient still on induction therapy). The addition of Bo was well tolerated. None of the cases had evidence of significant neuropathy, worsening of coagulopathy, IC bleed or a differentiation syndrome. Long term follow up is awaited to comment on the efficacy of this combination. In conclusion, the mechanism of ATO+Bo synergy is multi-factorial and appears to be predominantly due to increase in ROS activity and up regulation of UPR pathway leading to apoptosis. In spite of proteasomal inhibition by addition of Bo with ATO, PML-RARA continues to be degraded and this is mediated by up-regulation of autophagy pathway. ATO+Bo synergy was further confirmed in a pre-clinical model. This combination is also effective in ATO resistant cell lines with high levels of synergism. Figure 1 Figure 1. Disclosures Off Label Use: Bortezomib in the treatment of acute promyelocytic leukemia.


Blood ◽  
1991 ◽  
Vol 78 (6) ◽  
pp. 1413-1419 ◽  
Author(s):  
ZX Chen ◽  
YQ Xue ◽  
R Zhang ◽  
RF Tao ◽  
XM Xia ◽  
...  

Fifty patients with acute promyelocytic leukemia (APL) have been treated with all-trans retinoic acid (RA). In vitro induced differentiation of primarily cultured bone marrow cells from the patients, colony-forming unit granulocyte-macrophage (CFU-GM) and L-CFU colony-forming assays, and karyotype analysis were performed over the treatment course. The very high bone marrow complete remission (CR) rate (94%) suggested that all-trans RA was superior to conventional chemotherapeutic regimens for the treatment of APL. The leukemic clone was reduced by RA-induced terminal differentiation and loss of proliferation capacity of leukemic cells. Relapse after CR in about 40% of patients was the major reason for the failure of the RA treatment. Patients who relapsed after a chemotherapy-maintained CR could be effectively reinduced to second CR by RA. However, if relapse occurred after a CR maintained by both RA and chemotherapy, the sensitivity of newly emerged leukemic clones to RA was greatly reduced. Therefore, it is suggested that RA should be replaced by conventional chemotherapy as soon as CR is achieved. Laboratory studies proved valuable in selecting cases for RA therapy and in predicting therapeutic effects and prognosis.


Blood ◽  
1991 ◽  
Vol 78 (6) ◽  
pp. 1413-1419 ◽  
Author(s):  
ZX Chen ◽  
YQ Xue ◽  
R Zhang ◽  
RF Tao ◽  
XM Xia ◽  
...  

Abstract Fifty patients with acute promyelocytic leukemia (APL) have been treated with all-trans retinoic acid (RA). In vitro induced differentiation of primarily cultured bone marrow cells from the patients, colony-forming unit granulocyte-macrophage (CFU-GM) and L-CFU colony-forming assays, and karyotype analysis were performed over the treatment course. The very high bone marrow complete remission (CR) rate (94%) suggested that all-trans RA was superior to conventional chemotherapeutic regimens for the treatment of APL. The leukemic clone was reduced by RA-induced terminal differentiation and loss of proliferation capacity of leukemic cells. Relapse after CR in about 40% of patients was the major reason for the failure of the RA treatment. Patients who relapsed after a chemotherapy-maintained CR could be effectively reinduced to second CR by RA. However, if relapse occurred after a CR maintained by both RA and chemotherapy, the sensitivity of newly emerged leukemic clones to RA was greatly reduced. Therefore, it is suggested that RA should be replaced by conventional chemotherapy as soon as CR is achieved. Laboratory studies proved valuable in selecting cases for RA therapy and in predicting therapeutic effects and prognosis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3166-3166
Author(s):  
Mariam Thomas ◽  
Mahadeo A. Sukhai ◽  
Yali Xuan ◽  
Soheila A. Hamadanizadeh ◽  
Rashmi S. Goswami ◽  
...  

Abstract Acute promyelocytic leukemia (APL) is characterized by accumulation of abnormal promyelocytes in the bone marrow and peripheral blood, and sensitivity to treatment with all-trans retinoic acid. APL cases have a balanced chromosomal translocation involving retinoic acid receptor alpha (RARA) on chromosome 17. The resulting fusion proteins (X-RARA) are aberrant transcription factors and block ATRA-induced neutrophil differentiation. Loss of RARA signalling impairs granulopoiesis, but is not sufficient to cause a leukemic phenotype. We elucidated the identities of additional signalling pathways, which can potentially cooperate with X-RARA in APL, that are commonly modulated by multiple X-RARA. We used the U-937 hematopoetic cell line retrovirally transduced with NPM-RARA (Kamel-Reid et al, 2003), and NuMA-RARA, in addition to NB4 cells (expressing PML-RARA) to determine the common genes and pathways deregulated in APL. Gene expression analysis was carried out on RNA harvested in triplicate from control and X-RARA expressing cell lines, using the Affymetrix U133Plus2 array platform. Gene expression and pathways analysis of array data was carried out using a suite of analysis tools. Array data were validated in an independent sample set by real-time quantitative PCR. We observed a total of 311 genes deregulated at least 2-fold by NuMA-RARA (192 up-regulated, 119 down-regulated), 393 genes deregulated by NPM-RARA (292 up-regulated, 101 down-regulated), and 2056 genes deregulated by PML-RARA (1097 up-regulated, 959 down-regulated). A total of 65 genes, in 5 major interaction networks, were commonly deregulated by all three X-RARA (42/65 up-regulated, 23/65 down-regulated). The majority of these genes are involved in cellular signalling (14 genes, p-value 1.57E-07–7.77E-3), transcription (13 genes, p-value 5.68E-7–3.91E-3), cell proliferation (25 genes, p-value 9.73E-7–7.77E-3), apoptosis (26 genes, p-value 9.96E-7–7.71E-3), and cell movement (17 genes, p-value 1.43E-6–7.60E-3). Genes involved in the CEBPA interaction network (GFI1, TRIB2, ELA2), as well as other genes that we anticipated to be deregulated in APL including ID1, MMP9, and JUN were found through this analysis. NF-kB (p-value 2.10E-3), AHR (p-value 2.12E-3), IL-6 (p-value 5.40E-3), and G-protein coupled receptor (p-value 7.45E-3) signalling were among the top canonical pathways determined to be altered by X-RARA. Over-expression of a number of NF-kB downstream transcriptional targets, including VEGF, IL8, MMP9, cIAP2, and TNFAIP3, were also observed in multiple X-RARA expressing cell lines. In addition, in vitro results were compared to NuMA-RARA gene targets identified in primary bone marrow cultures derived from the hCG-NuMA-RARA transgenic mouse model (Sukhai et al, 2004). We observed that pathways involved in cell signalling, cell death, gene expression, proliferation, and cell cycle were significantly deregulated in both mouse and human datasets, indicating that these pathways may be important cooperating events in APL. Our data represent the first comparison of the genetic profiles of the variant fusion proteins NPM-RARA and NuMA-RARA in a haematopoietic cell system. Our studies are a significant step in identifying key targets that cooperate with X-RARA in the development of APL.


Sign in / Sign up

Export Citation Format

Share Document