Skip to main content

RETRACTED ARTICLE: Downregulated Poly-C binding protein-1 is a novel predictor associated with poor prognosis in Acute Myeloid Leukemia

This article was retracted on 12 September 2018

This article has been updated

Abstract

Background

Depletion of Poly-C binding protein-1(PCBP1) is implicated in various human malignancies. However, the underlying biological effect of PCBP1 in cancers, including acute myeloid leukemia (AML), still remains elusive. The purpose of this study was to examine the expression and clinical outcome of PCBP1in acute myeloid leukemia.

Methods

Bone marrow fluids of 88 newly diagnosed AML patients were sampled, and the PCBP1 mRNA expression level was evaluated using quantitative RT-PCR. The association between PCBP1 expression and clinicopathological features or the survival status of the patients was assessed by Chi-square test and Kaplan-Meier method.

Results

Comparing newly diagnosed AML patients to normal healthy donors, PCBP1 expression was significantly decreased in AML patients (Pā€‰<ā€‰0.001). Conversely, PCBP1 expression had accordingly recovered back to normal in patients with complete remission (Pā€‰<ā€‰0.001). Clinical feature analyses showed that PCBP1 expression was negatively correlated with white blood cell count (Pā€‰=ā€‰0.024). In addition, patients with low PCBP1 expression had poor disease-free survival (11.8Ā % vs. 45.3Ā %; Pā€‰=ā€‰0.01) and overall survival (18.2Ā % vs. 42.4Ā %; Pā€‰=ā€‰0.032), respectively.

Conclusions

Taken together, our results showed for the first time that expression of PCBP1 was down-regulated in newly diagnosed AML patients and might be an independent prognostic marker in AML and should to be further investigated.

Background

Acute myeloid leukemia (AML) is a clinically and genetically heterogeneous disease characterized by the differentiation arrest and the malignant clonal proliferation of hematopoietic progenitor cells [1, 2]. It is the most common myeloid leukemia and accounts for 70Ā % of acute leukemia in adults [3ā€“5]. Exposure to ionizing radiation, benzene, and cytotoxic chemotherapy is known to increase the incidence of AML [3]. The pathogenesis of AML is highly complex. Over the past decades, researching in genetic alterations, such as FLT3, NPM1, CEBPA gene mutations, resulted in prodigious improvement in the pathogenesis, prognostic classification and novel therapeutic approaches of AML [6, 7]. Despite the noteworthy progress in the development of genomic studies on AML, the prognosis classification and treatment for most AML patients still remains unsatisfactory [7ā€“10]. More than half of young adult patients and about 90Ā % of elder patients still die of the disease, due to either resistance to therapy or relapsed AML, and the 5-year survival rate is less than 30Ā % [11ā€“13]. As a result, more effective gene signatures are still required for risk-adapted treatment [14]. In addition, clinical outcomes can differ greatly in patients with identical karyotypes, suggesting that additional factors on AML diagnosis and prognosis should be identified and clarified [12]. Thus, investigation for more valuable diagnostic, prognostic factors and therapeutic targets implicated in AML pathogenesis is pressing for the performance of individual antitumor treatment.

Poly-C binding protein 1, also referred to as hnRNP-E1 or Ī±CP-1 [15], is broadly expressed in many human tissues and organs, including bone marrow, as well as the liver, heart, kidneys, brain, lungs, and placenta. Functionally, PCBP1 is known to play multiple roles in a wide spectrum of transcriptional and posttranscriptional events, including in mRNA transcription regulation, transportation, splicing, translation, and stabilization by means of its cis-DNA/RNA-binding activity [16ā€“18]. Increasing clinical and cellular evidence has demonstrated that PCBP1 plays pivotal roles in multiple pathological processes, including cancer apoptosis, metastasis, invasion and malignant transformation [17, 19ā€“21]. Recently, some studies indicated that aberrant expression of PCBP1 is associated with poor prognosis [22], and low PCBP1 expression could be a poor prognosis marker in diverse human cancers [17, 23].

Although aberrant PCBP1 expression levels are associated with cancer progression and metastases in many human solid cancers, there is not a straightforward experiment to demonstrate this role in tumor formation. Likewise, the function of PCBP1 in leukemia, including AML remains elusive. Herein, our study was carried out to investigate PCBP1 expression in AML and to understand the prognostic role of PCBP1 in AML patients.

Methods

Patients and tissue samples

A myeloid neoplasm with 20Ā % or more myeloblasts in the peripheral blood or bone marrow is considered to be AML according to the 2008 World Health Organization (WHO) criteria [24]. Initial diagnosis of AML samples prior to AML treatment were collected as newly diagnosed of acute myeloid leukemia. 88 patients, who were newly diagnosed with AML, were admitted for treatment at the First Affiliated Hospital of Sun Yat-sen University from 2012 to 2015. All patients underwent a bone marrow puncture at the time of diagnosis, with a signed informed consent. Morphology, Immunology, Cytogenetics and Molecular Biology (MICM) classification was performed on the bone marrow. Clinical features of all patients were summarized in TableĀ 1, and the risk status was predicted on the basis of validated cytogenetics and molecular abnormalities. According to NCCN guidelines version 2.2013, we classified acute myeloid leukemia risk-status into three categories: better-risk (inv(16),t(16;16),t(8;21),t(15;17) or NPM1 mutation in the absence of FLT3-ITD with normal cytogenetics), poor-risk (complex monosomal karyotype, āˆ’5, 5q-,-7,7q-,11q23 abnormality, inv(3), t(3;3), t(6;9), t(9;22) or normal cytogenetics with FLT3-ITD mutation) or intermediate-risk (neither better-risk nor poor-risk). Meanwhile, 24 healthy donors were recruited as normal control with their permission. Complete remission (CR) was defined by the presence of all the following: less than 5Ā % myeloid blasts in bone marrow, no leukemic blasts in peripheral blood, absolute neutrophil countsā€‰ā‰„ā€‰1,500/Ī¼l and platelet countsā€‰ā‰„ā€‰100,000/Ī¼l in peripheral blood, and no persistence of extramedullary disease [25]. The clinical characteristics of these newly diagnosed patients were listed in TableĀ 1. Sixty-seven patients were further followed up. In analysis of the disease-free survival, data were censored at the time of relapse, death or the last contact; in analysis of the overall survival, data were censored when the patients died or last known to be alive. This study was approved by the local ethic committee of the First Affiliated Hospital of Sun Yat-sen University and informed consents were obtained from each patient.

Table 1 Characteristics of AML patients in this study

Preparation of mononuclear cells and RNA extraction

Bone marrow fluid from each patient was obtained using a 6-ml EDTA-containing vacutainer. The collected sample was then diluted with physiological saline in 1:1 proportion, the diluted sample was carefully layered over the ficoll lymphocyte separation medium (LSM, MP Biomedicals, USA) and centrifuged at a speed of 400ā€‰Ć—ā€‰g at room temperature for 20Ā minutes. During centrifugation, differential migration results in the formation of four cell layers: plasma layer, mononuclear cell layer, LSM layer and RBC pellet. Erythrocytes and polynuclear leukocytes sedimented to the bottom of the tube; mononuclear cells were found at the plasma-LSM interface. After aspirating the mononuclear cells and transferring them to a new centrifuge tube, cells were washed twice with buffered balanced salt solution and re-suspended in 1Ā ml TRIzol reagent (Cwbio Cat: CW0580A). Total cellular RNAs were then extracted according to the guideline of the Ultrapure RNA Kit (Cwbio Cat: CW0581, China) and eluted in 30ā€“50Ā Ī¼l of RNase-free water. The RNA concentration and quality were determined by NanoDrop 1000 spectrophotometer (ThermoFisher Scientific, USA).

Reverse transcription

To obtain cDNA, 2Ā Ī¼g of total RNA were diluted into nuclease-free water to a total volume of 12Ā Ī¼l, mixed with 1Ā Ī¼l of anchored-oligo (dT) 18 primer (2.5Ā Ī¼M) and incubated for 10Ā minutes at 65Ā Ā°C to denature the template-primer mixture. After that, the remaining components of the RT mix, 4Ā Ī¼l transcriptor RT reaction buffer (5Ɨ), 0.5Ā Ī¼l protector RNase inhibitor (40 U/ul), 2Ā Ī¼l deoxynucleotide Mix (10Ā mM) and 0.5Ā Ī¼l transcriptor reverse transcriptase (20 U/Ī¼l) were added to the tube. The 20Ā Ī¼l reaction volumes were incubated at 55Ā Ā°C for 30Ā minutes followed by 5Ā minutes at 85Ā Ā°C, and the reaction was then halted by placing the tube on ice. All cDNAs were diluted to 1:10 in nuclease-free water and stored at -20Ā Ā°C for use.

Quantitative real-time PCR analysis for PCBP1 expression

In order to quantify PCBP1 mRNA expression levels, primers 5ā€™-CATCCGCTAAGAATTTAAAAAT-3ā€™ (forward) and 5ā€™-AAGACAGCAATTCCCAGC-3ā€™ (reverse) were used. Meanwhile 5ā€™-ACCACAGTCCATGCCATCAC-3ā€™ (forward) and 5ā€™-TCCACCACCCTGTTGCTGTA-3ā€™ (reverse) were used for amplification of the internal control gene GAPDH. Quantitative real-time PCR (qRT-PCR) were performed according to the instructions of RealMasterMix (SYBR Green) kit (Tiangen Biotech, China). Briefly, in each total volume of 20Ā Ī¼l reaction mixture, 9Ā Ī¼l of 2.5ā€‰Ć—ā€‰RealMasterMix/20ā€‰Ć—ā€‰SYBR Solution, 0.4Ā Ī¼l of forward and reverse primers(10Ā Ī¼M), 2Ā Ī¼l of diluted cDNA and 8.2Ā Ī¼l of ultrapure water were added. Amplification was conducted on a CFX96 Real-time fluorescent quantitative PCR detection system (Bio-Rad, USA) with the thermal cycling parameters as the initial denaturation at 95Ā Ā°C for 2Ā minutes, following by 40Ā cycles of 20Ā seconds at 95Ā Ā°C, 30Ā seconds at 60Ā Ā°C, 40Ā seconds at 68Ā Ā°C. Melting curves were drawn to identify the specificity of the PCR primers. Each experiment was repeated at least for three times.

Relative quantification of PCBP1 expression was calculated by the comparative cycle threshold method (the 2-Ī”Ī”CT method) [26], where Ī”Ī”CTā€‰=ā€‰Ī”CT,Target - Ī”CT,Control. The change in expression of PCBP1 was normalized to its internal control GAPDH, just as Ī”CT,Targetā€‰=ā€‰CT,Target - CT,GAPDH. Meanwhile, the smallest numeric values of 2-Ī”CT, Target was defined as ā€œ1ā€ as a control; the relative PCBP1 mRNA expression of other AML sample, CR patients and healthy donors were carried out by folds of the control.

Statistical analysis

Statistical analysis was performed by SPSS12.0 statistical software. The data was expressed as the meanā€‰Ā±ā€‰standard deviation (SD) or meanā€‰Ā±ā€‰SEM of at least 3 independent experiments. The Student T test was used to compare continuous variables. Different frequencies between groups were compared by the chi-square test or the Fisher exact test. Survival curves were obtained by the Kaplan-Meier method, and differences in survival among groups were analyzed by the log-rank test. P valuesā€‰<ā€‰0.05 were considered statistically significant.

Results

PCBP1 was significantly down-regulated in newly diagnosed AML patients

In order to explore whether PCBP1 is aberrantly expressed in AML patients, total RNA was isolated from bone marrows of AML patients and healthy donors, and the relative expression level of PCBP1 was detected using quantitative RT-PCR (qRT-PCR). The averaged relative value of PCBP1expression in newly diagnosed AML patients (averaged expression value 7.08, nā€‰=ā€‰88) was significantly lower than that in normal controls (averaged expression value 19.31, nā€‰=ā€‰24) (Fig.Ā 1, Pā€‰<ā€‰0.001). Furthermore, among 12 patients who achieved CR after one or two cycles of chemotherapy, PCBP1 expression level had markedly recovered back to their initial admission level (mean, 20.96 versus 6.25) (Fig.Ā 2, Pā€‰<ā€‰0.001), although it was higher than that of the normal controls, there was no statistical difference (mean 20.96 versus 19.31, pā€‰>ā€‰0.05) (Fig.Ā 3). Therefore, the results revealed that PCBP1 expression level in the majority of AML patients was negatively related to the state of the illness of AML.

Fig. 1
figure 1

PCBP1 was down-regulated in AML patients.The relative PCBP1 mRNA expression in newly diagnosed AML patients (nā€‰=ā€‰88) and healthy donors (nā€‰=ā€‰24) by qRT-PCR evaluation. PCBP1 expression was significantly decreased in newly diagnosed AML patients (average 7.08) compared with healthy donors (average 19.31; Pā€‰<ā€‰0.001). Each experiment was performed at least in triplicate

Fig. 2
figure 2

Comparison of PCBP1mRNA expression in 12 newly diagnosed/complete remission paired samples. Comparison of PCBP1 mRNA expression of patients in complete remission (CR) with their initially diagnosed counterparts.PCBP1 expression in patients achieved CR is 20.96ā€‰Ā±ā€‰3.22 versus 6.25ā€‰Ā±ā€‰1.72; paired t test, Pā€‰<ā€‰0.001

Fig. 3
figure 3

Comparison of PCBP1 expression level in CR patients with normal controls.Comparison of PCBP1 expression in CR patients with that in healthy donors.CR, 20.96ā€‰Ā±ā€‰3.22 versus donors, 19.31ā€‰Ā±ā€‰3.3, Pā€‰>ā€‰0.05

Low PCBP1 expression was associated with advanced AML clinical characteristics

To question the relationship of PCBP1 expression to AML characteristics, the 88 AML patients were classified into two groups according to the averaged PCBP1 expression, in which patients with PCBP1 expression value less than their averaged level (7.08) were classified into low-expression group (mean expression value 3.12, nā€‰=ā€‰63), whereas patients with equal PCBP1 level or above the mean value were assigned to the high-expression group (mean expression value 17.06, nā€‰=ā€‰25), of them, 71.6Ā % (63/88) patients showed low PCBP1 expression. PCBP1 expression and clinical features were summarized in TableĀ 2. The white blood cell count greater than or equal to 50ā€‰Ć—ā€‰109/L was identified in 30 (34.1Ā %) of 88 patients with AML and the frequency of PCBP1 underexpression was 86.7Ā % (26/30) in this subtype, whereas in 58 (65.9Ā %) AML patients with leukocyte count less than 50ā€‰Ć—ā€‰109/L, only 63.8Ā % (37/58) cases showed low-pcbp1 expression. Univariant analysis demonstrated that low PCBP1 expression was individually associated with high white blood cell count (Pā€‰=ā€‰0.024). However, there was no significant correlation of PCBP1 expression with other clinical risk-predicting factors including FAB category (Pā€‰=ā€‰0.413), risk status (Pā€‰=ā€‰0.143), CD7 expression (Pā€‰=ā€‰0.343), and CD56 expression (Pā€‰=ā€‰0.563) (Shown in TableĀ 2), indicating PCBP1 could be an independent prognostic marker.

Table 2 Association of PCBP1 expression to clinical features

Association of PCBP1 expression to survival of AML patients

To further investigate the prognostic value of PCBP1 expression in patients with AML, sixty-seven CR patients with non-M3 acute myeloid leukemia were followed up. The effect of PCBP1 expression on non-M3 AML patientsā€™ survival time was assessed by the Kaplan-Meier method with the log-rank test. Results revealed that the low-PCBP1 expression group had a clearly poor disease-free survival (11.8Ā % vs. 45.3Ā %; log rank Pā€‰=ā€‰0.01; shown in Fig.Ā 4a) and overall survival (18.2Ā % vs. 42.4Ā %; log rank Pā€‰=ā€‰0.032; shown in Fig.Ā 4b), confirming that PCBP1 could be an additional predictor in AML prognosis.

Fig. 4
figure 4

Kaplan-Meier curves of DFS and OS. The Kaplan-Meier survival analysis of the DFS (a) and OS (b) in 67 newly diagnosed non-M3 AML patients with CR. High PCBP1 group, nā€‰=ā€‰23; Low PCBP1 group, nā€‰=ā€‰44, pā€‰<ā€‰0.05

Discussion

Poly(C)-binding proteins (PCBPs), which are also well known as the heterogeneous nuclear ribonucleoprotein (hnRNP) family, were initially defined by their sequence-specific binding ability to polycytosine [poly (C)] in protein-nucleic acids interaction. In recent years, accumulating evidence has shown that PCBPs play a critical role in tumor development and progression [27,28]. Among PCBPs, hnRNP K, PCBP1, and PCBP2 have been intensively studied. HnRNP K, which is overexpressed in diverse neoplasms [29], can activate the transcriptional activation of oncogenes c-SRC and c-Myc [30,31], hinting its cancer-promoting potential. Upregulation of PCBP2 was also shown in the higher grades of human glioma [32] and PCBP2 can facilitate gastric cancer growth by targeting miR-34a [33]. More and more observations have described that PCBP1 could be implicated in malignancies [16, 23, 31] via in vitro cell line analyses and clinical observations. Mechanistically, PCBP1 silence facilitated androgen receptor (AR) protein expression by binding to the 3ā€™ untranslated region (UTR) of AR mRNA in prostate cancer LNCap cells [34]. The phosphorylated PCBP1 mediated by TGF Ī² signaling could promote epithelial-mesenchymal transition [35]. Accordingly, silencing endogenous PCBP1 in human SH-SY5Y cells resulted in a large body of altered transcripts, of which some are involved in Wnt signaling and TGF beta signaling [36]. NF-Kappa B pathway is also implicated in cancer metastasis, relapse and drug resistance, and PCBP1 is shown to inhibit NF-Kappa B activity by interacting with STAT3 pathway in HEK293 cells [37]. Furthermore, the other advances on the biological function and molecular mechanisms of PCBP1 involving in gene regulation were graphically represented in Fig.Ā 5 [38]. Recent data disclosed that PCBP1 modulated P63 expression via mRNA stability [39]. Our previous results also demonstrated that PCBP1 downregulates metastatic PRL-3 phosphatase translation by interacting with its 5ā€™ UTR [17].

Fig. 5
figure 5

Schematic representation of signaling network involving PCBP1. Adapted from Ref. [38]

It has also been demonstrated that aberrant expression of PCBP1 promotes tumor development and progression [19]. Recent studies have revealed PCBP1 could increase the expression of P21WAF1 to reduce cell proliferation and tumor growth and play as a tumor suppressor in human solid cancer [17, 40]. In addition, our previous study had clearly demonstrated that PCBP1 repressed metastatic PRL-3 translation, resulting in the decreased tumorigenic ability of tumor cells [17]. Interestingly, PRL-3 has been reported to be upregulated in de noveo FLT3-ITD-negative AML patients [41] and involved in AML leukemogenesis [41] [42], indicating PCBP1 as a tumorsuppressor gene in AML progression. Our data is comparable with above reports and found that PCBP1 expression in newly diagnosed AML is obviously lower than that observed in healthy control donors. The results are also well consistent with observations that PCBP1 expressions are usually downregulated in some human tumors, including liver [19], gall bladder [23], choriocarcinoma [22], colon and lung cancers [17], as well as in metastatic cervical cancer [21] and metastatic breast cancer cells [43].Then, we found that the expression of PCBP1 in 12 AML patients in complete remission after intensive chemotherapy was also markedly increased, compared to that at their initial diagnosis, suggesting the dysregulated expression of PCBP1 is consistent with cancer progression. Similar results are also reported in other studies, such as Pillai MR et al. reported that reduced PCBP1 expressed in higher grade squamous intraepithelial lesions of cervical tissues and even progressed to cervical cancer [21].In addition, we analyzed the clinicopathological factors of PCBP1 in AML. Our results showed that low level of PCBP1 expression is positively correlated with the high counts of white cells, which represented more aggressive clinical features. The difference in other aggressive clinical features, including age, risk status and leukemic blasts, was not significant. Moreover, when compared to the patients with high PCBP1 expression, low PCBP1 expression AML patients have poorer overall and disease-free survival rates, indicating that PCBP1 expression can be considered as a novel prognostic marker in initial diagnosis of AML. Given that PCBP1 is involved in gene regulation through multiple roles, including transcription, translation and mRNA stabilization. PCBP1 should possibly participate in the blockade of tumorigenesis by either inhibiting particular oncogene expression or enhancing tumor suppressor expression in AML progression. Whatever, the detailed underlying mechanism of PCBP1 in AML progression will be further investigated and a potent novel target could be dug out in the PCBP1-silencing tumors.

To the best of our knowledge, the current study is the first to demonstrate that PCBP1 is downregulated in AML patients and accounts for approximately 72Ā % of newly diagnosed AML patients compared to healthy donors. In this study, we screened PCBP1 expression and defined the average expression level of AML patients as a cutoff point to classify the patients into 2 groups, of them, 71.6Ā % (63/88) patients showed low PCBP1 expression. Similar work for confirming this novel gene was carried out in lung and colon carcinoma [17]. In that study, IHC analysis revealed that PCBP1protein expression is negative in 85.7Ā % (54/63) lung cancer tissue arrays and 91.7Ā % (66/72) colon cancer tissue arrays and the different expression level may be due to different cancer types. Furthermore, the real risk changes of PCBP1 downregulation in AML need to be further investigated.

Conclusion

This study showed that PCBP1 was downregulated in newly diagnosed acute myeloid leukemia, and the decreased PCBP1 expression level was associated with adverse prognosis in AML. Our findings are well consistent with observations that PCBP1 expression was usually downregulated in a variety of solid tumors. However, the underlying molecular mechanism for PCBP1 in AML progression is not well elucidated and further investigation will be required.

Change history

  • 12 September 2018

    This article [1] is retracted at request of the Editor.

Abbreviations

AML:

Acute myeloid leukemia

PCBP1:

Poly-C binding protein-1

CR:

Complete remission

DFS:

Disease-free survival

OS:

Overall survival

HnRNPs:

Heterogeneous nuclear ribonucleoproteins

PCBPs:

Poly(C)-binding proteins

References

  1. O'Donnell MR, Abboud CN, Altman J, Appelbaum FR, Coutre SE, Damon LE, et al. Acute myeloid leukemia. J Natl Compr Canc Netw. 2011;9(3):280ā€“317.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  2. Conway O'Brien E, Prideaux S, Chevassut T. The epigenetic landscape of acute myeloid leukemia. Adv Hematol. 2014;2014:103175.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  3. Estey E, Dohner H. Acute myeloid leukaemia. Lancet. 2006;368(9550):1894ā€“907.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  4. Brown P. Adding WT1 to childhood AML alphabet soup. Blood. 2009;113(23):5696ā€“7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Banerji V, Frumm SM, Ross KN, Li LS, Schinzel AC, Hahn CK, et al. The intersection of genetic and chemical genomic screens identifies GSK-3alpha as a target in human acute myeloid leukemia. J Clin Invest. 2012;122(3):935ā€“47.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  6. Frohling S, Scholl C, Gilliland DG, Levine RL. Genetics of myeloid malignancies: pathogenetic and clinical implications. J Clin Oncol. 2005;23(26):6285ā€“95.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Kralik JM, Kranewitter W, Boesmueller H, Marschon R, Tschurtschenthaler G, Rumpold H, et al. Characterization of a newly identified ETV6-NTRK3 fusion transcript in acute myeloid leukemia. Diagn Pathol. 2011;6:19.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  8. Wander SA, Levis MJ, Fathi AT. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther Adv Hematol. 2014;5(3):65ā€“77.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  9. Masetti R, Pigazzi M, Togni M, Astolfi A, Indio V, Manara E, et al. CBFA2T3-GLIS2 fusion transcript is a novel common feature in pediatric, cytogenetically normal AML, not restricted to FAB M7 subtype. Blood. 2013;121(17):3469ā€“72.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  10. Masetti R, Togni M, Astolfi A, Pigazzi M, Manara E, Indio V, et al. DHH-RHEBL1 fusion transcript: a novel recurrent feature in the new landscape of pediatric CBFA2T3-GLIS2-positive acute myeloid leukemia. Oncotarget. 2013;4(10):1712ā€“20.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  11. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood. 2005;106(4):1154ā€“63.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Ferrara F, Schiffer CA. Acute myeloid leukaemia in adults. Lancet. 2013;381(9865):484ā€“95.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  13. Zhao J, Lu Q, Zhu J, Fu J, Chen YX. Prognostic value of miR-96 in patients with acute myeloid leukemia. Diagn Pathol. 2014;9:76.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  14. Ma QL, Wang JH, Wang YG, Hu C, Mu QT, Yu MX, et al. High IDH1 expression is associated with a poor prognosis in cytogenetically normal acute myeloid leukemia. Int J Cancer. 2014.

  15. Makeyev AV, Liebhaber SA. The poly(C)-binding proteins: a multiplicity of functions and a search for mechanisms. RNA. 2002;8(3):265ā€“78.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. Huo LR, Zhong N. Identification of transcripts and translatants targeted by overexpressed PCBP1. Biochim Biophys Acta. 2008;1784(11):1524ā€“33.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Wang H, Vardy LA, Tan CP, Loo JM, Guo K, Li J, et al. PCBP1 suppresses the translation of metastasis-associated PRL-3 phosphatase. Cancer Cell. 2010;18(1):52ā€“62.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Lian WX, Yin RH, Kong XZ, Zhang T, Huang XH, Zheng WW, et al. THAP11, a novel binding protein of PCBP1, negatively regulates CD44 alternative splicing and cell invasion in a human hepatoma cell line. FEBS Lett. 2012;586(10):1431ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  19. Zhang T, Huang XH, Dong L, Hu D, Ge C, Zhan YQ, et al. PCBP-1 regulates alternative splicing of the CD44 gene and inhibits invasion in human hepatoma cell line HepG2 cells. Mol Cancer. 2010;9:72.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  20. Yang X, Zhang Z, Jia C, Li J, Yin L, Jiang S. The relationship between expression of c-ras, c-erbB-2, nm23, and p53 gene products and development of trophoblastic tumor and their predictive significance for the malignant transformation of complete hydatidiform mole. Gynecol Oncol. 2002;85(3):438ā€“44.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  21. Pillai MR, Chacko P, Kesari LA, Jayaprakash PG, Jayaram HN, Antony AC. Expression of folate receptors and heterogeneous nuclear ribonucleoprotein E1 in women with human papillomavirus mediated transformation of cervical tissue to cancer. J Clin Pathol. 2003;56(8):569ā€“74.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  22. Shi Z, Zhang T, Long W, Wang X, Zhang X, Ling X, et al. Down-regulation of poly(rC)-binding protein 1 correlates with the malignant transformation of hydatidiform moles. Int J Gynecol Cancer. 2012;22(7):1125ā€“9.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  23. Zhang HY, Dou KF. PCBP1 is an important mediator of TGF-beta-induced epithelial to mesenchymal transition in gall bladder cancer cell line GBC-SD. Mol Biol Rep. 2014;41(8):5519ā€“24.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  24. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937ā€“51.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  25. Yanada M, Takeuchi J, Sugiura I, Akiyama H, Usui N, Yagasaki F, et al. High complete remission rate and promising outcome by combination of imatinib and chemotherapy for newly diagnosed BCR-ABL-positive acute lymphoblastic leukemia: a phase II study by the Japan Adult Leukemia Study Group. J Clin Oncol. 2006;24(3):460ā€“6.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  26. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(āˆ’Delta Delta C(T)) Method. Methods. 2001;25(4):402ā€“8.

    ArticleĀ  CASĀ  Google ScholarĀ 

  27. Han N, Li W, Zhang M. The function of the RNA-binding protein hnRNP in cancer metastasis. J Cancer Res Ther. 2013;9:S129ā€“34.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  28. Carpenter B, MacKay C, Alnabulsi A, MacKay M, Telfer C, Melvin WT, et al. The roles of heterogeneous nuclear ribonucleoproteins in tumour development and progression. Biochim Biophys Acta. 2006;1765(2):85ā€“100.

    CASĀ  PubMedĀ  Google ScholarĀ 

  29. Eder S, Lamkowski A, Priller M, Port M, Steinestel K. Radiosensitization and downregulation of heterogeneous nuclear ribonucleoprotein K (hnRNP K) upon inhibition of mitogen/extracellular signal-regulated kinase (MEK) in malignant melanoma cells. Oncotarget. 2015.

  30. Ritchie SA, Pasha MK, Batten DJP, Sharma RK, Olson DJH, Ross ARS, et al. Identification of the SRC pyrimidine-binding protein (SPy) as hnRNP K: implications in the regulation of SRC1A transcription. Nucleic Acids Research. 2003;31(5):1502ā€“13.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  31. Evans JR, Mitchell SA, Spriggs KA, Ostrowski J, Bomsztyk K, Ostarek D, et al. Members of the poly (rC) binding protein family stimulate the activity of the c-myc internal ribosome entry segment in vitro and in vivo. Oncogene. 2003;22(39):8012ā€“20.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Han W, Xin Z, Zhao Z, Bao W, Lin X, Yin B, et al. RNA-binding protein PCBP2 modulates glioma growth by regulating FHL3. J Clin Invest. 2013;123(5):2103ā€“18.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  33. Hu CE, Liu YC, Zhang HD, Huang GJ. The RNA-binding protein PCBP2 facilitates gastric carcinoma growth by targeting miR-34a. Biochem Biophys Res Commun. 2014;448(4):437ā€“42.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  34. Cloke B, Shah K, Kaneda H, Lavery S, Trew G, Fusi L, et al. The poly(c)-binding protein-1 regulates expression of the androgen receptor. Endocrinology. 2010;151(8):3954ā€“64.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  35. Chaudhury A, Hussey GS, Ray PS, Jin G, Fox PL, Howe PH. TGF-beta-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI. Nat Cell Biol. 2010;12(3):286ā€“93.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  36. Huo LR, Ju W, Yan M, Zou JH, Yan W, He B, et al. Identification of differentially expressed transcripts and translatants targeted by knock-down of endogenous PCBP1. Biochim Biophys Acta. 2010;1804(10):1954ā€“64.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  37. Nishinakamura H, Minoda Y, Saeki K, Koga K, Takaesu G, Onodera M, et al. An RNA-binding protein alphaCP-1 is involved in the STAT3-mediated suppression of NF-kappaB transcriptional activity. Int Immunol. 2007;19(5):609ā€“19.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  38. Shi H, Zhou M, Tong X, Wang H. The function and mechanism of PCBP1 in gene regulation. Chemistry of Life. 2013;33(5):535ā€“42.

    CASĀ  Google ScholarĀ 

  39. Cho SJ, Jung YS, Chen X. Poly (C)-binding protein 1 regulates p63 expression through mRNA stability. PLoS One. 2013;8(8), e71724.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  40. Giles KM, Daly JM, Beveridge DJ, Thomson AM, Voon DC, Furneaux HM, et al. The 3 '-untranslated region of p21(WAF1) mRNA is a composite cis-acting sequence bound by RNA-binding proteins from breast cancer cells, including HuR and poly(C)-binding protein. J Biol Chem. 2003;278(5):2937ā€“46.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  41. Qu S, Liu B, Guo X, Shi H, Zhou M, Li L, et al. Independent oncogenic and therapeutic significance of phosphatase PRL-3 in FLT3-ITD-negative acute myeloid leukemia. Cancer. 2014;120(14):2130ā€“41.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  42. Park JE, Yuen HF, Zhou JB, Al-Aidaroos AQ, Guo K, Valk PJ, et al. Oncogenic roles of PRL-3 in FLT3-ITD induced acute myeloid leukaemia. EMBO Mol Med. 2013;5(9):1351ā€“66.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  43. Thakur S, Nakamura T, Calin G, Russo A, Tamburrino JF, Shimizu M, et al. Regulation of BRCA1 transcription by specific single-stranded DNA binding factors. Mol Cell Biol. 2003;23(11):3774ā€“87.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

Download references

Acknowledgements

This work was supported by Natural science Foundation of Guangdong (2015 program: PCBP1 in the role and mechanism of acute myeloid leukemia) to TX and Science and Technology Planning Project of Guangdong Province (No. 2013B021800127) to TX.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiuzhen Tong.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authorsā€™ contributions

XT conceived and designed the experiments. MZ carried out the experiments and participated in the data analysis and drafted the manuscript. All authors read and approved the final manuscript.

This article is retracted at request of the Editor. After publication of the article concerns were raised regarding inadequate documentation of the statistical methodology and use of inappropriate statistical analyses in the study. For these reasons the conclusions of the study cannot be considered to be supported by the data. The authors do not agree to this retraction.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, M., Tong, X. RETRACTED ARTICLE: Downregulated Poly-C binding protein-1 is a novel predictor associated with poor prognosis in Acute Myeloid Leukemia. Diagn Pathol 10, 147 (2015). https://doi.org/10.1186/s13000-015-0377-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13000-015-0377-y

Keywords