Skip to main content
Erschienen in: Molecular Cancer 1/2015

Open Access 01.12.2015 | Short communication

Epigenetic silencing of a long non-coding RNA KIAA0495 in multiple myeloma

verfasst von: Kwan Yeung Wong, Zhenhai Li, Xiaoqin Zhang, Gilberto Ka Kit Leung, Godfrey Chi-fung Chan, Chor Sang Chim

Erschienen in: Molecular Cancer | Ausgabe 1/2015

Abstract

In multiple myeloma, a long non-coding RNA, KIAA0495 (alias PDAM/TP73-AS1), had been found progressively downregulated from normal plasma cell to benign monoclonal gammopathy of undetermined significance to symptomatic myeloma. Herein, by methylation-specific PCR, the putative KIAA0495 promoter was found unmethylated in all healthy controls (N = 14) but methylated in 50 % of myeloma cell lines (N = 10). KIAA0495 methylation was shown inversely correlated with KIAA0495 expression. However, KIAA0495 methylation was detected in none of both primary myeloma samples at diagnosis (N = 61) and at relapse/progression (N = 16). Collectively, despite frequently methylated in cell lines, KIAA0495 methylation appeared unimportant in the pathogenesis or progression of myeloma.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12943-015-0444-8) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

KYW and ZZ carried out the experiments. KYW, ZZ and CSC drafted the manuscript. KYW and CSC conceived of the study. All authors participated in the design of the study. All authors read and approved the final manuscript.
Abkürzungen
lncRNA
Long non-coding RNA
MGUS
Monoclonal gammopathy of undetermined significance
miRNA
microRNA
MSP
Methylation-specific PCR
M-MSP
Methylated MSP
U-MSP
Unmethylated MSP
PCR
Polymerase chain reaction
RT-PCR
Reverse transcription-PCR
SMM
Smouldering multiple myeloma

Introduction

Long non-coding RNA (lncRNA) is a novel class of functional RNA molecules >200 nucleotides with little or no protein-coding capacity [1, 2]. Based on their genomic locations relative to annotated protein-coding genes, lncRNAs are broadly classified into intergenic, intragenic, antisense, pseudogene, or divergent transcripts [3, 4]. Functionally, lncRNAs play a role in, but not limited to, development, differentiation, and carcinogenesis [2, 5]. Moreover, lncRNAs are found dysregulated and therefore potentially to be oncogenic or tumour suppressive in human cancers [6, 7].
Table 1
Primer sequences and PCR reaction conditions
Primer set
Forward primer (5’ – 3’)
Reverse primer (5’ – 3’)
Product size (bp)
MgCl2/Tm/cycles
Reference
(I) Methylation-specific polymerase chain reaction (MSP)
   
KIAA0495
     
M-MSP
TGG AGA TAA CGG GTT TAG AGA AAT C
AAC GAA AAC AAA AAT AAA ACC TTC G
191
1.5 mM/58 °C/40X
--
U-MSP
TGG AGA TAA TGG GTT TAG AGA AAT T
AAA CAA AAA CAA AAA TAA AAC CTT CA
192
2.0 mM/58 °C/40X
--
(II) Reverse transcription-polymerase chain reaction (RT-PCR)
   
KIAA0495
GCT GCT TGC TGT ACG TGG TG
CGT GGC TGA CAC AAA CTT GC
178
1.5 mM/60 °C/35X
[16]
GAPDH
ACC ACA GTC CAT GCC ATC ACT
TCC ACC ACC CTG TTG CTG TA
452
1.5 mM/60 °C/24X
 
Keys: Tm, annealing temperature; M-MSP, MSP for the methylated allele; U-MSP, MSP for the unmethylated allele
Multiple myeloma is a form of haematological cancer originated from malignant transformation of plasma cells [8]. Clinically, this disease begins with benign monoclonal gammopathy of undetermined significance (MGUS), some of which may undergo asymptomatic smouldering multiple myeloma (SMM), and progresses to symptomatic myeloma [9]. Genetically, despite D-type cyclins are apparently upregulated in all patients, myeloma is a heterogeneous disease characterized by specific gains or losses of chromosomes, or reciprocal translocations, such as t(4;14)(p16.3;q32) and t(14;16)(q32;q23) [10].
DNA methylation refers to the catalytic addition of a methyl (−CH3) group to the cytosine ring of a CpG dinucleotide [11]. Human cancers are characterized by loss of global DNA methylation but gain of methylation at promoter-associated CpG islands (a cluster of CpG dinucleotides) and hence transcriptional silencing of specific tumour suppressor genes or miRNAs [12]. In myeloma, DNA methylation has been shown to mediate silencing of multiple tumour suppressor genes and miRNAs, and has been implicated in the pathogenesis and prognosis of the disease [13, 14].
Interestingly, by gene expression profiling, KIAA0495, a lncRNA transcribed from chromosome 1p36, has been shown to be progressively downregulated from normal plasma cell to MGUS to symptomatic myeloma [15]. Moreover, methylation-mediated silencing of KIAA0495 has been demonstrated in oligodendroglial tumours, leading to enhanced cisplatin resistance via upregulation of anti-apoptotic B-cell CLL/lymphoma 2-like 1 (BCL2L1) [16]. Hence, we hypothesized that DNA methylation may account for the progressive downregulation of KIAA0495 in the pathogenesis of myeloma. Herein, we have studied and reported methylation of KIAA0495 in myeloma cell lines, primary myeloma marrow samples at diagnosis, and at relapse/progression. Materials and methods have been incorporated as Additional file 1.

Findings

Methylation-specific PCR: KIAA0495 methylation in healthy controls and myeloma cell lines

While the putative KIAA0495 promoter region was found embedded in a CpG island (Additional file 2: Figure S1), MSP primers were designed to study methylation of this CpG island in a panel of healthy controls [peripheral (N = 10) and marrow (N = 3) buffy coat, CD138-sorted healthy plasma cell (N = 1)], and myeloma cell lines (N = 10) (Table 1). Direct sequencing analysis of M-MSP products from bisulfite-treated positive control showed expected conversion of unmethylated cytosine into uracil (turned to thymidine after PCR), with methylated cytosine remained unchanged, indicating complete bisulfite conversion and MSP specificity (Fig. 1a). None of 14 healthy controls showed KIAA0495 methylation (Fig. 1b). On the other hand, in myeloma cell lines, KMS-12-PE, LP-1, NCI-H929, OPM-2, and OCI-MY5 were partially methylated, whereas MOLP-8, RPMI-8226, U-266, WL-2, and JJN-3 were completely unmethylated for KIAA0495 (Fig. 1c). Moreover, the MSP methylation statuses of controls and cell lines were confirmed by quantitative bisulfite pyrosequencing (Fig. 1d). These data suggested that methylation of KIAA0495 was tumour-specific, consistent with other tumour suppressor protein-coding genes and non-coding miRNAs in myeloma [1720].

KIAA0495 methylation and expression in myeloma cell lines

To determine whether KIAA0495 methylation was associated with silencing of KIAA0495 expression, the expression level of KIAA0495 was measured by semi-quantitative RT-PCR and quantitative real-time PCR (qPCR), and correlated with the KIAA0495 methylation status as detected by MSP in myeloma cell lines. Of the 10 myeloma cell lines, KIAA0495 methylation was associated with lower expression as detected by both semi-quantitative RT-PCR (P = 0.0079; Fig. 2a) and quantitative real-time PCR (P = 0. 000155; Fig. 2b).
Moreover, myeloma cell lines with KIAA0495 methylation were treated with a hypomethylating agent, 5-AzadC, followed by pyrosequencing and qPCR analyses. Upon 5-AzadC treatment, LP-1 and OCI-MY5 cells, which were partially methylated for KIAA0495, showed progressive demethylation of the KIAA0495 promoter, as evidenced by the decreased methylation percentage on a stretch of seven consecutive CpG dinucleotides, together with concomitant re-expression of the KIAA0495 transcript, as illustrated by the increased expression relative to the untreated control (LP-1: P = 0.0013; OCI-MY5: P = 0.0013; Fig. 2c). Nevertheless, when 5-AzadC-treated cells were continually cultured with fresh medium in the absence of 5-AzadC for a further 6 days, methylation of the KIAA0495 promoter was restored, with simultaneous suppression of the KIAA0495 expression (LP-1: P = 0.0331; OCI-MY5: P = 0.0447; Fig. 2c). Taken together, the methylation of KIAA0495 was inversely correlated with KIAA0495 expression level in myeloma cell lines, similar to the methylation-mediated silencing of KIAA0495 demonstrated in glioma cell lines and primary oligodendroglial tumour cells [16], suggesting that methylation of the promoter-associated CpG island emerged to be one of the mechanisms resulting in the regulation of lncRNAs in cancer cells.

Methylation-specific PCR: KIAA0495 methylation in myeloma primary samples at diagnosis and at relapse

To examine if methylation of KIAA0495 was also detected in primary samples, methylation of KIAA0495 was studied in 61 primary samples at diagnosis and 16 primary samples at relapse by MSP. However, none of these samples showed methylation of KIAA0495 (Fig. 3), indicating methylation of KIAA0495 was rarely detected in primary myeloma samples. Hence, similar to the studies of miR-9-1, miR-9-3, and miR-124-1 [21, 22], these data suggested that methylation of certain tumour suppressor non-coding miRNAs or lncRNAs was acquired in vitro during continuous culture of myeloma cells, hence not pathogenic. Therefore, methylation was not the mechanism leading to the progressive downregulation of KIAA0495 from normal plasma cell to MGUS to symptomatic myeloma [15], suggestive of other mechanisms, such as histone modification and miRNA-mediated repression, may come into play. For instance, in gastric cancer cells, histone deacetylation was found to be associated in the downregulation of a tumour suppressor lncRNA, FENDRR [23]. Moreover, in breast cancer cells, miR-21 was shown to target and hence downregulate the expression of a tumour suppressor lncRNA, GAS5 [24]. Furthermore, while KIAA0495 resides on 1p36, which has been shown frequently deleted in newly diagnosed patients with myeloma [25], the loss of expression of KIAA0495 may be associated with chromosome deletion. Last but not least, inactivation of KIAA0495 may also be accounted by haploinsufficiency, which has been demonstrated in the TP53 in myeloma [26].

Conclusion

Methylation of KIAA0495 is tumour-specific, leading to reversible silencing of KIAA0495 expression in myeloma cell lines. On the other hand, it is rarely detected in primary samples of myeloma at diagnosis or at relapse, and hence not responsible for the progressive downregulation of KIAA0495 from normal plasma cell to MGUS to symptomatic myeloma, as shown by the gene expression profiling, and remains as a possible in vitro event acquired during continuous culture of myeloma cells.
Open AccessThis 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.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

KYW and ZZ carried out the experiments. KYW, ZZ and CSC drafted the manuscript. KYW and CSC conceived of the study. All authors participated in the design of the study. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155–9.CrossRefPubMed Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155–9.CrossRefPubMed
2.
Zurück zum Zitat Spizzo R, Almeida MI, Colombatti A, Calin GA. Long non-coding RNAs and cancer: a new frontier of translational research. Oncogene. 2012;31:4577–87.PubMedCentralCrossRefPubMed Spizzo R, Almeida MI, Colombatti A, Calin GA. Long non-coding RNAs and cancer: a new frontier of translational research. Oncogene. 2012;31:4577–87.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 2012;22:1775–89.PubMedCentralCrossRefPubMed Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 2012;22:1775–89.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15:7–21.CrossRefPubMed Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15:7–21.CrossRefPubMed
8.
Zurück zum Zitat Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer. 2012;12:335–48.CrossRefPubMed Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer. 2012;12:335–48.CrossRefPubMed
9.
Zurück zum Zitat Kyle RA, Durie BGM, Rajkumar SV, Landgren O, Blade J, Merlini G, et al. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management. Leukemia. 2010;24:1121–7.CrossRefPubMed Kyle RA, Durie BGM, Rajkumar SV, Landgren O, Blade J, Merlini G, et al. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management. Leukemia. 2010;24:1121–7.CrossRefPubMed
10.
Zurück zum Zitat Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK, et al. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23:2210–21.PubMedCentralCrossRefPubMed Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK, et al. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23:2210–21.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484–92.CrossRefPubMed Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484–92.CrossRefPubMed
13.
Zurück zum Zitat Chim CS, Kwong YL, Liang R. Gene hypermethylation in multiple myeloma: lessons from a cancer pathway approach. Clin Lymphoma Myeloma. 2008;8:331–9.CrossRefPubMed Chim CS, Kwong YL, Liang R. Gene hypermethylation in multiple myeloma: lessons from a cancer pathway approach. Clin Lymphoma Myeloma. 2008;8:331–9.CrossRefPubMed
14.
Zurück zum Zitat Wong KY, Huang X, Chim CS. DNA methylation of microRNA genes in multiple myeloma. Carcinogenesis. 2012;33:1629–38.CrossRefPubMed Wong KY, Huang X, Chim CS. DNA methylation of microRNA genes in multiple myeloma. Carcinogenesis. 2012;33:1629–38.CrossRefPubMed
15.
Zurück zum Zitat Zhan F, Barlogie B, Arzoumanian V, Huang Y, Williams DR, Hollmig K, et al. Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis. Blood. 2007;109:1692–700.PubMedCentralCrossRefPubMed Zhan F, Barlogie B, Arzoumanian V, Huang Y, Williams DR, Hollmig K, et al. Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis. Blood. 2007;109:1692–700.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Pang JCS, Li KKW, Lau KM, Ng YL, Wong J, Chung NYF, et al. KIAA0495/PDAM Is Frequently Downregulated in Oligodendroglial Tumors and Its Knockdown by siRNA Induces Cisplatin Resistance in Glioma Cells. Brain Pathol. 2010;20:1021–32.CrossRefPubMed Pang JCS, Li KKW, Lau KM, Ng YL, Wong J, Chung NYF, et al. KIAA0495/PDAM Is Frequently Downregulated in Oligodendroglial Tumors and Its Knockdown by siRNA Induces Cisplatin Resistance in Glioma Cells. Brain Pathol. 2010;20:1021–32.CrossRefPubMed
17.
Zurück zum Zitat Chim CS, Fung TK, Cheung WC, Liang R, Kwong YL. SOCS1 and SHP1 hypermethylation in multiple myeloma: implications for epigenetic activation of the Jak/STAT pathway. Blood. 2004;103:4630–5.CrossRefPubMed Chim CS, Fung TK, Cheung WC, Liang R, Kwong YL. SOCS1 and SHP1 hypermethylation in multiple myeloma: implications for epigenetic activation of the Jak/STAT pathway. Blood. 2004;103:4630–5.CrossRefPubMed
18.
Zurück zum Zitat Chim CS, Pang R, Fung TK, Choi CL, Liang R. Epigenetic dysregulation of Wnt signaling pathway in multiple myeloma. Leukemia. 2007;21:2527–36.CrossRefPubMed Chim CS, Pang R, Fung TK, Choi CL, Liang R. Epigenetic dysregulation of Wnt signaling pathway in multiple myeloma. Leukemia. 2007;21:2527–36.CrossRefPubMed
19.
Zurück zum Zitat Wong KY, Liang R, So CC, Jin DY, Costello JF, Chim CS. Epigenetic silencing of MIR203 in multiple myeloma. Br J Haematol. 2011;154:569–78.CrossRefPubMed Wong KY, Liang R, So CC, Jin DY, Costello JF, Chim CS. Epigenetic silencing of MIR203 in multiple myeloma. Br J Haematol. 2011;154:569–78.CrossRefPubMed
20.
Zurück zum Zitat Wong KY, Yim RLH, So CC, Jin D-Y, Liang R, Chim CS. Epigenetic inactivation of the MIR34B/C in multiple myeloma. Blood. 2011;118:5901–4.CrossRefPubMed Wong KY, Yim RLH, So CC, Jin D-Y, Liang R, Chim CS. Epigenetic inactivation of the MIR34B/C in multiple myeloma. Blood. 2011;118:5901–4.CrossRefPubMed
21.
Zurück zum Zitat Wong KY, So CC, Loong F, Chung LP, Lam WW, Liang R, et al. Epigenetic inactivation of the miR-124-1 in haematological malignancies. PloS one. 2011;6, e19027.PubMedCentralCrossRefPubMed Wong KY, So CC, Loong F, Chung LP, Lam WW, Liang R, et al. Epigenetic inactivation of the miR-124-1 in haematological malignancies. PloS one. 2011;6, e19027.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Zhang Q, Wang LQ, Wong KY, Li ZY, Chim CS. Infrequent DNA methylation of miR-9-1 and miR-9-3 in multiple myeloma. J Clin Pathol. 2015;68:557–61.CrossRefPubMed Zhang Q, Wang LQ, Wong KY, Li ZY, Chim CS. Infrequent DNA methylation of miR-9-1 and miR-9-3 in multiple myeloma. J Clin Pathol. 2015;68:557–61.CrossRefPubMed
23.
Zurück zum Zitat Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, et al. Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 2014;7:63.PubMedCentralCrossRefPubMed Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, et al. Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 2014;7:63.PubMedCentralCrossRefPubMed
24.
25.
Zurück zum Zitat Walker BA, Leone PE, Chiecchio L, Dickens NJ, Jenner MW, Boyd KD, et al. A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value. Blood. 2010;116:e56–65.CrossRefPubMed Walker BA, Leone PE, Chiecchio L, Dickens NJ, Jenner MW, Boyd KD, et al. A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value. Blood. 2010;116:e56–65.CrossRefPubMed
26.
Zurück zum Zitat Teoh PJ, Chung TH, Sebastian S, Choo SN, Yan J, Ng SB, et al. p53 haploinsufficiency and functional abnormalities in multiple myeloma. Leukemia. 2014;28:2066–74.PubMed Teoh PJ, Chung TH, Sebastian S, Choo SN, Yan J, Ng SB, et al. p53 haploinsufficiency and functional abnormalities in multiple myeloma. Leukemia. 2014;28:2066–74.PubMed
Metadaten
Titel
Epigenetic silencing of a long non-coding RNA KIAA0495 in multiple myeloma
verfasst von
Kwan Yeung Wong
Zhenhai Li
Xiaoqin Zhang
Gilberto Ka Kit Leung
Godfrey Chi-fung Chan
Chor Sang Chim
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2015
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0444-8

Weitere Artikel der Ausgabe 1/2015

Molecular Cancer 1/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.