Next Article in Journal
Long COVID Complicated by Fatal Cytomegalovirus and Aspergillus Infection of the Lungs: An Autopsy Case Report
Previous Article in Journal
The Role of Epinephelus coioides DUSP5 in Regulating Singapore Grouper Iridovirus Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

EBV and 1q Gains Affect Gene and miRNA Expression in Burkitt Lymphoma

Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Clinic Hamburg-Eppendorf, 20251 Hamburg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2023, 15(9), 1808; https://doi.org/10.3390/v15091808
Submission received: 18 July 2023 / Revised: 3 August 2023 / Accepted: 4 August 2023 / Published: 25 August 2023
(This article belongs to the Section Human Virology and Viral Diseases)

Abstract

:
Abnormalities of the long arm of chromosome 1 (1q) represent the most frequent secondary chromosomal aberrations in Burkitt lymphoma (BL) and are observed almost exclusively in EBV-negative BL cell lines (BL-CLs). To verify chromosomal abnormalities, we cytogenetically investigated EBV-negative BL patient material, and to elucidate the 1q gain impact on gene expression, we performed qPCR with six 1q-resident genes and analyzed miRNA expression in BL-CLs. We observed 1q aberrations in the form of duplications, inverted duplications, isodicentric chromosome idic(1)(q10), and the accumulation of 1q12 breakpoints, and we assigned 1q21.2–q32 as a commonly gained region in EBV-negative BL patients. We detected MCL1, ARNT, MLLT11, PDBXIP1, and FCRL5, and 64 miRNAs, showing EBV- and 1q-gain-dependent dysregulation in BL-CLs. We observed MCL1, MLLT11, PDBXIP1, and 1q-resident miRNAs, hsa-miR-9, hsa-miR-9*, hsa-miR-92b, hsa-miR-181a, and hsa-miR-181b, showing copy-number-dependent upregulation in BL-CLs with 1q gains. MLLT11, hsa-miR-181a, hsa-miR-181b, and hsa-miR-183 showed exclusive 1q-gains-dependent and FCRL5, hsa-miR-21, hsa-miR-155, hsa-miR-155*, hsa-miR-221, and hsa-miR-222 showed exclusive EBV-dependent upregulation. We confirmed previous data, e.g., regarding the EBV dependence of hsa-miR-17-92 cluster members, and obtained detailed information considering 1q gains in EBV-negative and EBV-positive BL-CLs. Altogether, our data provide evidence for a non-random involvement of 1q gains in BL and contribute to enlightening and understanding the EBV-negative and EBV-positive BL pathogenesis.

1. Introduction

Abnormalities affecting the long arm of chromosome 1 (1q) are among the most common chromosomal alterations in human neoplasm and are known to frequently occur as a secondary change during disease progression [1,2,3,4]. In B-cell non-Hodgkin’s lymphomas (B-NHL), 1q rearrangements are observed at high frequency and are associated with a shortened median survival [2,5]. In Burkitt lymphoma (BL), structural abnormalities of 1q represent the most frequent secondary chromosomal aberrations and have been seen in 31% of all cytogenetically characterized cases [6].
BL is a highly aggressive B-NHL [7]. According to the clinical characteristics, endemic (predominantly in tropical Africa, Epstein–Barr virus-positive (EBV+)), sporadic (outside of tropical Africa, rarely EBV+), and HIV-associated BL forms are common [8]. Cytogenetically, BL is defined by the presence of the chromosomal translocation t(8;14)(q24;q32) or its variants t(2;8)(p12;q24) and t(8;22)(q24;q11) [6,9,10]. These translocations induce the overexpression of the oncogene MYC due to its juxtaposition to the immunoglobulin heavy-chain (IGH) locus on chromosome 14, or in their variants to the immunoglobulin light-chain loci kappa (IGK) and lambda (IGL) on chromosomes 2 and 22, respectively. In addition to MYC rearrangements, duplications involving the whole 1q arm or parts of it, whole 1q arm translocations, or jumping translocations involving 1q are widely reported in BL [11,12,13,14,15]. Duplications of the 1q21 region are found in up to 44% of BL cases [16]; however, the precise nature of cytogenetic aberrations leading to these molecular rearrangements in BL has so far not been analyzed in detail [6,16]. Moreover, being the most frequent secondary chromosomal change, the prognostic significance of 1q gains in BL is controversial. According to some authors, abnormalities involving 1q are associated with a poor prognosis and short survival in BL patients [2,5,17,18,19], whereas in some other reports, they do not seem to have any impact on the outcomes [20,21], underlining the importance of detailed investigations of these frequent but poorly understood cytogenetic alterations.
Micro-RNAs (miRNAs) are a group of endogenously encoded, short (~22 nucleotides), single-stranded, non-coding but functional RNA molecules that influence almost every cellular process, including development, differentiation, proliferation, and apoptosis, in various organisms [22,23,24,25]. The tightly regulated interaction between miRNAs and their target(s) in healthy cells is lost in cancer, and this dysregulation allows the expression of proteins with oncogenic properties, which seem to play significant roles not only in the initiation and development of cancer and the promotion of neoplastic transformation and tumorigenesis, but also in contributing to the classification and prognosis of different cancer entities [23,25,26,27,28,29,30]. Additionally, in BL, aberrant miRNA and mRNA expression profiles are observed [31,32], whereas differences in expression associated with 1q aberrations have not yet been reported.
In our previous study, we cytogenetically analyzed well-established BL cell lines (BL-CLs) and observed aberrations involving 1q as the most frequent structural change, as well as an accumulation of 1q12 breaks [33]. Furthermore, we found a correlation between the absence of EBV and the presence of 1q gains, and defined 1q21.2–q22 as a minimal commonly gained region, when duplications and partial trisomies were considered together [33]. In order to prove this correlation on primary material, we cytogenetically analyzed three EBV-negative (EBV−) patient samples. Furthermore, we investigated EBV status- and 1q-gain-dependent gene and miRNA expression in BL-CLs in order to find specific patterns for EBV+ and EBV− BL. Altogether, our results indicate an accumulation of 1q aberrations in BL and a non-random involvement of 1q aberrations in EBV− and EBV+ BL by affecting mRNA and miRNA expression.

2. Materials and Methods

2.1. BL Patient Samples and Cell Lines

Three EBV− BL patient samples were selected from the archival methanol-acetic-acid-fixed cytogenetic preparations from the University Medical Center, Hamburg-Eppendorf, Hamburg, Germany (n = 1, patient-1 (male), Table 1), and the Center for Human Genetics of the University of Leuven, Leuven, Belgium (n = 2, patient-2 (male) and -3 (female), Table 1). All BL patients were studied at the time of primary diagnosis, were classified according to the WHO classification [7], and showed the typical histological features of BL. The G-banding karyotypes of the patients at initial diagnosis are summarized in Table 1. Our study was performed in accordance with the Declaration of Helsinki from 1975 and informed consent was obtained from all patients.
Since fresh, frozen primary material from patients was not available, gene and miRNA expression were investigated using BL-CLs. The main characteristics of the used BL-CLs are summarized in Table 2, and their updated karyotypes are published in our previous report [33].

2.2. Multicolor Fluorescence In Situ Hybridization and Probe Selection for Metaphase FISH Mapping of 1q Abnormalities

Multicolor fluorescence in situ hybridization (mFISH) was performed using the 24Xcyte color kit for human chromosomes (MetaSystems, Altlussheim, Germany) according to the supplier’s recommendations. Dual-color metaphase FISH was performed according to our previous publication [33]. For the metaphase FISH 37, partially overlapping BAC and PAC clones spanning the whole 1q-arm were selected from information archived (on January 2005) by the National Center for Biotechnology Information (NCBI) (http://www.ncbi.nlm.nih.gov/; 1 January 2005) and obtained from the RPCI-1, -3, -4 and -11 libraries (Roswell Park Cancer Institute, Buffalo, NY, USA) (Supplemental Table S1). Details of mFISH and FISH are summarized in the supplemental materials and methods sections. Karyotypes were described according to the International System for Human Cytogenetic Nomenclature (ISCN, 2020) [34] and revised using the online analysis webpage CyDAS (http://www.cydas.org/, 1 July 2023).

2.3. RNA Preparation, Reverse Transcription and Real-Time qPCR Analysis

Total RNA was isolated from 37 BL-CLs (Table 2) using the RNeasy Mini Kit (QIAGEN, Hilden, Germany) and cDNA synthesis was performed using SuperScript™ II RT (Invitrogen, Darmstadt, Germany) according to the manufacturer’s instructions. Real-time qPCR was performed on a Light Cycler Instrument (Roche, Basel, Switzerland) using the QuantiFast SYBR Green PCR Kit (QIAGEN). Primers for HMBS were obtained from Eurofins (Ebersberg, Germany). The QuantiTect Primer Assay from QIAGEN (Supplemental Table S2) was used for BCL9, MCL1, ARNT, MLLT11, PBXIP1 and FCRL5 amplification. The qPCR data were analyzed according to the comparative CT method (Applied Biosystems, Darmstadt, Germany) using the endogenous reference HMBS as a normalization factor. Normalized mRNA levels detected in BL-28, i.e., an EBV− BL-CL without 1q gains, were averaged and set to 1 and the amounts measured in the remaining BL-CLs were calculated relative to those levels. Each sample was measured in two independent qPCR experiments prepared as duplicates.

2.4. Micro RNA Analysis

MiRNA was isolated from 12 BL-CLs (Table 2, dark grey highlighted) during two independent days using the miRNeasy mini kit (QIAGEN) according to the manufacturer’s instructions. MiRNA expression analysis using miRCURY Hy3/Hy5 dual-color labeling and hybridization with the miRCURY™ LNA Array (EXIQON, Vedbaek, Denmark) was performed by the company EXIQON. The clustering was performed using log2(Hy3/Hy5) ratios, which passed the filtering criteria on variation across sample groups using a two-tailed t-test; a p-value < 0.05 and only miRNA with values showing a significant difference were considered.

3. Results

3.1. EBV-Negative BL Accumulate 1q Gains

Gains of 1q arm were mainly observed in EBV− BL-CLs [33,35,36]. We observed a correlation between the absence of EBV and gain of 1q in BL-CLs [33]. To verify this correlation on primary material, we analyzed the samples from three EBV− BL patients. To determine the exact position of the breakpoints and to uncover cryptic 1q rearrangements, we performed classical G-banding, mFISH and FISH with 37 specific clones covering the whole 1q arm (Supplemental Table S1). As expected, the characteristic MYC rearrangement for BL involving 8q24 was present in all investigated patient samples detected as t(8;14)(q24;q32) (Table 1). The gain of 1q-arm was observed in all three patient samples; in contrast, balanced translocations involving 1q, loss of 1q or rearrangements involving 1p were not observed.
In samples of patient-1 and -2, we found intra-chromosomal duplication of 1q and interestingly, they uncovered concomitant inversions of the duplicated regions. Moreover, the sample from patient-1 displayed the concomitant presence of two cytogenetic clones with a duplication and an inverted duplication of the same 1q region (Figure 1, Table 1). In the sample of patient-2, we observed several sub-clones characterized by distinct and specific chromosome 1 rearrangements including the presence of partial trisomy of 1q presented as a translocation of 1q (region 1q12–q44) to the recurring partner at 7p22 on the der(7) and inverted duplication of 1q41–q21.2 (Figure 1E, Table 1). In the sample of patient-3, we observed, with all the applied 1q probes, three hybridization signals with a breakpoint at 1q10 indicating the presence of an isodicentric chromosome 1 idic(1)(q10) (Figure 1E, Table 1).
Considering the duplications and inverted duplications detected in the samples from patient-1 and in two clones of patient-2, the proximal border of 1q gains was flanked by the pericentric heterochromatic region 1q12 and 1q21.2 and the distal border by 1q32.1 and 1q41 (Figure 1E, Table 1). This clustering indicates the region 1q21.2–1q32.1 as a commonly gained region in the primary material.
Taken together, we observed in all EBV− BL patient samples 1q gains as the most prominent structural aberration in the form of duplications and inverted duplications and accumulation of 1q12 breaks. We assigned 1q21.2–q32.1 as a commonly gained region for the primary material and detected an isodicentric chromosome 1 idic(1)(q10) in BL.

3.2. EBV and 1q Gains Affect the Expression of Genes Localized at 1q21.2–q22

Using BL-CLs, we defined 1q21.2–q22 as minimal commonly gained region, which was flanked in its centromeric margin by the clone RP11-54A4 (MCL1) and telomeric margin by RP11-42A2 (telomeric to MLLT11) (Figure 2G, Supplemental Table S1) [33]. In order to analyze whether EBV and/or 1q gains affect the expression of genes located within or flanking this minimal commonly gained region, we used 37 BL-CLs (Table 2) and performed qPCR with six genes, namely BCL9, MCL1, ARNT, MLLT11, PBXIP1 and FCRL5 (Figure 2 and Figure 3), which are reported to be involved in B-lymphocyte development and malignancy.
We observed similar BCL9 expression in all investigated groups with a slight increase in EBV+ BL-CLs with 1q gains (Figure 2A). Compared to EBV+ BL-CLs without 1q gains, the MCL1 and PBXIP1 levels measured in EBV+ BL-CLs with 1q gains were 2-fold higher (Figure 2B,E and Figure 3). We detected 1.5-fold higher ARNT expression in EBV− BL-CLs with 1q gains compared to EBV+ BL-CLs with 1q gains (Figure 2C and Figure 3). The MLLT11 levels in EBV− BL-CLs with 1q gains were 4-fold higher compared to EBV− BL-CLs without 1q gains and 2-fold higher compared to EBV+ BL-CLs with 1q gains (Figure 2D and Figure 3). Compared to EBV− BL-CL, the FCRL5 amounts in EBV+ BL-CLs with and without 1q gains were 4-fold and 57-fold higher, respectively (Figure 2F and Figure 3).
Taken together, MCL1, ARNT, MLLT11, PBXIP1 and FCRL5, genes located within the minimal commonly gained region 1q21.2–q22 as assigned in BL-CLs [33], showed EBV- and 1q-gains-dependent expression differences. We observed FCRL5 upregulated in EBV+ BL-CLs, MCL1 and PBXIP1 in EBV+ BL-CLs with 1q gains and MLLT11 and ARNT in EBV− BL-CLs with 1q gains.

3.3. BL Cell Lines Show EBV- and 1q-Gains-Dependent Specific miRNA Expression Patterns

Aberrant miRNA and mRNA expression profiles were detected in different cancer entities and MYC was shown to upregulate, among others, the oncomiR miR-17-92 cluster [31,32,37], whereas an association to the cytogenetic status, especially 1q gains, was not reported. Additionally, EBV is associated with BL malignancy, whereas the characteristics of EBV− BL have not been fully investigated Thus, in order to analyze whether EBV or 1q gains affect miRNA expression, we investigated 12 BL-CLs (Table 2) and detected 64 differentially expressed miRNAs (Figure 3 and Figure 4, Supplemental Tables S3 and S4). The miRNA and qPCR results from the previous subsection are summarized in Figure 3.
We observed 38 miRNAs that were dependent on 1q gains and significantly dysregulated in EBV+ and 18 in EBV− BL-CLs (Figure 3 and Figure 4, Supplemental Table S3). In contrast, 26 miRNAs were EBV-dependent and significantly dysregulated in BL-CLs with 1q gains and 22 in those without 1q gains (Figure 3 and Figure 4, Supplemental Table S4). Three miRNAs, namely hsa-miR-181a, hsa-miR-181b and hsa-miR-183, are independent of their EBV status upregulated in BL-CLs with 1q gains (Figure 4A, no. #6-#8). In addition, hsa-miR-125b, hsa-miR-27a, hsa-miR-99a, hsa-miR-100 and hsa-miRPlus-E1038 were upregulated and hsa-miR-675 and hsa-miR-138 were downregulated in EBV+ BL-CLs with 1q gains compared to EBV+ BL-CLs without 1q gains and EBV− BL-CLs with 1q gains (Figure 4A,B, no. #9-#15). The hsa-miR-17-92 cluster members (hsa-miR-17, hsa-miR-17*, hsa-miR-18a, hsa-miR-20a, hsa-miR-19b, hsa-miR-92a) and their paralogues (hsa-miR-18b, hsa-miR-20b, hsa-miR-106a and hsa-miR-93) were EBV-dependent and upregulated in BL-CLs with 1q gains (Figure 4C, no. #21-#31). Moreover, hsa-miR-21, hsa-miR-155/-155*, hsa-miR-221 and hsa-miR-222 were EBV-dependent and upregulated in EBV+ BL-CLs, both with and without 1q gains, whereby hsa-miR-155 was more downregulated in EBV− BL-CLs with 1q gains (Figure 4D,F, no. #39,#57-#60). Additionally, hsa-miR-9, hsa-miR-148a, hsa-miR-198, hsa-miR-335, hsa-miRPlus-E1117 and hsa-miRPlus-E1168 were upregulated and has-miR-22, hsa-miR-28-5p and hsa-miR-193b were downregulated in EBV− BL-CLs with 1q gains compared to EBV− BL-CLs without 1q gains (Figure 4D,E, no. #40-#48). We also observed both mature and passenger strands in five miRNAs: hsa-miR-9/-9*, hsa-miR-17/-17*, hsa-miR-20a/-20a* and hsa-miR-155/-155*, which were dysregulated in a similar way, whereas only the passenger strand for hsa-miR-124* was significantly dysregulated in the investigated BL-CLs (Figure 4A,C,D,F, no. #2,#40,#21,#22,#24,#25,#39,#58,#36). Interestingly, hsa-miR-1290 was downregulated in all investigated BL-CLs and showed a stronger downregulation in both EBV+ BL-CLs with 1q gains and EBV− BL-CLs without 1q gains (Figure 4A, no. #3).
It is also noteworthy that 5 of the 64 significantly dysregulated miRNAs were localized on the long arm of chromosome 1, namely hsa-miR-181a and hsa-miR-181b at 1q32.1, hsa-miR-9, hsa-miR-9* and hsa-miR-92b at 1q22, and all of them were upregulated in BL-CLs with 1q gains (Figure 4A,D, no. #6,#7,#2,#40,#33). While hsa-miR-92b was upregulated in EBV+ BL-CLs with 1q gains, hsa-miR-181a and hsa-miR-181b were both upregulated in EBV+ and EBV− BL-CLs with 1q gains. In contrast, hsa-miR-9 and hsa-miR-9* were upregulated in EBV− BL-CLs with 1q gains. Additionally, hsa-miR-9* was downregulated in EBV+ BL-CLs with 1q gains and upregulated in those without 1q gains (Figure 4A,D, no. #6,#7,#2,#40,#33, Supplemental Tables S3 and S4).
Taken together, we observed 64 miRNAs that were EBV- and 1q-gains-dependent dysregulated in BL-CLs. Interestingly, five miRNAs, namely hsa-miR-181a, hsa-miR-181b, hsa-miR-9, hsa-miR-9* and hsa-miR-92b localized to 1q, were upregulated in BL-CLs with 1q gains. Additionally, hsa-miR-181a, hsa-miR-181b and hsa-miR-183 were upregulated in BL-CLs with 1q gains independent of their EBV status and hsa-miR-21, hsa-miR-155, hsa-miR-155*, hsa-miR-221, and hsa-miR-222 were upregulated in EBV+ BL-CLs independent of their 1q gains status.

4. Discussion

Chromosomal rearrangements affecting 1q are the most common secondary abnormalities observed in BL [6,33]. In our previous report, we described a correlation between the absence of EBV and gain of 1q in BL-CLs, and verified 1q21.2–q22 as a minimal commonly gained region [33]. To determine whether primary material shows a similar correlation in this study, we carried out a cytogenetic investigation on three EBV− BL patient samples. Additionally, we performed miRNA and gene expression analyses in BL-CLs to elucidate specific patterns depending on 1q gains and EBV characterizing BL in detail.
In all EBV− BL patient samples, we observed 1q gains in the form of intra-chromosomal duplication of 1q, translocation of the whole 1q arm and isodicentric chromosome 1 idic(1)(q10), resulting in partial tetrasomy of the 1q arm. While isodicentric chromosome idic(1)(p12) [12] and isochromosome i(1)(q10) [13] have already been reported, this is the first report describing supernumerary idic(1)(q10) in BL. Isochromosomes implicate the presence of a tetrasomy of the involved chromosome arm and imply the dysregulated expression of (proto-)oncogenes encoded by the affected chromosome arm [12,13]. Moreover, patient-1 displayed the concomitant presence of two cytogenetic clones bearing a duplication and an inverted duplication of the same 1q region. This is, to the best of our knowledge, the first report that shows that chromosomal duplications and inversions are temporarily separated events and that, at least in this case, the inversion followed the duplication of the 1q material.
The long arm of chromosome 1 is prone to breaks in cancer and 1q gains involve large regions covering nearly the whole 1q arm [2,33,38]. In our study, the proximal border of the duplicated 1q region in EBV− patients was mapped to the pericentric heterochromatic region 1q12 and 1q21.2 and the telomeric breakpoints were clustered in chromosomal band 1q32 and 1qter, indicating 1q21–2-1q32 as the minimal commonly gained region, which covers the minimal commonly gained region 1q21.2–q22 assigned in BL-CLs [33]. In contrast to the heterogeneous distribution of the distal breakpoints, we observed an accumulation of 1q gains with a centromeric breakpoint at the constitutive heterochromatin band 1q12 in both BL-CLs [33] and primary material indicating 1q12 as a non-random target of rearrangements in BL. The recurrence of breakpoints involving the pericentric heterochromatin band 1q12 was also observed in other NHL entities [2,3,39,40,41,42], pointing to 1q12 as an instability region that may contribute to lymphomagenesis. The juxtaposition of euchromatin regions to the constitutive heterochromatin might lead to the repression of gene(s) adjacent to the breakpoint. Thus, an excess of heterochromatin may result in the silencing of target genes by the formation of aberrant transcriptionally silent domains [41,43]. Heterochromatin region 1q12 tends to be situated close to the membrane of the nucleus and this perinuclear localization of heterochromatin is associated with transcriptional silencing [43,44]. Nevertheless, the peripheral localization of 1q12 heterochromatin is disrupted in lymphoma cells harboring 1q12 rearrangements; therefore, the information required for gene silencing might be lost in tumor cells [43]. Hence, it is highly suggestive to think that the loss of the silencing mechanisms of target genes in BL with 1q12 alterations could be potentiated by the gain of 1q regions harboring those target genes. In fact, the chromosomal pericentromeric regions are often composed of duplicated low copy repeat sequences, suggesting a link between duplications and genomic instability at the pericentric heterochromatin regions of chromosomes [45].
Gains of the 1q-arm are mainly observed in EBV− BL-CLs [33,35,36], and authors have hypothesized that genetic information similar to that included in viral genome exists on 1q [36]. A gene dosage effect resulting in higher copy numbers of gene(s), potentially relevant for proliferation, has been discussed to explain the biological consequences underlying chromosomal trisomies [46]. Additionally, 1q-resident genes MDM4 and hsa-miR-181b were overexpressed following 1q gains in BL and were discussed as candidate disease drivers [47,48,49]. In this regard, commonly overrepresented regions and partial trisomies are of particular significance and can delineate subregions bearing genes that are important for pathogenicity. Therefore, we assumed that candidate genes important for BL malignancy might exist in the minimal commonly gained region 1q21.2–q22 delineated in BL-CLs [33]. This region is flanked in its centromeric margin by the clone RP11-54A4 (covers MCL1 gene) and telomeric margin by RP11-42A2 (telomeric to MLLT11). Thus, we decided to investigate the expression of BCL9, MCL1, ARNT, MLLT11, PBXIP1 and FCRL5 genes known to be involved in B-cell development and malignancies and localized or adjacent to the minimal commonly gained region 1q21.2–q22. BCL9 oncogene, a member of the Wnt signaling pathway, are aberrantly expressed in human multiple myeloma (MM) and colon carcinoma [50] and overexpressed following t(1;14)(q21;q32)/BCL9-IGH in pre-B-cell acute lymphoblastic leukemia (ALL) [51]. BCL9 is localized at 1q21.2 but above the minimal commonly gained region [33]. Accordingly, we observed neither EBV- nor 1q-gains-dependent differences in BCL9 expression. MLLT11 is involved in t(1;11)(q21;q23) and high MLLT11 expression, which is detected in pediatric acute myeloid leukemia (AML), is associated with poor outcomes [52]. MCL1 is a member of the BCL2 family of anti-apoptotic, prosurvival proteins and is frequently overexpressed in high-grade NHL lymphomas and MM [53,54,55]. Furthermore, MYC-driven lymphomas are highly dependent on MCL1 for survival [56]. In addition, MCL1 is induced after the EBV transformation of B-cells [57,58]. FCRL5, a B-cell membrane protein involved in t(1;14)(q21;q32), is upregulated in BL-CLs with 1q21 abnormalities and is induced by EBV [59,60]. PBXIP1, involved in organogenesis and tumorigenesis, is overexpressed in various tumors [61,62]. ARNT is involved in xenobiotic metabolism and hypoxia responses and stimulates angiogenesis in different malignancies [63]. Increased ARNT expression is associated with drug resistance in high-risk MM [64] and t(1;12)(q21;p13)/TEL(ETV6)-ARNT, resulting in an impaired transcriptional response and hematopoietic cellular differentiation, is associated with AML [65,66]. The involvement of BPXIP1 or ARNT in BL or an association with EBV has not yet been reported. Here, we confirmed the EBV dependence of MCL1 and FCRL5. Our results show that MCL1 and PBXIP1 increased in 1q-gained EBV+ and MLLT11 and ARNT increased in EBV− BL-CLs with 1q gains. It indicates a dosage-dependent dysregulation of genes located at the gained 1q bands in BL, strengthening its role in the pathogenesis of BL with 1q gains.
One of the mechanisms used to control gene expression includes post-transcriptional regulation by miRNAs. Expression of miRNAs in BL was investigated in different experimental approaches. However, a relation to EBV and 1q gains has not yet been reported. Here, we investigated miRNA expression in MYC-Ig translocation-positive BL-CLs and observed 64 miRNAs that were EBV- and 1q-gains-dependent dysregulated in BL-CLs: 21 of them were known to be dysregulated in BL, 10 miRNAs were not delineated yet and the remaining 33, were described for the first time in this paper to be dysregulated in BL. However, these 33 miRNAs were found to be dysregulated in other cancer entities (s. references in Supplemental Table S5).
The most extensively investigated oncomiR hsa-miR-17-92 cluster, involved in B-cell development, is dysregulated in different cancer entities and EBV-infected B-cells and is known to be under the transcriptional control of MYC. It is also differentially regulated in MYC-driven cancers, among others in BL [25,31,32,37,67,68,69,70]. We confirmed its EBV dependence and observed the hsa-miR-17-92 cluster members and their paralogues upregulated in EBV+ BL-CLs with 1q gains, indicating an additive regulatory effect caused by 1q gains in the presence of EBV. Additionally, MYC was shown to stimulate hsa-miR-9 and repress hsa-miR-15a, hsa-miR-22, hsa-miR-23a, hsa-miR-100/let-7a-2/hsa-miR-125b-1 clusters and hsa-miR-28. Altogether, they are involved in hematopoiesis, proliferation, apoptosis, cell cycle control and survival [68,71,72,73,74]. Our data indicate that these MYC-regulated miRNAs are additionally affected by EBV and 1q gains in BL and, in case of hsa-miR-28-5p, a possible regulation independent of EBV and 1q gains.
Dysregulation of some miRNAs was directly linked to frequent chromosomal alterations involving their chromosomal loci and increased hsa-miR-9 expression was associated with the presence of 1q gains [75]. In this regard, 5 of the 64 miRNAs observed as dysregulated in our study are localized to 1q, namely hsa-miR-9, hsa-miR-9* and hsa-miR-92b at 1q22 and hsa-miR-181a and hsa-miR-181b at 1q32.1. Thus, the upregulation of these 1q resident miRNAs and four 1q resident genes (MCL1, PBXIP1, MLLT11 and ARNT) in BL-CLs with 1q gains confirms previous results and supports the assumption of a copy-number effect associated with these chromosomal aberrations [75].
DNA methyltransferases, responsible for the methylation of the CpG islands, are involved in gene expression and are repressed by hsa-miR-148a [72]. Hsa-miR-1275, a key regulator of apoptosis, autophagy and different signaling pathways, is involved in the development of different cancer types [76]. Hsa-miR-222/-221 clusters, involved in cell cycle control, are frequently upregulated in cancer and are induced by EBV [77,78,79]. Additionally, a strong relation between EBV presence and hsa-miR-21 and hsa-miR-155 expression was confirmed [80,81]. Both miRNAs are involved in hematopoiesis and are upregulated in cancer [25]. In another study, hsa-miR-155 was upregulated and the hsa-miR-183-96-182 cluster was downregulated in EBV+ BL-CLs [82]. Compared to GC B-cells, hsa-miR-182-5p, hsa-miR-183-5p and hsa-miR-148a-3p were upregulated and hsa-miR-28-5p, hsa-miR-222-3p, hsa-miR-221-3p and hsa-miR-1275 were downregulated in endemic BL tumor cells [31]. We confirmed the EBV dependence of hsa-miR-21, hsa-miR-155/-155*, hsa-miR-183-96-182 clusters and hsa-miR-222/-221 clusters, whereas our data indicate a different control mechanism involving hsa-miR-148a, the hsa-miR-183-96-182 family member and hsa-miR-1275 associated with 1q gains in EBV− and EBV+ BL. A different cellular origin within the BL subtypes is hypothesized. While EBV+ BL was assumed to originate from late-GC or memory (post-GC) B-cells, EBV− (mainly sporadic) BL appears to develop from early centroblasts [83,84]. Thus, the use of different experimental designs, cancer entities and control cell lines/tissues may cause the above-mentioned expression differences regarding these miRNAs observed in our study and previous studies.
In summary, we were able to confirm the already-known EBV dependence of some miRNAs in BL-CLs and achieved additional information taking 1q gains and EBV status into account. We observed five 1q resident miRNAs (hsa-miR-9, hsa-miR-9*, hsa-miR-92b, hsa-miR-181a and hsa-miR-181b) that were upregulated in BL-CLs with 1q gains, three miRNAs (hsa-miR-181a, hsa-miR-181b and hsa-miR-183) upregulated in BL-CLs with 1q gains, independent of their EBV status, and five miRNAs (hsa-miR-21, hsa-miR-155, hsa-miR-155*, hsa-miR-221, and hsa-miR-222) upregulated in EBV+ BL-CLs, independent of their 1q gain status. Altogether, our data indicate EBV- and 1q-gains- dependent concerted action of 64 miRNAs in BL-CLs, which may contribute to the EBV− and EBV+ BL malignancies. The miRNAs, here observed as dysregulated, are involved in proliferation, invasion, migration, cell cycle control, apoptosis and epigenetic regulation like histone-modifications or promoter methylation. Their dysregulation in cancer provides tumor cells with an attractive environment, benefits and unhampered growth. It will be challenging to elucidate the functional relevance of the individual miRNAs observed as dysregulated in our study.

5. Conclusions

We cytogenetically investigated EBV− BL primary material and analyzed EBV− and EBV+ BL-CLs in order to find 1q-gains-dependent differences in gene and miRNA expression. We observed an accumulation of 1q gains in EBV− BL cases and EBV- and 1q-gains-dependent specific gene and miRNA expression patterns involving five 1q resident mRNAs and 64 miRNAs in BL-CLs. Altogether, our data indicate a non-random involvement of 1q gains in BL and contribute to a better understanding of EBV− and EBV+ BL pathogenesis. Future investigations should be focused on primary material to confirm the effects of 1q gains in gene and miRNA expression, as well as on the influence of the condensed heterochromatin 1q12 band in the pathogenesis of BL. These would enlighten the molecular genetic differences in EBV− and EBV+ BL and provide new markers to develop novel, precise and personalized therapeutic approaches to improve diagnosis and prognosis of BL.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/v15091808/s1, “Supplemental_data_Akyuz_2023”. Table S1: List of the FISH probes used in this study; Table S2: List of the qPCR primers used in this study; Table S3: Summary of the significantly dysregulated miRNA regarding their EBV status; Table S4: Summary of the significantly dysregulated miRNA regarding their 1q gain status; Table S5: Expression profiles of the significantly dysregulated miRNAs in different cancer entities used in our study. Please note that the references [85,86]-are cited only in the “Supplementary Materials” section and [31,32,70,71,74,80,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205] only in the supplementary Table S5. The reference [34] is cited both in the main text and in the “Supplementary Materials” section. The references [25,31,32,69,70,71,72,74,80,82] are cited both in the main text and in the supplementary Table S5.

Author Contributions

Conceptualization, J.D.; methodology, J.D.; validation, investigation, N.A.; resources, J.D. and C.B.; data curation, N.A.; writing—original draft preparation, N.A. and S.J.; writing—review and editing, N.A., S.J., S.G., C.B. and J.D.; visualization, N.A.; supervision, J.D.; project administration, J.D.; funding acquisition, J.D. and C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the grant 2004.034.1 (J.D.) from the Wilhelm Sander Foundation.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki (PV4767 (16 September 2014)).

Informed Consent Statement

Patients’ specimens were collected in the diagnostic units for haematological malignancies in the context of routine diagnostics and only surplus material was analyzed for the study. Informed consent was obtained from the patients in accordance to the local laws approved by the local ethic committees for collecting and archiving their surplus material for further experimental studies in the participating institutes.

Data Availability Statement

Not applicable.

Acknowledgments

We want to thank Marianne Klokow, Bettina Keller, Petra Behrmann and Anna Schellenberg for their excellent technical assistance. Furthermore, we want to thank Eva Maria Murga Penas for providing the results. We are grateful to our patients for their valuable donation for scientific purposes.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Johansson, B.; Brondum-Nielsen, K.; Billstrom, R.; Schiodt, I.; Mitelman, F. Translocations between the long arms of chromosomes 1 and 5 in hematologic malignancies are strongly associated with neoplasms of the myeloid lineages. Cancer Genet. Cytogenet. 1997, 99, 97–101. [Google Scholar] [CrossRef] [PubMed]
  2. Itoyama, T.; Nanjungud, G.; Chen, W.; Dyomin, V.G.; Teruya-Feldstein, J.; Jhanwar, S.C.; Zelenetz, A.D.; Chaganti, R.S. Molecular cytogenetic analysis of genomic instability at the 1q12-22 chromosomal site in B-cell non-Hodgkin lymphoma. Genes Chromosomes Cancer 2002, 35, 318–328. [Google Scholar] [CrossRef] [PubMed]
  3. Sawyer, J.R.; Tricot, G.; Lukacs, J.L.; Binz, R.L.; Tian, E.; Barlogie, B.; Shaughnessy, J., Jr. Genomic instability in multiple myeloma: Evidence for jumping segmental duplications of chromosome arm 1q. Genes Chromosomes Cancer 2005, 42, 95–106. [Google Scholar] [CrossRef] [PubMed]
  4. Gunnarsson, R.; Dilorenzo, S.; Lundin-Strom, K.B.; Olsson, L.; Biloglav, A.; Lilljebjorn, H.; Rissler, M.; Wahlberg, P.; Lundmark, A.; Castor, A.; et al. Mutation, methylation, and gene expression profiles in dup(1q)-positive pediatric B-cell precursor acute lymphoblastic leukemia. Leukemia 2018, 32, 2117–2125. [Google Scholar] [CrossRef]
  5. Offit, K.; Jhanwar, S.C.; Ladanyi, M.; Filippa, D.A.; Chaganti, R.S. Cytogenetic analysis of 434 consecutively ascertained specimens of non-Hodgkin’s lymphoma: Correlations between recurrent aberrations, histology, and exposure to cytotoxic treatment. Genes Chromosomes Cancer 1991, 3, 189–201. [Google Scholar] [CrossRef]
  6. Mitelman, F.; Johansson, B.; Mertens, F. Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer (2022). Available online: https://mitelmandatabase.isb-cgc.org (accessed on 25 August 2022).
  7. Jaffe, E.S.; Stein, H.; Vardiman, J.W. World Health Organization classification of tumours. In Tumours of Haematopoietic and Lymphoid Tissue; IARC Press: Lyon, France, 2001; pp. 135–137. [Google Scholar]
  8. Thorley-Lawson, D.A.; Allday, M.J. The curious case of the tumour virus: 50 years of Burkitt’s lymphoma. Nat. Rev. Microbiol. 2008, 6, 913–924. [Google Scholar] [CrossRef]
  9. Dalla-Favera, R.; Bregni, M.; Erikson, J.; Patterson, D.; Gallo, R.C.; Croce, C.M. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc. Natl. Acad. Sci. USA 1982, 79, 7824–7827. [Google Scholar] [CrossRef]
  10. Taub, R.; Kirsch, I.; Morton, C.; Lenoir, G.; Swan, D.; Tronick, S.; Aaronson, S.; Leder, P. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc. Natl. Acad. Sci. USA 1982, 79, 7837–7841. [Google Scholar] [CrossRef]
  11. Salaverria, I.; Zettl, A.; Bea, S.; Hartmann, E.M.; Dave, S.S.; Wright, G.W.; Boerma, E.J.; Kluin, P.M.; Ott, G.; Chan, W.C.; et al. Chromosomal alterations detected by comparative genomic hybridization in subgroups of gene expression-defined Burkitt’s lymphoma. Haematologica 2008, 93, 1327–1334. [Google Scholar] [CrossRef]
  12. Roug, A.S.; Wendtland, P.; Bendix, K.; Kjeldsen, E. Supernumerary isochromosome 1, idic(1)(p12), leading to tetrasomy 1q in Burkitt lymphoma. Cytogenet. Genome Res. 2014, 142, 7–13. [Google Scholar] [CrossRef]
  13. Rim, J.H.; Kim, H.S.; Shin, S.; Park, S.J.; Choi, J.R. Isochromosome 1q in childhood Burkitt lymphoma: The first reported case in Korea. Ann. Lab. Med. 2015, 35, 663–665. [Google Scholar] [CrossRef] [PubMed]
  14. Aukema, S.M.; Theil, L.; Rohde, M.; Bauer, B.; Bradtke, J.; Burkhardt, B.; Bonn, B.R.; Claviez, A.; Gattenlohner, S.; Makarova, O.; et al. Sequential karyotyping in Burkitt lymphoma reveals a linear clonal evolution with increase in karyotype complexity and a high frequency of recurrent secondary aberrations. Br. J. Haematol. 2015, 170, 814–825. [Google Scholar] [CrossRef] [PubMed]
  15. Bessenyei, B.; Ujfalusi, A.; Balogh, E.; Olah, E.; Szegedi, I.; Kiss, C. Jumping translocation of chromosome 1q associated with good clinical outcome in a case of Burkitt leukemia. Cancer Genet. 2011, 204, 207–210. [Google Scholar] [CrossRef] [PubMed]
  16. Douglass, E.C.; Magrath, I.T.; Lee, E.C.; Whang-Peng, J. Serial cytogenetic studies of nonendemic Burkitt’s lymphoma cell lines. J. Natl. Cancer Inst. 1980, 65, 891–895. [Google Scholar]
  17. Cigudosa, J.C.; Parsa, N.Z.; Louie, D.C.; Filippa, D.A.; Jhanwar, S.C.; Johansson, B.; Mitelman, F.; Chaganti, R.S. Cytogenetic analysis of 363 consecutively ascertained diffuse large B-cell lymphomas. Genes Chromosomes Cancer 1999, 25, 123–133. [Google Scholar] [CrossRef]
  18. Garcia, J.L.; Hernandez, J.M.; Gutierrez, N.C.; Flores, T.; Gonzalez, D.; Calasanz, M.J.; Martinez-Climent, J.A.; Piris, M.A.; Lopez-Capitan, C.; Gonzalez, M.B.; et al. Abnormalities on 1q and 7q are associated with poor outcome in sporadic Burkitt’s lymphoma. A cytogenetic and comparative genomic hybridization study. Leukemia 2003, 17, 2016–2024. [Google Scholar] [CrossRef]
  19. Lones, M.A.; Sanger, W.G.; Le Beau, M.M.; Heerema, N.A.; Sposto, R.; Perkins, S.L.; Buckley, J.; Kadin, M.E.; Kjeldsberg, C.R.; Meadows, A.; et al. Chromosome abnormalities may correlate with prognosis in Burkitt/Burkitt-like lymphomas of children and adolescents: A report from Children’s Cancer Group Study CCG-E08. J. Pediatr. Hematol./Oncol. 2004, 26, 169–178. [Google Scholar] [CrossRef]
  20. Onciu, M.; Schlette, E.; Zhou, Y.; Raimondi, S.C.; Giles, F.J.; Kantarjian, H.M.; Medeiros, L.J.; Ribeiro, R.C.; Pui, C.H.; Sandlund, J.T. Secondary chromosomal abnormalities predict outcome in pediatric and adult high-stage Burkitt lymphoma. Cancer 2006, 107, 1084–1092. [Google Scholar] [CrossRef]
  21. Poirel, H.A.; Cairo, M.S.; Heerema, N.A.; Swansbury, J.; Auperin, A.; Launay, E.; Sanger, W.G.; Talley, P.; Perkins, S.L.; Raphael, M.; et al. Specific cytogenetic abnormalities are associated with a significantly inferior outcome in children and adolescents with mature B-cell non-Hodgkin’s lymphoma: Results of the FAB/LMB 96 international study. Leukemia 2009, 23, 323–331. [Google Scholar] [CrossRef]
  22. Place, R.F.; Li, L.C.; Pookot, D.; Noonan, E.J.; Dahiya, R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc. Natl. Acad. Sci. USA 2008, 105, 1608–1613. [Google Scholar] [CrossRef]
  23. Filipowicz, W.; Bhattacharyya, S.N.; Sonenberg, N. Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat. Rev. Genet. 2008, 9, 102–114. [Google Scholar] [CrossRef] [PubMed]
  24. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed]
  25. Weiss, C.N.; Ito, K. A Macro View of MicroRNAs: The Discovery of MicroRNAs and Their Role in Hematopoiesis and Hematologic Disease. Int. Rev. Cell Mol. Biol. 2017, 334, 99–175. [Google Scholar] [PubMed]
  26. Calin, G.A.; Croce, C.M. MicroRNA signatures in human cancers. Nature reviews. Cancer 2006, 6, 857–866. [Google Scholar]
  27. Calin, G.A.; Ferracin, M.; Cimmino, A.; Di Leva, G.; Shimizu, M.; Wojcik, S.E.; Iorio, M.V.; Visone, R.; Sever, N.I.; Fabbri, M.; et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N. Engl. J. Med. 2005, 353, 1793–1801. [Google Scholar] [CrossRef]
  28. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef]
  29. Di Lisio, L.; Martinez, N.; Montes-Moreno, S.; Piris-Villaespesa, M.; Sanchez-Beato, M.; Piris, M.A. The role of miRNAs in the pathogenesis and diagnosis of B-cell lymphomas. Blood 2012, 120, 1782–1790. [Google Scholar] [CrossRef]
  30. Di Lisio, L.; Sánchez-Beato, M.; Gómez-López, G.; Rodríguez, M.E.; Montes-Moreno, S.; Mollejo, M.; Menárguez, J.; Martínez, M.A.; Alves, F.J.; Pisano, D.G.; et al. MicroRNA signatures in B-cell lymphomas. Blood Cancer J. 2012, 2, e57. [Google Scholar] [CrossRef]
  31. Oduor, C.I.; Kaymaz, Y.; Chelimo, K.; Otieno, J.A.; Ong’echa, J.M.; Moormann, A.M.; Bailey, J.A. Integrative microRNA and mRNA deep-sequencing expression profiling in endemic Burkitt lymphoma. BMC Cancer 2017, 17, 761. [Google Scholar] [CrossRef]
  32. Robaina, M.C.; Faccion, R.S.; Mazzoccoli, L.; Rezende, L.M.; Queiroga, E.; Bacchi, C.E.; Thomas-Tikhonenko, A.; Klumb, C.E. miR-17-92 cluster components analysis in Burkitt lymphoma: Overexpression of miR-17 is associated with poor prognosis. Ann. Hematol. 2016, 95, 881–891. [Google Scholar] [CrossRef]
  33. Murga Penas, E.M.; Schilling, G.; Behrmann, P.; Klokow, M.; Vettorazzi, E.; Bokemeyer, C.; Dierlamm, J. Comprehensive cytogenetic and molecular cytogenetic analysis of 44 Burkitt lymphoma cell lines: Secondary chromosomal changes characterization, karyotypic evolution, and comparison with primary samples. Genes Chromosomes Cancer 2014, 53, 497–515. [Google Scholar] [CrossRef] [PubMed]
  34. McGowan-Jordan, J.H.R.; Moore, S. (Eds.) ISCN 2020 An International System for Human Cytogenomic Nomenclature (2020); Karger: Basel, Switzerland, 2020. [Google Scholar]
  35. Sobol, H.; Benziane, A.; Kerangueven, F.; Yin, L.; Noguchi, T.; Pauly, S.; Eisinger, F.; Longy, M.; Romeo, G.; Lenoir, G.; et al. Genome-wide search for loss of heterozygosity in Burkitt lymphoma cell lines. Genes Chromosomes Cancer 2002, 33, 217–224. [Google Scholar] [CrossRef] [PubMed]
  36. Berger, R.; Bernheim, A. [Is there a functional equivalence between abnormalities of the long arm of chromosome 1 and the presence of Epstein-Barr virus in continuous lines of Burkitt’s lymphoma?]. Comptes Rendus L’academie Sci. Ser. III Sci. Vie 1984, 298, 143–145. [Google Scholar]
  37. O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843. [Google Scholar] [CrossRef]
  38. Davidsson, J.; Andersson, A.; Paulsson, K.; Heidenblad, M.; Isaksson, M.; Borg, A.; Heldrup, J.; Behrendtz, M.; Panagopoulos, I.; Fioretos, T.; et al. Tiling resolution array comparative genomic hybridization, expression and methylation analyses of dup(1q) in Burkitt lymphomas and pediatric high hyperdiploid acute lymphoblastic leukemias reveal clustered near-centromeric breakpoints and overexpression of genes in 1q22-32.3. Hum. Mol. Genet. 2007, 16, 2215–2225. [Google Scholar]
  39. Dierlamm, J.; Wlodarska, I.; Michaux, L.; Vermeesch, J.R.; Meeus, P.; Stul, M.; Criel, A.; Verhoef, G.; Thomas, J.; Delannoy, A.; et al. FISH identifies different types of duplications with 12q13-15 as the commonly involved segment in B-cell lymphoproliferative malignancies characterized by partial trisomy 12. Genes Chromosomes Cancer 1997, 20, 155–166. [Google Scholar] [CrossRef]
  40. Busson-Le Coniat, M.; Salomon-Nguyen, F.; Dastugue, N.; Maarek, O.; Lafage-Pochitaloff, M.; Mozziconacci, M.J.; Baranger, L.; Brizard, F.; Radford, I.; Jeanpierre, M.; et al. Fluorescence in situ hybridization analysis of chromosome 1 abnormalities in hematopoietic disorders: Rearrangements of DNA satellite II and new recurrent translocations. Leukemia 1999, 13, 1975–1981. [Google Scholar] [CrossRef]
  41. Le Baccon, P.; Leroux, D.; Dascalescu, C.; Duley, S.; Marais, D.; Esmenjaud, E.; Sotto, J.J.; Callanan, M. Novel evidence of a role for chromosome 1 pericentric heterochromatin in the pathogenesis of B-cell lymphoma and multiple myeloma. Genes Chromosomes Cancer 2001, 32, 250–264. [Google Scholar] [CrossRef]
  42. Lestou, V.S.; Gascoyne, R.D.; Salski, C.; Connors, J.M.; Horsman, D.E. Uncovering novel inter- and intrachromosomal chromosome 1 aberrations in follicular lymphomas by using an innovative multicolor banding technique. Genes Chromosomes Cancer 2002, 34, 201–210. [Google Scholar] [CrossRef]
  43. Barki-Celli, L.; Lefebvre, C.; Le Baccon, P.; Nadeau, G.; Bonnefoix, T.; Usson, Y.; Vourc’h, C.; Khochbin, S.; Leroux, D.; Callanan, M. Differences in nuclear positioning of 1q12 pericentric heterochromatin in normal and tumor B lymphocytes with 1q rearrangements. Genes Chromosomes Cancer 2005, 43, 339–349. [Google Scholar] [CrossRef]
  44. Andrulis, E.D.; Neiman, A.M.; Zappulla, D.C.; Sternglanz, R. Perinuclear localization of chromatin facilitates transcriptional silencing. Nature 1998, 394, 592–595. [Google Scholar] [CrossRef]
  45. Horvath, J.E.; Bailey, J.A.; Locke, D.P.; Eichler, E.E. Lessons from the human genome: Transitions between euchromatin and heterochromatin. Hum. Mol. Genet. 2001, 10, 2215–2223. [Google Scholar] [CrossRef] [PubMed]
  46. Dürrbaum, M.; Storchová, Z. Effects of aneuploidy on gene expression: Implications for cancer. FEBS J. 2016, 283, 791–802. [Google Scholar] [CrossRef] [PubMed]
  47. Toujani, S.; Dessen, P.; Ithzar, N.; Danglot, G.; Richon, C.; Vassetzky, Y.; Robert, T.; Lazar, V.; Bosq, J.; Da Costa, L.; et al. High resolution genome-wide analysis of chromosomal alterations in Burkitt’s lymphoma. PLoS ONE 2009, 4, e7089. [Google Scholar] [CrossRef] [PubMed]
  48. Scholtysik, R.; Kreuz, M.; Klapper, W.; Burkhardt, B.; Feller, A.C.; Hummel, M.; Loeffler, M.; Rosolowski, M.; Schwaenen, C.; Spang, R.; et al. Detection of genomic aberrations in molecularly defined Burkitt’s lymphoma by array-based, high resolution, single nucleotide polymorphism analysis. Haematologica 2010, 95, 2047–2055. [Google Scholar] [CrossRef] [PubMed]
  49. Hullein, J.; Slabicki, M.; Rosolowski, M.; Jethwa, A.; Habringer, S.; Tomska, K.; Kurilov, R.; Lu, J.; Scheinost, S.; Wagener, R.; et al. MDM4 Is Targeted by 1q Gain and Drives Disease in Burkitt Lymphoma. Cancer Res. 2019, 79, 3125–3138. [Google Scholar] [CrossRef] [PubMed]
  50. Mani, M.; Carrasco, D.E.; Zhang, Y.; Takada, K.; Gatt, M.E.; Dutta-Simmons, J.; Ikeda, H.; Diaz-Griffero, F.; Pena-Cruz, V.; Bertagnolli, M.; et al. BCL9 promotes tumor progression by conferring enhanced proliferative, metastatic, and angiogenic properties to cancer cells. Cancer Res. 2009, 69, 7577–7586. [Google Scholar] [CrossRef]
  51. Willis, T.G.; Zalcberg, I.R.; Coignet, L.J.; Wlodarska, I.; Stul, M.; Jadayel, D.M.; Bastard, C.; Treleaven, J.G.; Catovsky, D.; Silva, M.L.; et al. Molecular cloning of translocation t(1;14)(q21;q32) defines a novel gene (BCL9) at chromosome 1q21. Blood 1998, 91, 1873–1881. [Google Scholar] [CrossRef]
  52. Tse, W.; Meshinchi, S.; Alonzo, T.A.; Stirewalt, D.L.; Gerbing, R.B.; Woods, W.G.; Appelbaum, F.R.; Radich, J.P. Elevated expression of the AF1q gene, an MLL fusion partner, is an independent adverse prognostic factor in pediatric acute myeloid leukemia. Blood 2004, 104, 3058–3063. [Google Scholar] [CrossRef]
  53. Kozopas, K.M.; Yang, T.; Buchan, H.L.; Zhou, P.; Craig, R.W. MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2. Proc. Natl. Acad. Sci. USA 1993, 90, 3516–3520. [Google Scholar] [CrossRef]
  54. Cho-Vega, J.H.; Rassidakis, G.Z.; Admirand, J.H.; Oyarzo, M.; Ramalingam, P.; Paraguya, A.; McDonnell, T.J.; Amin, H.M.; Medeiros, L.J. MCL-1 expression in B-cell non-Hodgkin’s lymphomas. Hum. Pathol. 2004, 35, 1095–1100. [Google Scholar] [CrossRef]
  55. Michels, J.; O’Neill, J.W.; Dallman, C.L.; Mouzakiti, A.; Habens, F.; Brimmell, M.; Zhang, K.Y.; Craig, R.W.; Marcusson, E.G.; Johnson, P.W.; et al. Mcl-1 is required for Akata6 B-lymphoma cell survival and is converted to a cell death molecule by efficient caspase-mediated cleavage. Oncogene 2004, 23, 4818–4827. [Google Scholar] [CrossRef]
  56. Kelly, G.L.; Grabow, S.; Glaser, S.P.; Fitzsimmons, L.; Aubrey, B.J.; Okamoto, T.; Valente, L.J.; Robati, M.; Tai, L.; Fairlie, W.D.; et al. Targeting of MCL-1 kills MYC-driven mouse and human lymphomas even when they bear mutations in p53. Genes Dev. 2014, 28, 58–70. [Google Scholar] [CrossRef]
  57. Wang, S.; Rowe, M.; Lundgren, E. Expression of the Epstein Barr virus transforming protein LMP1 causes a rapid and transient stimulation of the Bcl-2 homologue Mcl-1 levels in B-cell lines. Cancer Res. 1996, 56, 4610–4613. [Google Scholar]
  58. Kenney, J.L.; Guinness, M.E.; Curiel, T.; Lacy, J. Antisense to the epstein-barr virus (EBV)-encoded latent membrane protein 1 (LMP-1) suppresses LMP-1 and bcl-2 expression and promotes apoptosis in EBV-immortalized B cells. Blood 1998, 92, 1721–1727. [Google Scholar] [CrossRef]
  59. Mohan, J.; Dement-Brown, J.; Maier, S.; Ise, T.; Kempkes, B.; Tolnay, M. Epstein-Barr virus nuclear antigen 2 induces FcRH5 expression through CBF1. Blood 2006, 107, 4433–4439. [Google Scholar] [CrossRef]
  60. Hatzivassiliou, G.; Miller, I.; Takizawa, J.; Palanisamy, N.; Rao, P.H.; Iida, S.; Tagawa, S.; Taniwaki, M.; Russo, J.; Neri, A.; et al. IRTA1 and IRTA2, novel immunoglobulin superfamily receptors expressed in B cells and involved in chromosome 1q21 abnormalities in B cell malignancy. Immunity 2001, 14, 277–289. [Google Scholar] [CrossRef]
  61. Zhang, G.Y.; Liu, A.H.; Li, G.M.; Wang, J.R. HPIP Silencing Prevents Epithelial-Mesenchymal Transition Induced by TGF-beta1 in Human Ovarian Cancer Cells. Oncol. Res. 2016, 24, 33–39. [Google Scholar] [CrossRef]
  62. Shi, S.; Zhao, J.; Wang, J.; Mi, D.; Ma, Z. HPIP silencing inhibits TGF-beta1-induced EMT in lung cancer cells. Int. J. Mol. Med. 2017, 39, 479–483. [Google Scholar] [CrossRef]
  63. Vorrink, S.U.; Domann, F.E. Regulatory crosstalk and interference between the xenobiotic and hypoxia sensing pathways at the AhR-ARNT-HIF1alpha signaling node. Chem. Biol. Interact. 2014, 218, 82–88. [Google Scholar] [CrossRef]
  64. Wu, C.; Yang, T.; Liu, Y.; Lu, Y.; Yang, Y.; Liu, X.; Liu, X.; Ye, L.; Sun, Y.; Wang, X.; et al. ARNT/HIF-1beta links high-risk 1q21 gain and microenvironmental hypoxia to drug resistance and poor prognosis in multiple myeloma. Cancer Med. 2018, 7, 3899–3911. [Google Scholar] [CrossRef] [PubMed]
  65. Salomon-Nguyen, F.; Della-Valle, V.; Mauchauffe, M.; Busson-Le Coniat, M.; Ghysdael, J.; Berger, R.; Bernard, O.A. The t(1;12)(q21;p13) translocation of human acute myeloblastic leukemia results in a TEL-ARNT fusion. Proc. Natl. Acad. Sci. USA 2000, 97, 6757–6762. [Google Scholar] [CrossRef] [PubMed]
  66. Nguyen-Khac, F.; Della Valle, V.; Lopez, R.G.; Ravet, E.; Mauchauffe, M.; Friedman, A.D.; Huang, L.E.; Fichelson, S.; Ghysdael, J.; Bernard, O.A. Functional analyses of the TEL-ARNT fusion protein underscores a role for oxygen tension in hematopoietic cellular differentiation. Oncogene 2006, 25, 4840–4847. [Google Scholar] [CrossRef]
  67. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: A comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef]
  68. Chang, T.C.; Yu, D.; Lee, Y.S.; Wentzel, E.A.; Arking, D.E.; West, K.M.; Dang, C.V.; Thomas-Tikhonenko, A.; Mendell, J.T. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat. Genet. 2008, 40, 43–50. [Google Scholar] [CrossRef] [PubMed]
  69. Onnis, A.; De Falco, G.; Antonicelli, G.; Onorati, M.; Bellan, C.; Sherman, O.; Sayed, S.; Leoncini, L. Alteration of microRNAs regulated by c-Myc in Burkitt lymphoma. PLoS ONE 2010, 5, e12960. [Google Scholar] [CrossRef]
  70. Cao, P.; Zhang, M.; Wang, L.; Sai, B.; Tang, J.; Luo, Z.; Shuai, C.; Zhang, L.; Li, Z.; Wang, Y.; et al. miR-18a reactivates the Epstein-Barr virus through defective DNA damage response and promotes genomic instability in EBV-associated lymphomas. BMC Cancer 2018, 18, 1293. [Google Scholar] [CrossRef]
  71. Zhang, X.; Chen, X.; Lin, J.; Lwin, T.; Wright, G.; Moscinski, L.C.; Dalton, W.S.; Seto, E.; Wright, K.; Sotomayor, E.; et al. Myc represses miR-15a/miR-16-1 expression through recruitment of HDAC3 in mantle cell and other non-Hodgkin B-cell lymphomas. Oncogene 2012, 31, 3002–3008. [Google Scholar] [CrossRef]
  72. Croce, C.M. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009, 10, 704–714. [Google Scholar] [CrossRef]
  73. Psathas, J.N.; Thomas-Tikhonenko, A. MYC and the art of microRNA maintenance. Cold Spring Harb. Perspect. Med. 2014, 4, a014175. [Google Scholar] [CrossRef]
  74. Schneider, C.; Setty, M.; Holmes, A.B.; Maute, R.L.; Leslie, C.S.; Mussolin, L.; Rosolen, A.; Dalla-Favera, R.; Basso, K. MicroRNA 28 controls cell proliferation and is down-regulated in B-cell lymphomas. Proc. Natl. Acad. Sci. USA 2014, 111, 8185–8190. [Google Scholar] [CrossRef]
  75. Wilting, S.M.; Snijders, P.J.; Verlaat, W.; Jaspers, A.; van de Wiel, M.A.; van Wieringen, W.N.; Meijer, G.A.; Kenter, G.G.; Yi, Y.; le Sage, C.; et al. Altered microRNA expression associated with chromosomal changes contributes to cervical carcinogenesis. Oncogene 2013, 32, 106–116. [Google Scholar] [CrossRef]
  76. Chong, Z.X.; Yeap, S.K.; Ho, W.Y.; Fang, C.M. Unveiling the tumour-regulatory roles of miR-1275 in cancer. Pathol. Res. Pract. 2022, 230, 153745. [Google Scholar] [CrossRef]
  77. Galardi, S.; Mercatelli, N.; Giorda, E.; Massalini, S.; Frajese, G.V.; Ciafrè, S.A.; Farace, M.G. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J. Biol. Chem. 2007, 282, 23716–23724. [Google Scholar] [CrossRef]
  78. Bazot, Q.; Paschos, K.; Skalska, L.; Kalchschmidt, J.S.; Parker, G.A.; Allday, M.J. Epstein-Barr Virus Proteins EBNA3A and EBNA3C Together Induce Expression of the Oncogenic MicroRNA Cluster miR-221/miR-222 and Ablate Expression of Its Target p57KIP2. PLoS Pathog. 2015, 11, e1005031. [Google Scholar] [CrossRef] [PubMed]
  79. Andrade, T.A.; Evangelista, A.F.; Campos, A.H.; Poles, W.A.; Borges, N.M.; Camillo, C.M.; Soares, F.A.; Vassallo, J.; Paes, R.P.; Zerbini, M.C.; et al. A microRNA signature profile in EBV+ diffuse large B-cell lymphoma of the elderly. Oncotarget 2014, 5, 11813–11826. [Google Scholar] [CrossRef] [PubMed]
  80. Rosato, P.; Anastasiadou, E.; Garg, N.; Lenze, D.; Boccellato, F.; Vincenti, S.; Severa, M.; Coccia, E.M.; Bigi, R.; Cirone, M.; et al. Differential regulation of miR-21 and miR-146a by Epstein-Barr virus-encoded EBNA2. Leukemia 2012, 26, 2343–2352. [Google Scholar] [CrossRef] [PubMed]
  81. Gibcus, J.H.; Tan, L.P.; Harms, G.; Schakel, R.N.; de Jong, D.; Blokzijl, T.; Möller, P.; Poppema, S.; Kroesen, B.J.; van den Berg, A. Hodgkin lymphoma cell lines are characterized by a specific miRNA expression profile. Neoplasia 2009, 11, 167–176. [Google Scholar] [CrossRef]
  82. Oussaief, L.; Fendri, A.; Chane-Woon-Ming, B.; Poirey, R.; Delecluse, H.J.; Joab, I.; Pfeffer, S. Modulation of MicroRNA Cluster miR-183-96-182 Expression by Epstein-Barr Virus Latent Membrane Protein 1. J. Virol. 2015, 89, 12178–12188. [Google Scholar] [CrossRef]
  83. Bellan, C.; Lazzi, S.; Hummel, M.; Palummo, N.; de Santi, M.; Amato, T.; Nyagol, J.; Sabattini, E.; Lazure, T.; Pileri, S.A.; et al. Immunoglobulin gene analysis reveals 2 distinct cells of origin for EBV-positive and EBV-negative Burkitt lymphomas. Blood 2005, 106, 1031–1036. [Google Scholar] [CrossRef]
  84. Amato, T.; Abate, F.; Piccaluga, P.; Iacono, M.; Fallerini, C.; Renieri, A.; De Falco, G.; Ambrosio, M.R.; Mourmouras, V.; Ogwang, M.; et al. Clonality Analysis of Immunoglobulin Gene Rearrangement by Next-Generation Sequencing in Endemic Burkitt Lymphoma Suggests Antigen Drive Activation of BCR as Opposed to Sporadic Burkitt Lymphoma. Am. J. Clin. Pathol. 2016, 145, 116–127. [Google Scholar] [CrossRef] [PubMed]
  85. Dierlamm, J.; Baens, M.; Stefanova-Ouzounova, M.; Hinz, K.; Wlodarska, I.; Maes, B.; Steyls, A.; Driessen, A.; Verhoef, G.; Gaulard, P.; et al. Detection of t(11;18)(q21;q21) by interphase fluorescence in situ hybridization using API2 and MLT specific probes. Blood J. Am. Soc. Hematol. 2000, 96, 2215–2218. [Google Scholar]
  86. Hiller, B.; Bradtke, J.; Balz, H.; Rieder, H. CyDAS Online Analysis Site. Available online: http://www.cydas.org/OnlineAnalysis (accessed on 1 January 2010).
  87. Dierlamm, J.; Wlodarska, I.; Michaux, L.; La Starza, R.; Zeller, W.; Mecucci, C.; Van den Berghe, H. Successful use of the same slide for consecutive fluorescence in situ hybridization experiments. Genes Chromosomes Cancer 1996, 16, 261–264. [Google Scholar] [CrossRef]
  88. Khosravi, A.; Alizadeh, S.; Jalili, A.; Shirzad, R.; Saki, N. The impact of Mir-9 regulation in normal and malignant hematopoiesis. Oncol. Rev. 2018, 12, 348. [Google Scholar] [CrossRef]
  89. Nowek, K.; Wiemer, E.A.C.; Jongen-Lavrencic, M. The versatile nature of miR-9/9(*) in human cancer. Oncotarget 2018, 9, 20838–20854. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, W.; Corrigan-Cummins, M.; Hudson, J.; Maric, I.; Simakova, O.; Neelapu, S.S.; Kwak, L.W.; Janik, J.E.; Gause, B.; Jaffe, E.S.; et al. MicroRNA profiling of follicular lymphoma identifies microRNAs related to cell proliferation and tumor response. Haematologica 2012, 97, 586–594. [Google Scholar] [CrossRef] [PubMed]
  91. Sun, Y.; Ju, X.L.; Li, D.; Zhou, P.P.; Li, X.; Luo, R.H. miR-1290 promotes proliferation and suppresses apoptosis in acute myeloid leukemia by targeting FOXG1/SOCS3. J. Biol. Regul. Homeost. Agents 2019, 33, 1703–1713. [Google Scholar]
  92. Zhou, W.; Wang, S.; Ying, Y.; Zhou, R.; Mao, P. miR-196b/miR-1290 participate in the antitumor effect of resveratrol via regulation of IGFBP3 expression in acute lymphoblastic leukemia. Oncol. Rep. 2017, 37, 1075–1083. [Google Scholar] [CrossRef]
  93. Hu, N.; Chen, L.; Wang, C.; Zhao, H. MALAT1 knockdown inhibits proliferation and enhances cytarabine chemosensitivity by upregulating miR-96 in acute myeloid leukemia cells. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 112, 108720. [Google Scholar] [CrossRef]
  94. Huang, T.; Fu, Y.; Wang, S.; Xu, M.; Yin, X.; Zhou, M.; Wang, X.; Chen, C. miR-96 acts as a tumor suppressor via targeting the BCR-ABL1 oncogene in chronic myeloid leukemia blastic transformation. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 119, 109413. [Google Scholar] [CrossRef]
  95. Iwai, N.; Yasui, K.; Tomie, A.; Gen, Y.; Terasaki, K.; Kitaichi, T.; Soda, T.; Yamada, N.; Dohi, O.; Seko, Y.; et al. Oncogenic miR-96-5p inhibits apoptosis by targeting the caspase-9 gene in hepatocellular carcinoma. Int. J. Oncol. 2018, 53, 237–245. [Google Scholar] [CrossRef]
  96. Xie, W.; Sun, F.; Chen, L.; Cao, X. miR-96 promotes breast cancer metastasis by suppressing MTSS1. Oncol. Lett. 2018, 15, 3464–3471. [Google Scholar] [CrossRef]
  97. Xie, L.; Ushmorov, A.; Leithäuser, F.; Guan, H.; Steidl, C.; Färbinger, J.; Pelzer, C.; Vogel, M.J.; Maier, H.J.; Gascoyne, R.D.; et al. FOXO1 is a tumor suppressor in classical Hodgkin lymphoma. Blood J. Am. Soc. Hematol. 2012, 119, 3503–3511. [Google Scholar] [CrossRef]
  98. Hu, J.; Lv, G.; Zhou, S.; Zhou, Y.; Nie, B.; Duan, H.; Zhang, Y.; Yuan, X. The Downregulation of MiR-182 Is Associated with the Growth and Invasion of Osteosarcoma Cells through the Regulation of TIAM1 Expression. PLoS ONE 2015, 10, e0121175. [Google Scholar] [CrossRef]
  99. Zhang, S.; Zhang, Q.; Shi, G.; Yin, J. MiR-182-5p regulates BCL2L12 and BCL2 expression in acute myeloid leukemia as a potential therapeutic target. Biomed. Pharmacother. Biomed. Pharmacother. 2018, 97, 1189–1194. [Google Scholar] [CrossRef]
  100. Piatopoulou, D.; Avgeris, M.; Marmarinos, A.; Xagorari, M.; Baka, M.; Doganis, D.; Kossiva, L.; Scorilas, A.; Gourgiotis, D. miR-125b predicts childhood acute lymphoblastic leukaemia poor response to BFM chemotherapy treatment. Br. J. Cancer 2017, 117, 801–812. [Google Scholar] [CrossRef]
  101. Liu, X.; Liao, W.; Peng, H.; Luo, X.; Luo, Z.; Jiang, H.; Xu, L. miR-181a promotes G1/S transition and cell proliferation in pediatric acute myeloid leukemia by targeting ATM. J. Cancer Res. Clin. Oncol. 2016, 142, 77–87. [Google Scholar] [CrossRef]
  102. Verduci, L.; Azzalin, G.; Gioiosa, S.; Carissimi, C.; Laudadio, I.; Fulci, V.; Macino, G. microRNA-181a enhances cell proliferation in acute lymphoblastic leukemia by targeting EGR1. Leuk. Res. 2015, 39, 479–485. [Google Scholar] [CrossRef]
  103. Pichiorri, F.; Suh, S.S.; Ladetto, M.; Kuehl, M.; Palumbo, T.; Drandi, D.; Taccioli, C.; Zanesi, N.; Alder, H.; Hagan, J.P.; et al. MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc. Natl. Acad. Sci. USA 2008, 105, 12885–12890. [Google Scholar] [CrossRef]
  104. Shen, H.; Weng, X.D.; Liu, X.H.; Yang, D.; Wang, L.; Guo, J.; Wang, M.; Wang, X.; Diao, C.H. miR-181a-5p is downregulated and inhibits proliferation and the cell cycle in prostate cancer. Int. J. Clin. Exp. Pathol. 2018, 11, 3969–3976. [Google Scholar]
  105. Zhou, G.; Cao, Y.; Dong, W.; Lin, Y.; Wang, Q.; Wu, W.; Hua, X.; Ling, Y.; Xie, X.; Hu, S.; et al. The clinical characteristics and prognostic significance of AID, miR-181b, and miR-155 expression in adult patients with de novo B-cell acute lymphoblastic leukemia. Leuk. Lymphoma 2017, 58, 2118–2126. [Google Scholar] [CrossRef] [PubMed]
  106. Visone, R.; Veronese, A.; Rassenti, L.Z.; Balatti, V.; Pearl, D.K.; Acunzo, M.; Volinia, S.; Taccioli, C.; Kipps, T.J.; Croce, C.M. miR-181b is a biomarker of disease progression in chronic lymphocytic leukemia. Blood J. Am. Soc. Hematol. 2011, 118, 3072–3079. [Google Scholar] [CrossRef] [PubMed]
  107. Li, J.G.; Ding, Y.; Huang, Y.M.; Chen, W.L.; Pan, L.L.; Li, Y.; Chen, X.L.; Chen, Y.; Wang, S.Y.; Wu, X.N. FAMLF is a target of miR-181b in Burkitt lymphoma. Braz. J. Med. Biol. Res. Rev. Bras. De Pesqui. Medicas E Biol. 2017, 50, e5661. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, X.; Zuo, D.; Yuan, Y.; Yang, X.; Hong, Z.; Zhang, R. MicroRNA-183 promotes cell proliferation via regulating programmed cell death 6 in pediatric acute myeloid leukemia. J. Cancer Res. Clin. Oncol. 2017, 143, 169–180. [Google Scholar] [CrossRef] [PubMed]
  109. Renou, L.; Boelle, P.Y.; Deswarte, C.; Spicuglia, S.; Benyoucef, A.; Calvo, J.; Uzan, B.; Belhocine, M.; Cieslak, A.; Landman-Parker, J.; et al. Homeobox protein TLX3 activates miR-125b expression to promote T-cell acute lymphoblastic leukemia. Blood Adv. 2017, 1, 733–747. [Google Scholar] [CrossRef] [PubMed]
  110. Zhang, H.; Luo, X.Q.; Zhang, P.; Huang, L.B.; Zheng, Y.S.; Wu, J.; Zhou, H.; Qu, L.H.; Xu, L.; Chen, Y.Q. MicroRNA patterns associated with clinical prognostic parameters and CNS relapse prediction in pediatric acute leukemia. PLoS ONE 2009, 4, e7826. [Google Scholar] [CrossRef]
  111. Tili, E.; Michaille, J.J.; Luo, Z.; Volinia, S.; Rassenti, L.Z.; Kipps, T.J.; Croce, C.M. The down-regulation of miR-125b in chronic lymphocytic leukemias leads to metabolic adaptation of cells to a transformed state. Blood J. Am. Soc. Hematol. 2012, 120, 2631–2638. [Google Scholar] [CrossRef]
  112. Dioguardi, M.; Caloro, G.A.; Laino, L.; Alovisi, M.; Sovereto, D.; Crincoli, V.; Aiuto, R.; Coccia, E.; Troiano, G.; Lo Muzio, L. Circulating miR-21 as a Potential Biomarker for the Diagnosis of Oral Cancer: A Systematic Review with Meta-Analysis. Cancers 2020, 12, 936. [Google Scholar] [CrossRef]
  113. Zhang, T.J.; Zhou, J.D.; Zhang, W.; Lin, J.; Ma, J.C.; Wen, X.M.; Yuan, Q.; Li, X.X.; Xu, Z.J.; Qian, J. H19 overexpression promotes leukemogenesis and predicts unfavorable prognosis in acute myeloid leukemia. Clin. Epigenetics 2018, 10, 47. [Google Scholar] [CrossRef]
  114. Li, Y.; Tian, Z.; Tan, Y.; Lian, G.; Chen, S.; Chen, S.; Li, J.; Li, X.; Huang, K.; Chen, Y. Bmi-1-induced miR-27a and miR-155 promote tumor metastasis and chemoresistance by targeting RKIP in gastric cancer. Mol. Cancer 2020, 19, 109. [Google Scholar] [CrossRef]
  115. Sun, Y.P.; Lu, F.; Han, X.Y.; Ji, M.; Zhou, Y.; Zhang, A.M.; Wang, H.C.; Ma, D.X.; Ji, C.Y. MiR-424 and miR-27a increase TRAIL sensitivity of acute myeloid leukemia by targeting PLAG1. Oncotarget 2016, 7, 25276–25290. [Google Scholar] [CrossRef] [PubMed]
  116. Scheibner, K.A.; Teaboldt, B.; Hauer, M.C.; Chen, X.; Cherukuri, S.; Guo, Y.; Kelley, S.M.; Liu, Z.; Baer, M.R.; Heimfeld, S.; et al. MiR-27a functions as a tumor suppressor in acute leukemia by regulating 14-3-3θ. PLoS ONE 2012, 7, e50895. [Google Scholar] [CrossRef]
  117. Rane, J.K.; Erb, H.H.; Nappo, G.; Mann, V.M.; Simms, M.S.; Collins, A.T.; Visakorpi, T.; Maitland, N.J. Inhibition of the glucocorticoid receptor results in an enhanced miR-99a/100-mediated radiation response in stem-like cells from human prostate cancers. Oncotarget 2016, 7, 51965–51980. [Google Scholar] [CrossRef]
  118. Si, X.; Zhang, X.; Hao, X.; Li, Y.; Chen, Z.; Ding, Y.; Shi, H.; Bai, J.; Gao, Y.; Cheng, T.; et al. Upregulation of miR-99a is associated with poor prognosis of acute myeloid leukemia and promotes myeloid leukemia cell expansion. Oncotarget 2016, 7, 78095–78109. [Google Scholar] [CrossRef]
  119. Li, X.J.; Luo, X.Q.; Han, B.W.; Duan, F.T.; Wei, P.P.; Chen, Y.Q. MicroRNA-100/99a, deregulated in acute lymphoblastic leukaemia, suppress proliferation and promote apoptosis by regulating the FKBP51 and IGF1R/mTOR signalling pathways. Br. J. Cancer 2013, 109, 2189–2198. [Google Scholar] [CrossRef] [PubMed]
  120. Arribas, A.J.; Gómez-Abad, C.; Sánchez-Beato, M.; Martinez, N.; Dilisio, L.; Casado, F.; Cruz, M.A.; Algara, P.; Piris, M.A.; Mollejo, M. Splenic marginal zone lymphoma: Comprehensive analysis of gene expression and miRNA profiling. Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc. 2013, 26, 889–901. [Google Scholar] [CrossRef] [PubMed]
  121. Berg, V.; Rusch, M.; Vartak, N.; Jüngst, C.; Schauss, A.; Waldmann, H.; Hedberg, C.; Pallasch, C.P.; Bastiaens, P.I.; Hallek, M.; et al. miRs-138 and -424 control palmitoylation-dependent CD95-mediated cell death by targeting acyl protein thioesterases 1 and 2 in CLL. Blood J. Am. Soc. Hematol. 2015, 125, 2948–2957. [Google Scholar] [CrossRef]
  122. Augello, C.; Gianelli, U.; Savi, F.; Moro, A.; Bonoldi, E.; Gambacorta, M.; Vaira, V.; Baldini, L.; Bosari, S. MicroRNA as potential biomarker in HCV-associated diffuse large B-cell lymphoma. J. Clin. Pathol. 2014, 67, 697–701. [Google Scholar] [CrossRef]
  123. Amaar, Y.G.; Reeves, M.E. RASSF1C regulates miR-33a and EMT marker gene expression in lung cancer cells. Oncotarget 2019, 10, 123–132. [Google Scholar] [CrossRef]
  124. Guan, X.; Gu, S.; Yuan, M.; Zheng, X.; Wu, J. MicroRNA-33a-5p overexpression sensitizes triple-negative breast cancer to doxorubicin by inhibiting eIF5A2 and epithelial-mesenchymal transition. Oncol. Lett. 2019, 18, 5986–5994. [Google Scholar] [CrossRef]
  125. Sun, X.; Liu, H.; Li, T.; Qin, L. MicroRNA-339-5p inhibits cell proliferation of acute myeloid leukaemia by directly targeting SOX4. Mol. Med. Rep. 2018, 18, 5261–5269. [Google Scholar] [CrossRef] [PubMed]
  126. Almeida, R.S.; Costa, E.S.M.; Coutinho, L.L.; Garcia Gomes, R.; Pedrosa, F.; Massaro, J.D.; Donadi, E.A.; Lucena-Silva, N. MicroRNA expression profiles discriminate childhood T- from B-acute lymphoblastic leukemia. Hematol. Oncol. 2019, 37, 103–112. [Google Scholar] [CrossRef] [PubMed]
  127. Pashaei, E.; Pashaei, E.; Ahmady, M.; Ozen, M.; Aydin, N. Meta-analysis of miRNA expression profiles for prostate cancer recurrence following radical prostatectomy. PLoS ONE 2017, 12, e0179543. [Google Scholar] [CrossRef] [PubMed]
  128. Yang, J.R.; Shi, M.X.; Zeng, Y. LncRNA HAND2-AS1 inhibits proliferation and promotes apoptosis of chronic myeloid leukemia cells by sponging with micRNA-1275. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2103–2111. [Google Scholar]
  129. Ozdogan, H.; Gur Dedeoglu, B.; Oztemur Islakoglu, Y.; Aydos, A.; Kose, S.; Atalay, A.; Yegin, Z.A.; Avcu, F.; Uckan Cetinkaya, D.; Ilhan, O. DICER1 gene and miRNA dysregulation in mesenchymal stem cells of patients with myelodysplastic syndrome and acute myeloblastic leukemia. Leuk. Res. 2017, 63, 62–71. [Google Scholar] [CrossRef]
  130. Mraz, M.; Malinova, K.; Kotaskova, J.; Pavlova, S.; Tichy, B.; Malcikova, J.; Stano Kozubik, K.; Smardova, J.; Brychtova, Y.; Doubek, M.; et al. miR-34a, miR-29c and miR-17-5p are downregulated in CLL patients with TP53 abnormalities. Leukemia 2009, 23, 1159–1163. [Google Scholar] [CrossRef]
  131. Iqbal, J.; Shen, Y.; Huang, X.; Liu, Y.; Wake, L.; Liu, C.; Deffenbacher, K.; Lachel, C.M.; Wang, C.; Rohr, J.; et al. Global microRNA expression profiling uncovers molecular markers for classification and prognosis in aggressive B-cell lymphoma. Blood J. Am. Soc. Hematol. 2015, 125, 1137–1145. [Google Scholar] [CrossRef]
  132. Li, L.; Zhu, L.; Wang, Y.; Zhou, D.; Zhu, J.; Xie, W.; Ye, X. Profiling of microRNAs in AML cells following overexpression or silencing of the VEGF gene. Oncol. Lett. 2017, 13, 105–110. [Google Scholar] [CrossRef]
  133. Xu, Y.; Fang, F.; Zhang, J.; Josson, S.; St Clair, W.H.; St Clair, D.K. miR-17* suppresses tumorigenicity of prostate cancer by inhibiting mitochondrial antioxidant enzymes. PLoS ONE 2010, 5, e14356. [Google Scholar] [CrossRef]
  134. Dong, L.; Deng, J.; Sun, Z.M.; Pan, A.P.; Xiang, X.J.; Zhang, L.; Yu, F.; Chen, J.; Sun, Z.; Feng, M.; et al. Interference with the β-catenin gene in gastric cancer induces changes to the miRNA expression profile. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2015, 36, 6973–6983. [Google Scholar] [CrossRef]
  135. Luo, L.H.; Jin, M.; Wang, L.Q.; Xu, G.J.; Lin, Z.Y.; Yu, D.D.; Yang, S.L.; Ran, R.Z.; Wu, G.; Zhang, T. Long noncoding RNA TCL6 binds to miR-106a-5p to regulate hepatocellular carcinoma cells through PI3K/AKT signaling pathway. J. Cell. Physiol. 2020, 235, 6154–6166. [Google Scholar] [CrossRef] [PubMed]
  136. Li, N.; Miao, Y.; Shan, Y.; Liu, B.; Li, Y.; Zhao, L.; Jia, L. MiR-106b and miR-93 regulate cell progression by suppression of PTEN via PI3K/Akt pathway in breast cancer. Cell Death Dis. 2017, 8, e2796. [Google Scholar] [CrossRef] [PubMed]
  137. Fonseca-Sanchéz, M.A.; Pérez-Plasencia, C.; Fernández-Retana, J.; Arechaga-Ocampo, E.; Marchat, L.A.; Rodríguez-Cuevas, S.; Bautista-Piña, V.; Arellano-Anaya, Z.E.; Flores-Pérez, A.; Diaz-Chávez, J.; et al. microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncol. Rep. 2013, 30, 2399–2410. [Google Scholar] [CrossRef] [PubMed]
  138. Papageorgiou, S.G.; Kontos, C.K.; Tsiakanikas, P.; Stavroulaki, G.; Bouchla, A.; Vasilatou, D.; Bazani, E.; Lazarakou, A.; Scorilas, A.; Pappa, V. Elevated miR-20b-5p expression in peripheral blood mononuclear cells: A novel, independent molecular biomarker of favorable prognosis in chronic lymphocytic leukemia. Leuk. Res. 2018, 70, 1–7. [Google Scholar] [CrossRef] [PubMed]
  139. Hatzl, S.; Geiger, O.; Kuepper, M.K.; Caraffini, V.; Seime, T.; Furlan, T.; Nussbaumer, E.; Wieser, R.; Pichler, M.; Scheideler, M.; et al. Increased Expression of miR-23a Mediates a Loss of Expression in the RAF Kinase Inhibitor Protein RKIP. Cancer Res. 2016, 76, 3644–3654. [Google Scholar] [CrossRef]
  140. Xishan, Z.; Xianjun, L.; Ziying, L.; Guangxin, C.; Gang, L. The malignancy suppression role of miR-23a by targeting the BCR/ABL oncogene in chromic myeloid leukemia. Cancer Gene Ther. 2014, 21, 397–404. [Google Scholar] [CrossRef] [PubMed]
  141. Lin, R.; Zhan, M.; Yang, L.; Wang, H.; Shen, H.; Huang, S.; Huang, X.; Xu, S.; Zhang, Z.; Li, W.; et al. Deoxycholic acid modulates the progression of gallbladder cancer through N6-methyladenosine-dependent microRNA maturation. Oncogene 2020, 39, 4983–5000. [Google Scholar] [CrossRef]
  142. Ma, H.; Wang, L.Y.; Yang, R.H.; Zhou, Y.; Zhou, P.; Kong, L. Identification of reciprocal microRNA-mRNA pairs associated with metastatic potential disparities in human prostate cancer cells and signaling pathway analysis. J. Cell. Biochem. 2019, 120, 17779–17790. [Google Scholar] [CrossRef]
  143. Pal, S.; Baiocchi, R.A.; Byrd, J.C.; Grever, M.R.; Jacob, S.T.; Sif, S. Low levels of miR-92b/96 induce PRMT5 translation and H3R8/H4R3 methylation in mantle cell lymphoma. EMBO J. 2007, 26, 3558–3569. [Google Scholar] [CrossRef]
  144. Pan, Y.; Hu, J.; Ma, J.; Qi, X.; Zhou, H.; Miao, X.; Zheng, W.; Jia, L. MiR-193a-3p and miR-224 mediate renal cell carcinoma progression by targeting alpha-2,3-sialyltransferase IV and the phosphatidylinositol 3 kinase/Akt pathway. Mol. Carcinog. 2018, 57, 1067–1077. [Google Scholar] [CrossRef]
  145. Fan, Q.; Hu, X.; Zhang, H.; Wang, S.; Zhang, H.; You, C.; Zhang, C.Y.; Liang, H.; Chen, X.; Ba, Y. MiR-193a-3p is an Important Tumour Suppressor in Lung Cancer and Directly Targets KRAS. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2017, 44, 1311–1324. [Google Scholar] [CrossRef] [PubMed]
  146. Sun, H.; Sun, Y.; Chen, Q.; Xu, Z. LncRNA KCNQ1OT1 contributes to the progression and chemoresistance in acute myeloid leukemia by modulating Tspan3 through suppressing miR-193a-3p. Life Sci. 2020, 241, 117161. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, Q.; Lu, G.; Cai, Y.; Li, Y.; Xu, R.; Ke, Y.; Zhang, S. MiR-124-5p inhibits the growth of high-grade gliomas through posttranscriptional regulation of LAMB1. Neuro-oncology 2014, 16, 637–651. [Google Scholar] [CrossRef] [PubMed]
  148. von Frowein, J.; Pagel, P.; Kappler, R.; von Schweinitz, D.; Roscher, A.; Schmid, I. MicroRNA-492 is processed from the keratin 19 gene and up-regulated in metastatic hepatoblastoma. Hepatology 2011, 53, 833–842. [Google Scholar] [CrossRef] [PubMed]
  149. Wang, K.; Lü, H.; Qu, H.; Xie, Q.; Sun, T.; Gan, O.; Hu, B. miR-492 Promotes Cancer Progression by Targeting GJB4 and Is a Novel Biomarker for Bladder Cancer. OncoTargets Ther. 2019, 12, 11453–11464. [Google Scholar] [CrossRef] [PubMed]
  150. Di, Z.; Di, M.; Fu, W.; Tang, Q.; Liu, Y.; Lei, P.; Gu, X.; Liu, T.; Sun, M. Integrated Analysis Identifies a Nine-microRNA Signature Biomarker for Diagnosis and Prognosis in Colorectal Cancer. Front. Genet. 2020, 11, 192. [Google Scholar] [CrossRef]
  151. Casabonne, D.; Benavente, Y.; Seifert, J.; Costas, L.; Armesto, M.; Arestin, M.; Besson, C.; Hosnijeh, F.S.; Duell, E.J.; Weiderpass, E.; et al. Serum levels of hsa-miR-16-5p, hsa-miR-29a-3p, hsa-miR-150-5p, hsa-miR-155-5p and hsa-miR-223-3p and subsequent risk of chronic lymphocytic leukemia in the EPIC study. Int. J. Cancer 2020, 147, 1315–1324. [Google Scholar] [CrossRef]
  152. Rokah, O.H.; Granot, G.; Ovcharenko, A.; Modai, S.; Pasmanik-Chor, M.; Toren, A.; Shomron, N.; Shpilberg, O. Downregulation of miR-31, miR-155, and miR-564 in chronic myeloid leukemia cells. PLoS ONE 2012, 7, e35501. [Google Scholar]
  153. Schneider, E.; Staffas, A.; Röhner, L.; Krowiorz, K.; Heuser, M.; Döhner, K.; Bullinger, L.; Döhner, H.; Fogelstrand, L.; Rouhi, A.; et al. MicroRNA-155 is upregulated in MLL-rearranged AML but its absence does not affect leukemia development. Exp. Hematol. 2016, 44, 1166–1171. [Google Scholar] [CrossRef]
  154. El-Maadawy, E.A.; Elshal, M.F.; Bakry, R.M.; Moussa, M.M.; El-Naby, S.; Talaat, R.M. Regulation of CD4+CD25+FOXP3+ cells in Pediatric Acute Lymphoblastic Leukemia (ALL): Implication of cytokines and miRNAs. Mol. Immunol. 2020, 124, 1–8. [Google Scholar] [CrossRef]
  155. Ferrajoli, A.; Shanafelt, T.D.; Ivan, C.; Shimizu, M.; Rabe, K.G.; Nouraee, N.; Ikuo, M.; Ghosh, A.K.; Lerner, S.; Rassenti, L.Z.; et al. Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B chronic lymphocytic leukemia. Bood J. Am. Soc. Hematol. 2013, 122, 1891–1899. [Google Scholar] [CrossRef]
  156. Fulci, V.; Chiaretti, S.; Goldoni, M.; Azzalin, G.; Carucci, N.; Tavolaro, S.; Castellano, L.; Magrelli, A.; Citarella, F.; Messina, M.; et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Bood J. Am. Soc. Hematol. 2007, 109, 4944–4951. [Google Scholar] [CrossRef] [PubMed]
  157. Han, B.; Wang, S.; Zhao, H. MicroRNA-21 and microRNA-155 promote the progression of Burkitt’s lymphoma by the PI3K/AKT signaling pathway. Int. J. Clin. Exp. Pathol. 2020, 13, 89–98. [Google Scholar] [PubMed]
  158. Borchert, G.M.; Holton, N.W.; Larson, E.D. Repression of human activation induced cytidine deaminase by miR-93 and miR-155. BMC Cancer 2011, 11, 347. [Google Scholar] [CrossRef]
  159. Kluiver, J.; Haralambieva, E.; de Jong, D.; Blokzijl, T.; Jacobs, S.; Kroesen, B.J.; Poppema, S.; van den Berg, A. Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes Chromosomes Cancer 2006, 45, 147–153. [Google Scholar] [CrossRef] [PubMed]
  160. Lin, X.; Wang, Z.; Zhang, R.; Feng, W. High serum microRNA-335 level predicts aggressive tumor progression and unfavorable prognosis in pediatric acute myeloid leukemia. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2015, 17, 358–364. [Google Scholar] [CrossRef] [PubMed]
  161. Ronchetti, D.; Lionetti, M.; Mosca, L.; Agnelli, L.; Andronache, A.; Fabris, S.; Deliliers, G.L.; Neri, A. An integrative genomic approach reveals coordinated expression of intronic miR-335, miR-342, and miR-561 with deregulated host genes in multiple myeloma. BMC Med. Genom. 2008, 1, 37. [Google Scholar] [CrossRef]
  162. Saleh, L.M.; Wang, W.; Herman, S.E.; Saba, N.S.; Anastas, V.; Barber, E.; Corrigan-Cummins, M.; Farooqui, M.; Sun, C.; Sarasua, S.M.; et al. Ibrutinib downregulates a subset of miRNA leading to upregulation of tumor suppressors and inhibition of cell proliferation in chronic lymphocytic leukemia. Leukemia 2017, 31, 340–349. [Google Scholar] [CrossRef]
  163. Saccomani, V.; Grassi, A.; Piovan, E.; Bongiovanni, D.; Di Martino, L.; Minuzzo, S.; Tosello, V.; Zanovello, P. miR-22-3p Negatively Affects Tumor Progression in T-Cell Acute Lymphoblastic Leukemia. Cells 2020, 9, 1726. [Google Scholar] [CrossRef]
  164. Qu, H.; Zheng, G.; Cheng, S.; Xie, W.; Liu, X.; Tao, Y.; Xie, B. Serum miR-22 is a novel prognostic marker for acute myeloid leukemia. J. Clin. Lab. Anal. 2020, 34, e23370. [Google Scholar] [CrossRef]
  165. Wei, D.; Miao, Y.; Yu, L.; Wang, D.; Wang, Y. Downregulation of microRNA-198 suppresses cell proliferation and invasion in retinoblastoma by directly targeting PTEN. Mol. Med. Rep. 2018, 18, 595–602. [Google Scholar] [CrossRef] [PubMed]
  166. Marin-Muller, C.; Li, D.; Bharadwaj, U.; Li, M.; Chen, C.; Hodges, S.E.; Fisher, W.E.; Mo, Q.; Hung, M.C.; Yao, Q. A tumorigenic factor interactome connected through tumor suppressor microRNA-198 in human pancreatic cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 5901–5913. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, S.; Zhang, X.; Yang, C.; Xu, S. MicroRNA-198-5p inhibits the migration and invasion of non-small lung cancer cells by targeting fucosyltransferase 8. Clin. Exp. Pharmacol. Physiol. 2019, 46, 955–967. [Google Scholar] [CrossRef] [PubMed]
  168. Zhu, G.; Wang, Z.; Mijiti, M.; Du, G.; Li, Y.; Dangmurenjiafu, G. MiR-28-5p promotes human glioblastoma cell growth through inactivation of FOXO1. Int. J. Clin. Exp. Pathol. 2019, 12, 2972–2980. [Google Scholar]
  169. Xiao, F.; Cheng, Z.; Wang, P.; Gong, B.; Huang, H.; Xing, Y.; Liu, F. MicroRNA-28-5p inhibits the migration and invasion of gastric cancer cells by suppressing AKT phosphorylation. Oncol. Lett. 2018, 15, 9777–9785. [Google Scholar] [CrossRef] [PubMed]
  170. Bartolomé-Izquierdo, N.; de Yébenes, V.G.; Álvarez-Prado, A.F.; Mur, S.M.; Lopez Del Olmo, J.A.; Roa, S.; Vazquez, J.; Ramiro, A.R. miR-28 regulates the germinal center reaction and blocks tumor growth in preclinical models of non-Hodgkin lymphoma. Bood J. Am. Soc. Hematol. 2017, 129, 2408–2419. [Google Scholar] [CrossRef]
  171. Wang, X.X.; Zhang, H.; Li, Y. Preliminary study on the role of miR-148a and DNMT1 in the pathogenesis of acute myeloid leukemia. Mol. Med. Rep. 2019, 19, 2943–2952. [Google Scholar] [CrossRef]
  172. Li, X.; Zou, W.; Wang, Y.; Liao, Z.; Li, L.; Zhai, Y.; Zhang, L.; Gu, S.; Zhao, X. Plasma-based microRNA signatures in early diagnosis of breast cancer. Mol. Genet. Genom. Med. 2020, 8, e1092. [Google Scholar] [CrossRef]
  173. Bhayadia, R.; Krowiorz, K.; Haetscher, N.; Jammal, R.; Emmrich, S.; Obulkasim, A.; Fiedler, J.; Schwarzer, A.; Rouhi, A.; Heuser, M.; et al. Endogenous Tumor Suppressor microRNA-193b: Therapeutic and Prognostic Value in Acute Myeloid Leukemia. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1007–1016. [Google Scholar] [CrossRef]
  174. Chen, J.; Zhang, X.; Lentz, C.; Abi-Daoud, M.; Paré, G.C.; Yang, X.; Feilotter, H.E.; Tron, V.A. miR-193b Regulates Mcl-1 in Melanoma. Am. J. Pathol. 2011, 179, 2162–2168. [Google Scholar] [CrossRef]
  175. Raychaudhuri, M.; Bronger, H.; Buchner, T.; Kiechle, M.; Weichert, W.; Avril, S. MicroRNAs miR-7 and miR-340 predict response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. Treat. 2017, 162, 511–521. [Google Scholar] [CrossRef] [PubMed]
  176. Yu, Z.; Ni, L.; Chen, D.; Zhang, Q.; Su, Z.; Wang, Y.; Yu, W.; Wu, X.; Ye, J.; Yang, S.; et al. Identification of miR-7 as an oncogene in renal cell carcinoma. J. Mol. Histol. 2013, 44, 669–677. [Google Scholar] [CrossRef]
  177. Sun, H.; Zhang, Z.; Luo, W.; Liu, J.; Lou, Y.; Xia, S. MiR-7 Functions as a Tumor Suppressor by Targeting the Oncogenes TAL1 in T-Cell Acute Lymphoblastic Leukemia. Technol. Cancer Res. Treat. 2020, 19, 1533033820934130. [Google Scholar] [CrossRef] [PubMed]
  178. Xin, L.; Liu, L.; Liu, C.; Zhou, L.Q.; Zhou, Q.; Yuan, Y.W.; Li, S.H.; Zhang, H.T. DNA-methylation-mediated silencing of miR-7-5p promotes gastric cancer stem cell invasion via increasing Smo and Hes1. J. Cell. Physiol. 2020, 235, 2643–2654. [Google Scholar] [CrossRef] [PubMed]
  179. Organista-Nava, J.; Gómez-Gómez, Y.; Illades-Aguiar, B.; Del Carmen Alarcón-Romero, L.; Saavedra-Herrera, M.V.; Rivera-Ramírez, A.B.; Garzón-Barrientos, V.H.; Leyva-Vázquez, M.A. High miR-24 expression is associated with risk of relapse and poor survival in acute leukemia. Oncol. Rep. 2015, 33, 1639–1649. [Google Scholar] [CrossRef]
  180. Xu, D.F.; Wang, L.S.; Zhou, J.H. Long non-coding RNA CASC2 suppresses pancreatic cancer cell growth and progression by regulating the miR-24/MUC6 axis. Int. J. Oncol. 2020, 56, 494–507. [Google Scholar] [CrossRef]
  181. Jiang, W.; Meng, K.; Sheng, G.; Yang, T. MicroRNA-24 inhibits the proliferation, migration and invasion and enhances chemosensitivity of human gastric cancer by targeting DND1. J. BUON Off. J. Balk. Union Oncol. 2020, 25, 1001–1006. [Google Scholar]
  182. Wang, F.; Ye, B.G.; Liu, J.Z.; Kong, D.L. miR-487b and TRAK2 that form an axis to regulate the aggressiveness of osteosarcoma, are potential therapeutic targets and prognostic biomarkers. J. Biochem. Mol. Toxicol. 2020, 34, e22511. [Google Scholar] [CrossRef]
  183. Chen, X.; Lin, Z.F.; Xi, W.J.; Wang, W.; Zhang, D.; Yang, F.; Li, Y.F.; Huo, Y.; Zhang, T.Z.; Jiang, Y.H.; et al. DNA methylation-regulated and tumor-suppressive roles of miR-487b in colorectal cancer via targeting MYC, SUZ12, and KRAS. Cancer Med. 2019, 8, 1694–1709. [Google Scholar] [CrossRef]
  184. Gattolliat, C.H.; Thomas, L.; Ciafrè, S.A.; Meurice, G.; Le Teuff, G.; Job, B.; Richon, C.; Combaret, V.; Dessen, P.; Valteau-Couanet, D.; et al. Expression of miR-487b and miR-410 encoded by 14q32.31 locus is a prognostic marker in neuroblastoma. Br. J. Cancer 2011, 105, 1352–1361. [Google Scholar] [CrossRef]
  185. Hua, K.T.; Hong, J.B.; Sheen, Y.S.; Huang, H.Y.; Huang, Y.L.; Chen, J.S.; Liao, Y.H. miR-519d Promotes Melanoma Progression by Downregulating EphA4. Cancer Res. 2018, 78, 216–229. [Google Scholar] [CrossRef] [PubMed]
  186. Xie, Q.; Wang, S.; Zhao, Y.; Zhang, Z.; Qin, C.; Yang, X. MiR-519d impedes cisplatin-resistance in breast cancer stem cells by down-regulating the expression of MCL-1. Oncotarget 2017, 8, 22003–22013. [Google Scholar] [CrossRef] [PubMed]
  187. Xu, Z.; Li, Z.; Wang, W.; Xia, Y.; He, Z.; Li, B.; Wang, S.; Huang, X.; Sun, G.; Xu, J.; et al. MIR-1265 regulates cellular proliferation and apoptosis by targeting calcium binding protein 39 in gastric cancer and, thereby, impairing oncogenic autophagy. Cancer Lett. 2019, 449, 226–236. [Google Scholar] [CrossRef]
  188. Luo, M.; Zhang, Q.; Xia, M.; Hu, F.; Ma, Z.; Chen, Z.; Guo, A.Y. Differential Co-expression and Regulatory Network Analysis Uncover the Relapse Factor and Mechanism of T Cell Acute Leukemia. Mol. Ther. Nucleic Acids 2018, 12, 184–194. [Google Scholar] [CrossRef] [PubMed]
  189. Wei, J.; Yang, L.; Wu, Y.N.; Xu, J. Serum miR-1290 and miR-1246 as Potential Diagnostic Biomarkers of Human Pancreatic Cancer. J. Cancer 2020, 11, 1325–1333. [Google Scholar] [CrossRef]
  190. Chen, G.; Chen, Z.; Zhao, H. MicroRNA-155-3p promotes glioma progression and temozolomide resistance by targeting Six1. J. Cell. Mol. Med. 2020, 24, 5363–5374. [Google Scholar] [CrossRef]
  191. Zhang, G.; Zhong, L.; Luo, H.; Wang, S. MicroRNA-155-3p promotes breast cancer progression through down-regulating CADM1. OncoTargets Ther. 2019, 12, 7993–8002. [Google Scholar] [CrossRef]
  192. Tang, B.; Lei, B.; Qi, G.; Liang, X.; Tang, F.; Yuan, S.; Wang, Z.; Yu, S.; He, S. MicroRNA-155-3p promotes hepatocellular carcinoma formation by suppressing FBXW7 expression. J. Exp. Clin. Cancer Res. CR 2016, 35, 93. [Google Scholar] [CrossRef]
  193. Yim, R.L.; Wong, K.Y.; Kwong, Y.L.; Loong, F.; Leung, C.Y.; Chu, R.; Lam, W.W.; Hui, P.K.; Lai, R.; Chim, C.S. Methylation of miR-155-3p in mantle cell lymphoma and other non-Hodgkin’s lymphomas. Oncotarget 2014, 5, 9770–9782. [Google Scholar] [CrossRef]
  194. Gimenes-Teixeira, H.L.; Lucena-Araujo, A.R.; Dos Santos, G.A.; Zanette, D.L.; Scheucher, P.S.; Oliveira, L.C.; Dalmazzo, L.F.; Silva-Júnior, W.A.; Falcão, R.P.; Rego, E.M. Increased expression of miR-221 is associated with shorter overall survival in T-cell acute lymphoid leukemia. Exp. Hematol. Oncol. 2013, 2, 10. [Google Scholar] [CrossRef]
  195. Cammarata, G.; Augugliaro, L.; Salemi, D.; Agueli, C.; La Rosa, M.; Dagnino, L.; Civiletto, G.; Messana, F.; Marfia, A.; Bica, M.G.; et al. Differential expression of specific microRNA and their targets in acute myeloid leukemia. Am. J. Hematol. 2010, 85, 331–339. [Google Scholar] [CrossRef] [PubMed]
  196. Zhang, L.; Li, H.; Yuan, M.; Li, M.; Zhang, S. Cervical Cancer Cells-Secreted Exosomal microRNA-221-3p Promotes Invasion, Migration and Angiogenesis of Microvascular Endothelial Cells in Cervical Cancer by Down-Regulating MAPK10 Expression. Cancer Manag. Res. 2019, 11, 10307–10319. [Google Scholar] [CrossRef] [PubMed]
  197. Felli, N.; Fontana, L.; Pelosi, E.; Botta, R.; Bonci, D.; Facchiano, F.; Liuzzi, F.; Lulli, V.; Morsilli, O.; Santoro, S.; et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc. Natl. Acad. Sci. USA 2005, 102, 18081–18086. [Google Scholar] [CrossRef] [PubMed]
  198. Frenquelli, M.; Muzio, M.; Scielzo, C.; Fazi, C.; Scarfò, L.; Rossi, C.; Ferrari, G.; Ghia, P.; Caligaris-Cappio, F. MicroRNA and proliferation control in chronic lymphocytic leukemia: Functional relationship between miR-221/222 cluster and p27. Blood J. Am. Soc. Hematol. 2010, 115, 3949–3959. [Google Scholar] [CrossRef] [PubMed]
  199. Song, Q.; An, Q.; Niu, B.; Lu, X.; Zhang, N.; Cao, X. Role of miR-221/222 in Tumor Development and the Underlying Mechanism. J. Oncol. 2019, 2019, 7252013. [Google Scholar] [CrossRef]
  200. Gao, R.; Wang, Z.; Liu, Q.; Yang, C. MicroRNA-105 plays an independent prognostic role in esophageal cancer and acts as an oncogene. Cancer Biomark. Sect. A Dis. Markers 2020, 27, 173–180. [Google Scholar] [CrossRef]
  201. Shang, J.C.; Yu, G.Z.; Ji, Z.W.; Wang, X.Q.; Xia, L. MiR-105 inhibits gastric cancer cells metastasis, epithelial-mesenchymal transition by targeting SOX9. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6160–6169. [Google Scholar]
  202. Li, J.; Zhang, Z.; Chen, F.; Hu, T.; Peng, W.; Gu, Q.; Sun, Y. The Diverse Oncogenic and Tumor Suppressor Roles of microRNA-105 in Cancer. Front. Oncol. 2019, 9, 518. [Google Scholar] [CrossRef]
  203. Pekarsky, Y.; Croce, C.M. Role of miR-15/16 in CLL. Cell Death Differ. 2015, 22, 6–11. [Google Scholar] [CrossRef]
  204. Lovat, F.; Nigita, G.; Distefano, R.; Nakamura, T.; Gasparini, P.; Tomasello, L.; Fadda, P.; Ibrahimova, N.; Catricalà, S.; Palamarchuk, A.; et al. Combined loss of function of two different loci of miR-15/16 drives the pathogenesis of acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2020, 117, 12332–12340. [Google Scholar] [CrossRef]
  205. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Analysis of the 1q abnormalities using dual-color metaphase FISH. Representative images showing 1q duplication (A) and inverted duplication (C) in patient-1 using the BAC clones RP11-553K8 (green) and RP11-42A2 (red). The abnormal chromosomes are indicated by arrowheads. In (A,C), representative dual color FISH images are shown and in (B,D), the corresponding G-banding images are shown (E) Summary of the 1q aberrations achieved by FISH on primary material. The BAC clones selected for this study and their cytogenetic positions are indicated on the right side of the 1q ideogram. The vertical lines indicate the overrepresented chromosomal region as assessed by FISH and correspond to the following patients (from left to right); duplications: patient-1; inverted duplications: patients-1 and -2; partial trisomies: patients-2 and -3. The minimal commonly gained-region 1q21.2–q22 delineated in our previous report using BL-CLs [33] is shaded in grey and indicated with CL (cell lines), and the commonly gained region 1q21.2–q32 delineated in our present study using primary material is indicated with PM (primary material). Pat1–3: Patient-1–3.
Figure 1. Analysis of the 1q abnormalities using dual-color metaphase FISH. Representative images showing 1q duplication (A) and inverted duplication (C) in patient-1 using the BAC clones RP11-553K8 (green) and RP11-42A2 (red). The abnormal chromosomes are indicated by arrowheads. In (A,C), representative dual color FISH images are shown and in (B,D), the corresponding G-banding images are shown (E) Summary of the 1q aberrations achieved by FISH on primary material. The BAC clones selected for this study and their cytogenetic positions are indicated on the right side of the 1q ideogram. The vertical lines indicate the overrepresented chromosomal region as assessed by FISH and correspond to the following patients (from left to right); duplications: patient-1; inverted duplications: patients-1 and -2; partial trisomies: patients-2 and -3. The minimal commonly gained-region 1q21.2–q22 delineated in our previous report using BL-CLs [33] is shaded in grey and indicated with CL (cell lines), and the commonly gained region 1q21.2–q32 delineated in our present study using primary material is indicated with PM (primary material). Pat1–3: Patient-1–3.
Viruses 15 01808 g001aViruses 15 01808 g001b
Figure 2. Differential expression of six genes located at 1q21.2–q22 in BL-CLs. The figure shows the expression level of BCL9 (A), MCL1 (B), ARNT (C), MLLT11 (D), PBXIP1 (E) and FCRL5 (F) relative to their mRNA level detected in the BL-28, an EBV− BL cell line without 1q aberrations. The localization of the investigated genes, their transcriptional direction (indicated with an arrow) and the BAC clones covering their position are highlighted on the 1q ideogram (G). Expression levels in EBV-negative (EBV−) BL-CLs are shown as empty bars and in EBV-positive (EBV+) BL-CLs as black bars. Note that 4 EBV− and 12 EBV+ BL-CLs without (w/o) 1q gains and 13 EBV− and 8 EBV+ BL-CLs with 1q gains were investigated in this study. Each sample was measured in two independent qPCR experiments prepared as duplicates. *: p < 0.05; **: p < 0.01; ***: p < 0.001; ****: p < 0.0001 (two-way ANOVA followed by Tukey’s multiple comparisons test).
Figure 2. Differential expression of six genes located at 1q21.2–q22 in BL-CLs. The figure shows the expression level of BCL9 (A), MCL1 (B), ARNT (C), MLLT11 (D), PBXIP1 (E) and FCRL5 (F) relative to their mRNA level detected in the BL-28, an EBV− BL cell line without 1q aberrations. The localization of the investigated genes, their transcriptional direction (indicated with an arrow) and the BAC clones covering their position are highlighted on the 1q ideogram (G). Expression levels in EBV-negative (EBV−) BL-CLs are shown as empty bars and in EBV-positive (EBV+) BL-CLs as black bars. Note that 4 EBV− and 12 EBV+ BL-CLs without (w/o) 1q gains and 13 EBV− and 8 EBV+ BL-CLs with 1q gains were investigated in this study. Each sample was measured in two independent qPCR experiments prepared as duplicates. *: p < 0.05; **: p < 0.01; ***: p < 0.001; ****: p < 0.0001 (two-way ANOVA followed by Tukey’s multiple comparisons test).
Viruses 15 01808 g002
Figure 3. Summary of the expression profiles of the significantly dysregulated mRNAs and miRNAs in MYC translocation positive BL-CLs depending on their EBV and 1q gain status. The 1q resident miRNAs hsa-miR-9, hsa-miR-9*, hsa-miR-92b, hsa-miR-181a and hsa-miR181b are highlighted in bold letters. The significantly dysregulated mRNAs are indicated in bold and italic letters. The drawing of a hairpin structure indicates miRNA and curved line with a 5′-Cap and 3′-poly-A tail indicates mRNA.
Figure 3. Summary of the expression profiles of the significantly dysregulated mRNAs and miRNAs in MYC translocation positive BL-CLs depending on their EBV and 1q gain status. The 1q resident miRNAs hsa-miR-9, hsa-miR-9*, hsa-miR-92b, hsa-miR-181a and hsa-miR181b are highlighted in bold letters. The significantly dysregulated mRNAs are indicated in bold and italic letters. The drawing of a hairpin structure indicates miRNA and curved line with a 5′-Cap and 3′-poly-A tail indicates mRNA.
Viruses 15 01808 g003
Figure 4. EBV- and 1q-gains-dependent miRNA expression in BL-CLs. Expression profiles of 64 significantly dysregulated miRNAs in BL-CLs depending on their EBV and 1q gain status are shown as diagrams (AF). All miRNAs are numbered according to their appearance in this figure indicated under their names. Value: log2 normalized Hy3/Hy5 ratios. Statistical analysis: two-way ANOVA followed by Tukey’s multiple comparisons test, * indicates the comparison between the BL-CL groups with 1q gains (1q+) vs. without 1q gains (1q−) (p < 0.05) and # indicates the comparison between the BL-CL groups EBV-positive (EBV+) vs. EBV-negative (EBV−) (p < 0.05).
Figure 4. EBV- and 1q-gains-dependent miRNA expression in BL-CLs. Expression profiles of 64 significantly dysregulated miRNAs in BL-CLs depending on their EBV and 1q gain status are shown as diagrams (AF). All miRNAs are numbered according to their appearance in this figure indicated under their names. Value: log2 normalized Hy3/Hy5 ratios. Statistical analysis: two-way ANOVA followed by Tukey’s multiple comparisons test, * indicates the comparison between the BL-CL groups with 1q gains (1q+) vs. without 1q gains (1q−) (p < 0.05) and # indicates the comparison between the BL-CL groups EBV-positive (EBV+) vs. EBV-negative (EBV−) (p < 0.05).
Viruses 15 01808 g004aViruses 15 01808 g004b
Table 1. Cytogenetic features of patients analyzed in this study using G-banding, mFISH and FISH.
Table 1. Cytogenetic features of patients analyzed in this study using G-banding, mFISH and FISH.
PatientG-Banding KaryotypePartial mFISH KaryotypePartial FISH Karyotype a
Pat1 44-45,XY,dup(1)(q22q31),-3,-4, t(8;14)(q24;q32),del(11)(q23),+mar[cp25]45,XY,dup(1)(q22q31),t(8;14)(q24;q32)I. dup(1)(q12q32.1)/
pUC1.77 to RP11-31E23 (PTPRC)
II. dup(1)(q32.1q12)/
RP11-31E23 (PTPRC) to pUC1.77
Pat2 45-47,XY,dup(1)(q21q44)[4], der(6)t(1;6)(q21;q27)[1],del(6)(q22q25)[1], der(7)t(1;7)(q21;p22)[3], t(8;14)(q24;q32)[cp9],+11 [cp9]I.46,XY,der(7)t(1;7)(q;p),t(8;14)(q24;q32)
II.43,XY,dup(1q),t(8;14)(q24;q32)
I. der(7)t(1;7)(q12;p22)/
pUC1.77 to 1qter
II. dup(1)(q41q21.2)/
RP11-74E6 (PTPN14) to RP11-54A4 (MCL1)
Pat3 47,XX,+idic(1)(q10),t(8;14)(q24;q32),
del(10)(p14),der(14)t(8;14)[10]
47,XX,+idic(1)(q10),t(8;14)(q24;q32), der(14)t(8;14)idic(1)(q10)/
centromere 1 to 1qter
a Used FISH probes are indicated after the slash and are summarized in Supplemental Table S2. Pat1–3: Patient-1–3.
Table 2. Characteristics of the Burkitt lymphoma cell lines used in this study.
Table 2. Characteristics of the Burkitt lymphoma cell lines used in this study.
No.BL Cell Line aEBV Status1q Gain StatusMYC TranslocationSexAgeOrder No.Purchased
1BL-28t(8;14)(q24;q32)m19Prof. G. Klein b
2BLUE-1t(8;14)(q24;q32)m29ACC 594DSMZ c
3DG-75t(8;14)(q24;q32)m10ACC 83DSMZ c
4Ramost(8;14)(q24;q32)m3ACC 603DSMZ c
5BL-2par-tri; delt(8;22)(q24;q11)m7Prof. G. Klein b
6BL-92par-trit(8;14)(q24;q32)m14IARC1503IARC d
7ST-486par-trit(8;14)(q24;q32)n.a.n.a.Prof. G. Klein b
8BL-41inv-dup; par-trit(8;14)(q24;q32)m8ACC 160DSMZ c
9CA-46inv-dupt(8;14)(q24;q32)n.a.n.a.ACC 73DSMZ c
10CW698inv-dupt(8;14)(q24;q32)n.a.n.a.Prof. G. Klein b
11BL-30dupt(8;14)(q24;q32)m19IARC116AIARC d
12BL-31dupt(8;14)(q24;q32)m14IARC142AIARC d
13BL-70dupt(8;14)(q24;q32)m16ACC 233DSMZ c
14Loukesdupt(8;14)(q24;q32)n.a.n.a.Prof. G. Klein b
15MN-60dupt(8;14)(q24;q32)m20ACC 138DSMZ c
16Tanouedupt(8;14)(q24;q32)m11ACC 399DSMZ c
17BL-49delt(8;22)(q24;q11)m3Prof. G. Klein b
18AG876+t(8;14)(q24;q32)m8Prof. G. Klein b
19Akuba+t(8;22)(q24;q11)n.a.n.a.Prof. G. Klein b
20BL-18+t(8;14)(q24;q32)m3Prof. G. Klein b
21BL-60+t(8;22)(q24;q11)f4Prof. G. Klein b
22DAUDI+t(8;14)(q24;q32)m16ACC 78DSMZ c
23DOHH-2+t(8;14;18)(q24;q32;q21)f9ACC 47DSMZ c
24Jijoye M13+t(8;14)(q24;q32)m7Prof. G. Klein b
25LY-91+t(2;8)(p12;q24)f7Prof. G. Klein b
26Naliaka+t(8;14)(q24;q32)n.a.n.a.Prof. G. Klein b
27Rael+t(8;14)(q24;q32)n.a.n.a.Prof. G. Klein b
28Raji+t(8;14)(q24;q32)m12ACC 319DSMZ c
29Switzer+t(8;14)(q24;q32)m16Prof. G. Klein b
30BL-16+par-trit(8;14)(q24;q32)f5Prof. G. Klein b
31JI+par-trit(2;8)(p12;q24)f34Prof. G. Klein b
32LY-47+par-trit(8;22)(q24;q11)mn.a.Prof. G. Klein b
33Seraphina+par-trit(8;14)(q24;q32)f7Prof. G. Klein b
34Silfere+par-trit(8;14)(q24;q32)f6Prof. G. Klein b
35LY-66+inv-dupt(2;8)(p12;q24)m13Prof. G. Klein b
36NAMALWA+inv-dupt(8;14)(q24;q32)f3ACC 24DSMZ c
37KK124+dup; delt(8;22)(q24;q11)mn.a.Prof. G. Klein b
All 37 BL-CLs listed here were used for qPCR experiments. a Dark grey highlighted BL-CLs were used for both qPCR and miRNA analysis. b Prof. G. Klein (Department of Microbiology & Tumor Biology, Karolinska Institute, Stockholm, Sweden). c DSMZ-German Collection of Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany). d International Agency for Research on Cancer (IARC, Lyon, France). EBV status is indicated as − (negative) and + (positive) in the third column. A minus (−) in the fourth column indicates no 1q gains. Del: Deletion, dup: Duplication, inv-dup: Inverted duplication, Par-tri: Partial trisomy, m: Male. f: Female. n.a.: Not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Akyüz, N.; Janjetovic, S.; Ghandili, S.; Bokemeyer, C.; Dierlamm, J. EBV and 1q Gains Affect Gene and miRNA Expression in Burkitt Lymphoma. Viruses 2023, 15, 1808. https://doi.org/10.3390/v15091808

AMA Style

Akyüz N, Janjetovic S, Ghandili S, Bokemeyer C, Dierlamm J. EBV and 1q Gains Affect Gene and miRNA Expression in Burkitt Lymphoma. Viruses. 2023; 15(9):1808. https://doi.org/10.3390/v15091808

Chicago/Turabian Style

Akyüz, Nuray, Snjezana Janjetovic, Susanne Ghandili, Carsten Bokemeyer, and Judith Dierlamm. 2023. "EBV and 1q Gains Affect Gene and miRNA Expression in Burkitt Lymphoma" Viruses 15, no. 9: 1808. https://doi.org/10.3390/v15091808

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop