MiRNAs/lncRNAs as Potential Biomarkers for Non-Hodgkin Lymphoma: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Oncology | Cell Biology
Contributor:

Non-Hodgkin lymphoma (NHL) is a heterogeneous malignancy with variable patient outcomes. There is still a lack of understanding about the different players involved in lymphomagenesis, and the identification of new diagnostic and prognostic biomarkers is urgent. MicroRNAs and long non-coding RNAs emerged as master regulators of B-cell development, and their deregulation has been associated with the initiation and progression of lymphomagenesis. They can function by acting alone or, as recently proposed, by creating competing endogenous RNA (ceRNA) networks. The study of miRNAs’ and lncRNAs’ deregulation in NHL, either alone or as ceRNAs networks, offers new insights into the molecular mechanisms underlying lymphoma pathogenesis and opens a window of opportunity to identify potential diagnostic and prognostic biomarkers.

  • lymphoma
  • non-Hodgkin’s lymphoma
  • miRNAs
  • lncRNAs
  • biomarkers
  • ceRNA network

1. Introduction

Non-Hodgkin lymphomas (NHL) are a very heterogeneous group of lymphoproliferative malignancies characterized by the infiltration of lymphoid tissues [1]. The majority of NHL are derived from B cells (85% to 90%), while the remaining are derived from T cells or NK cells [1].
Recently, non-coding RNAs (ncRNAs), which were once thought to be “junk RNA”, have emerged as essential players in the molecular events of normal B-cell development and in lymphomagenesis [5]. MicroRNAs (miRNAs) are undoubtedly the class of ncRNAs most studied over the years, especially due to their relevant biological function in gene regulation [6]. MiRNAs are characterized as small ncRNAs with ~22 nucleotides in length, present in all eukaryotic cells, and highly conserved. They function as gene regulators at a post-transcriptional level through binding to the 3′ untranslated region (UTR) of a target mRNA, which results in their repression or degradation [7]. Recently, the role of miRNAs as regulatory players in B-cell lymphomas is being unveiled, and they have been proposed as potential biomarkers for the diagnosis, prognosis, and prediction of therapy response [8]. To date, it is established that miRNAs can be found in circulation, not only in its cell-free form but also encapsulated in extracellular vesicles (such as exosomes), which permit them to function in a paracrine manner during lymphoma development and progression (reviewed by Fernandes et al. [9]).
Recent studies have shown that another class of ncRNAs, known as long non-coding RNAs (lncRNAs), are also master regulators of multiple protein-coding genes and are involved in all cancer hallmarks [10,11]. LncRNAs are characterized for being more than 200 nucleotides long and can be further classified based on their biogenesis loci in intronic, exonic, intergenic, or overlapping sense/antisense lncRNAs, and divergent/convergent lncRNAs (Figure 1) [12]. These molecules exhibit relatively low expression but high tissue and disease-specific expression patterns [13]. Among the different functions of lncRNAs in gene expression regulation is the remarkable interplay between lncRNAs and miRNAs, which has the ability to balance miRNA function as miRNA sponges/decoys, creating a competitive endogenous RNA (ceRNA) network [14,15]. LncRNAs can sequester miRNAs by presenting biding sequences for miRNAs and impairing their functional interaction with mRNA [16]. Moreover, one lncRNA has the ability to sponge various miRNAs through different biding sites, as seen, for example, for lncRNA MALAT1, which was demonstrated to target miR-101, miR-129, and miR-199a [17,18,19,20]. Therefore, the miRNA regulatory network is more intricate than previously thought by adding another regulatory layer to the network involving lncRNAs. Recent studies have shown that lncRNAs regulate cell differentiation, and their deregulation plays a key role in the pathogenesis of cancer [21,22]. In fact, some studies have been analyzing the expression pattern of lncRNAs in the different B-cell lymphoma subtypes. However, compared to solid tumors, there is still a limited number of studies analyzing the role of lncRNAs during normal B-cell development and as key players in B-cell malignancies.
Figure 1. LncRNA can be classified based on: (A) the genomic location between two coding genes in: intronic, exonic, intergenic, and overlapping lncRNA; (B) the template strand from which they are transcribed in: sense and antisense lncRNA; and (C) the direction of lncRNA transcription in: divergent and convergentlncRNA. Arrows indicate the transcription direction. Red, blue, yellow, and green boxes represent exons from different coding genes.

2. MiRNAs and lncRNAs Deregulation in Lymphomagenesis

2.1. The Role of miRNAs in B-Cell Lymphomagenesis

Considering that B-cell development is a highly regulated process, it is not surprising that miRNAs have been implicated in the regulation of most of the stages comprising this process (Figure 2). Interestingly, during B-cell development, most miRNAs show a stage-specific expression pattern, highlighting their stage-specific function [23]. The process involving B-cell differentiation seems to be prone to malignant transformation, with increasing evidence showing that disruption of the miRNA network takes part in the initiation and maintenance of lymphomagenesis.
Figure 2. miRNA and lncRNA expression during the different stages of B-cell development. During B-cell development, miRNAs and lncRNAs show a stage-specific expression pattern. For example, miR-181a-5p, miR-150-5p, miR-132-3p, and miR-126-3p were shown to be differentially expressed during the development stages of B cells; in particular, miR-181a-5p ectopic overexpression in common lymphoid progenitors results in an increasing total number of B cells. Conversely, overexpression of miR-23a-5p in HSCs results in the inhibition of B-cell development. MiRNAs are involved in the modulation of the checkpoint of pro to pre-B-cell transition. MiR-132-3p shows a stage-specific and BCR-dependent expression, being normally expressed after the pro-B stage; miR-24-3p, miR-34a, and miR-150-5p, when overexpressed, block the transition at pro to pre-B-cell. In secondary lymphoid tissues, miR-155 and miR-181b are highly expressed in activated B-cells in germinal centers. miR-155 and miR-181b-deficient B cells have defective antibody class switching and differentiation into plasma cells; both miRNAs target activation-induced cytidine deaminase (AID) and PU.1, which promote antibody class switching and antibody production. Other miRNAs, e.g., miR-9, miR-125b, and the miR-30 family, are expressed in GC B cells and enhance plasma-cell differentiation. Concerning lncRNAs regulation of B-cell development, lncRNAs MYB-AS1, SMAS-AS1, and LEF-AS1 were found to play a role in early B cells; CRNDE is overexpressed during proliferating stages, such as pre-B-cells and centroblasts in the GC. LncRNA XIST modulates the X-linked gene regulation from antigen naïve B-cells to activated B-cells during B-cell stimulation. Expression of lncRNAs PVT1 and RP11-132N15.3 were associated with the expression of AID in the GC. (Abbreviations: B-cell receptor (BCR); Follicular B cells (FO B-cells); Hematopoietic stem cells (HSCs); Marginal zone B-cells (MZ B-cells)).

2.2. The Role of LncRNAs in B-Cell Lymphomagenesis

Regarding the regulatory role of lncRNA during the different stages of B-cell development and as drivers of B-cell malignancies, there is still scarcer information when compared to miRNAs. LncRNA expression profiling studies have reported that lncRNA exhibits cell-type-specific expression patterns during the different stages of B-cell differentiation (Figure 2) [64,65,66,67]. Consequently, each B-cell subset can be differentiated using its unique lncRNA expression profile [67]. Petri et al., using a guilt-by-association method, analyzed lncRNAs originated from protein-coding genes with known functions in B-cell development and identified antisense lncRNAs, such as MYB-AS1, SMAS-AS1, and LEF-AS1, with roles in early B cells, associated with RAG2, VPREB1, DNTT, LEF1, SMAD1, and MYB expression [66]. On the other hand, lncRNA colorectal neoplasia differentially expressed (CRNDE) showed high expression during the proliferating stages, such as pre-B cells and centroblasts in the GC, which was tightly associated with the expression of mitotic cell cycle genes [66]. In fact, CRNDE was previously demonstrated to be linked to cell-cycle and metabolic changes during proliferation [68,69]. Brazão et al. identified the expression of PVT1 and some uncharacterized lincRNAs, such as LINC00487, LINC00877, and RP11-132N15.3, associated with the expression of AID and SERPINA9, both specifically expressed in GC centroblasts and centrocytes. Of note, RP11-132N15.3 is described to be encoded approximately 240 kilobases upstream of BCL6 [65,66]. Additionally, based on mice models, several lncRNAs demonstrated a PAX5-dependent expression, a transcription factor involved in B-cell commitment, which were shown to be bound by PAX5 and to have human orthologs previously described [65].
Non-coding antisense transcripts of PU.1 were reported to inhibit the expression of PU.1 at a translation level, which could indicate its pivotal role in lymphomagenesis given the regulatory function of PU.1 in B-cell differentiation [72,73].
On the other hand, over the past few years, some studies have been trying to unveil the mechanistic pathways associated with the deregulation of lncRNAs during lymphomagenesis. In this instance, TP53 has been linked to the expression of some lncRNAs in different lymphoma subtypes. In fact, Blume et al. demonstrated, for the first time, the association of lncRNAs and the p53 pathway in CLL and lymphoma by inducing a p53-dependent DNA damage response, which resulted in increased expression of two lncRNAs, NEAT1 and lincRNA-p21, regulating apoptosis or cell-cycle arrest and DNA repair [74]. In DLBCL, p53 can directly bind to the promoter region of the lncRNA PANDA, which inactivates the MAPK/ERK signaling pathway, suppressing the proliferation of DLBCL cells by a G0/G1 cell-cycle arrest [75]. Peng et al. demonstrated that lncRNA HULC regulates DLBCL cell apoptosis and cell proliferation via the upregulation of antiapoptotic BCL2 protein and cyclin D1 [76]. The Wnt/β-catenin signaling pathway was shown to be activated by lncRNA FIRRE through promoting the nuclear translocation of β-catenin [77]. LncRNA DBH-AS1, found to be upregulated in DLBCL, was identified as a positive regulator of cell proliferation, migration, and invasion via binding to the RNA-binding protein BUD13 homolog (BUD13), which in turn regulates fibronectin 1 expression [78].

2.3. The Role of LncRNAs as ceRNAs in B-Cell Lymphomagenesis

Karreth et al. demonstrated that lncRNA BRAFP1, which is aberrantly expressed in B-cell lymphomas, acts as a ceRNA with BRAF mRNA, increasing its stability and BRAF levels by sequestering specific BRAF-targeting miRNAs, such as miR-134, miR-543, and miR-653. Consequently, BRAF activates MAPK signaling, resulting in DLBCL cells’ proliferation [87]. In fact, NEAT1 was identified as an MYC-regulated transcript promoting DLBCL cells proliferation and lymphomagenesis by regulating the miR-34b-5p-GLI1 pathway [88]. Interestingly, NEAT1, along with LincRNA-p21, were also identified as p53-dependent DNA damage response machinery in lymphoma and CLL [74].

Another reported upregulated lncRNA in DLBCL is MALAT1, whose ceRNA function is through sponging miR-195, resulting in the activation of the immune checkpoint molecule PD-L1 and consequently promoting cell proliferation, migration, and immune escape. Moreover, MALAT1 can induce CD8+ T cell apoptosis and epithelial–mesenchymal transition (EMT)-like processes by regulating the Ras/ERK signaling pathway [92]. In MCL, the knockdown of MALAT1 resulted in cell-cycle arrest and impaired proliferation due to the upregulation of p21 and p27 by EZH2 [83]. MiR-423-5p was reported to be involved in a ceRNA network with lncRNA FOXP4-AS1 in MCL cells. Mechanistically, FOXP4-AS1 acts as a sponge to miR-423-5p, upregulating the expression of NACC1, which results in MCL cell proliferation, migration, and invasion [93].

3. MiRNAs and lncRNAs as Potential Biomarkers for NHL

The presence of circulating tumor-associated components, known as “tumor circulome”, which can be easily assessed, appears as a potential option as cancer biomarkers for liquid biopsies (Figure 3) [107]. One of the major components of “tumor circulome”, highly present in circulation, are the miRNAs [108]. MiRNAs emerged as excellent biomarker candidates due to their high stability in biological samples and their high specificity and sensitivity (Table 1) [8]. Similarly, increasing evidence has proposed lncRNAs are promising cancer diagnostic and prognostic biomarkers, especially given their high cell type, tissue, and disease type-specific expression (Table 2). Moreover, lncRNAs have been considered stable and can also be detected in circulation [109]. However, the majority of studies analyzing deregulated lncRNAs in lymphoma have been performed on tissue samples and cell lines.

This entry is adapted from the peer-reviewed paper 10.3390/biomedicines9121934

This entry is offline, you can click here to edit this entry!
Video Production Service