Myelodysplastic neoplasms (MDS) are a group of myeloid neoplasms characterized by clonal proliferation of hematopoietic stem cells (HSC) and genetic and epigenetic abnormalities leading to ineffective hematopoiesis, peripheral cytopenias and a propensity to the development of acute myeloid leukemia (AML) [
1,
2]. Diagnosis is based on the full blood count parameters, the bone marrow morphology and blast count and the presence of cytogenetic and molecular abnormalities, mainly mutations [
2]. The most recent World Health Organization (WHO) classification, 5
th edition recognizes two main groups: a. MDS with defining genetic abnormalities and b. MDS, morphologically defined [
3]. Following correct diagnosis and accurate classification, prognosis estimation and risk stratification is crucial to tailor therapy. The revised International Prognostic Scoring System (IPSS-R) is widely used for the risk stratification of MDS patients considering the number and depth of cytopenias and cytogenetic abnormalities [
4]; while most recently the molecular IPSS (IPSS-M) combined genomic aberrations with hematologic and cytogenetic abnormalities and provided an improved risk stratification of patients with MDS [
5]. In general, low risk patients are managed either expectantly or with recombinant human erythropoietin or luspatercept [
6], whereas high risk patients are being offered hypomethylating agents (HMAs) and/or allogeneic hematopoetic stem transplantation (AlloSCT), that remains the only curative modality. Despite all this progress, there is currently no widely accepted predictive model nor a serviceable biomarker of response that can offer a timely and valid estimation of the expected benefit from these available treatment options.
In terms of pathophysiology, genes regulating epigenetic modifications seem to be the most commonly mutated in patients with MDS [
7]. Among epigenetic modifiers, non-coding RNA molecules, especially micro-RNAs (miRNAs) and long non-coding RNAs (lncRNAs) have recently attracted research interest. Until fairly recently, it was believed that the molecules that are important for the function of a cell are those described by the “Central Dogma” of biology, namely messenger RNAs and proteins. However, almost three decades ago the discovery of microRNAs (miRNAs) in plants [
8] and animals [
9,
10] changed this perception. Subsequent research efforts have demonstrated that large parts of an organism’s genome will be transcribed at one time point or another into RNA, but will not be translated into an amino acid sequence. These RNA transcripts have been referred to as non-coding RNAs (ncRNAs). There are many recognizable classes of ncRNAs, each having a distinct function. These include the above-mentioned miRNAs, transfer RNAs (tRNAs) [
11], ribosomal RNAs (rRNAs) [
12], piwi-interacting (piRNAs) [
13], small nucleolar RNAs (snoRNAs) [
14], long intergenic ncRNAs (lincRNAs) [
15] etc. The full extent of distinct classes of ncRNAs that are encoded within the human genome is currently unknown, but are believed to be numerous.
Functionally ncRNAs are divided into two main categories: housekeeping ncRNAs, which are involved in generic cellular functions and regulatory ncRNAs, which primarily regulate gene expression in multiple levels. Hence, their regulatory role in cellular physiology, including normal hematopoiesis, is important, as is their participation in initiation and progression of neoplasia. Indeed, several studies have demonstrated the role of ncRNAs in solid and hematological malignancies, either from a pathophysiologic point of view or as prognostic biomarkers [
16,
17].
In this review we present a comprehensive summary of findings regarding the emerging role of various ncRNAs in MDS biology, patients’ prognosis and response to therapy.
1.1. miRNAs in hematopoiesis and MDS pathogenesis
HSC are multipotent, self-renewing progenitors that generate all blood cells [
18]. Many genetic and epigenetic regulatory mechanisms are involved in the homeostasis and differentiation of the normal hematopoietic system, including various miRNAs [
19,
20]. MiRNAs belong to a large family of naturally occurring, endogenous, single-stranded ~22-nucleotide-long noncoding RNAs that interact with their target RNA in a sequence-dependent manner, leading to their degradation or translational repression, rendering them significant regulators of posttranscriptional gene expression [
21,
22]. Each specific miRNA can target multiple mRNAs, while each mRNA may be targeted by several miRNAs. To date, more than 3700 human miRNAs have been identified [
23]. MiRNAs are crucial regulators in normal and malignant hematopoiesis [
24,
25]. Chen et al were among the first researchers to identify three miRNAs namely miR-181, miR-223 and miR-142 that were specifically expressed in hematopoietic cells with a dynamic regulation during the early stages of hematopoiesis. MiRNAs implicated in the self-renewal of HSC in mouse models were miR-33 [
26], miR-99 [
27], and miR-125a [
28]. In addition, at least 33 different miRNAs were found to be expressed in CD34+ HSC playing a role in many different cellular processes and blocking differentiation into mature cells [
29]. On the other hand, oncogenic miRNAs (oncomiRs) negatively regulate the expression of tumor suppressor genes, whereas tumour suppressor miRNAs are negative regulators of oncogenes [
30,
31,
32]. The first two oncomiRs that were found to be implicated in cancer were miR-15a and miR-16a in chronic lymphocytic leukemia with deletion 13q14 [
33].
Abnormal expression of miRNAs has also been implicated in MDS in various differently prepared samples and using different techniques and statistical methods [
34]. For instance, miR-150 plays an important role in the regulation of erythropoiesis and megakaryocytopoiesis and its deregulation has been linked to MDS development [
35,
36]. The main target of miR-150 is MYB. MYB or c-Myb is a regulatory transcription factor of the haematopoietic system and gastrointestinal tract preserving the balance between cell division, differentiation and survival [
37]. Deregulation of MYB activity has been associated with several hematologic disorders [
38]. In a zebrafish model, hyperactivity of MYB led to MDS [
38]. In another study, investigators found that MYB was a direct target of miR-150-5p in MDS cells [
36]. In these cells, MYB was increased and its knockdown significantly inhibited cellular proliferation and diminished the proliferation-promoting effect of the inhibitor miR-150-5p. [
36].
Moreover, miR-145 affects megakaryocyte and erythroid differentiation by targeting Fli-1, a megakaryocyte and erythroid regulatory transcription factor [
39]. The miR-17-92 is a polycistronic miR cluster, consisting of miR-17, miR-18a, miR-19a, miR-19b, miR-20a, and miR-92a, which is often overexpressed in certain malignancies. This cluster targets the tumor suppressor PTEN and the pro-apoptotic protein Bim by inhibiting their expression [
40]. By targeting the pro-apoptotic protein Bim, miR-17-92 cluster ensures survival of haematopoietic stem and progenitor cells, playing a crucial role in hematopoiesis [
41]. Moreover, two other members of miR-17-92 cluster namely miR-17-5p and miR-20a that down-regulate E2F1 were found to be under-expressed in high-risk MDS patients constituting favorable prognostic markers associated with increased overall survival (OS) [
42]. In the same study, investigators found that let-7a that down-regulates KRAS was under-expressed in patients with intermediate- or high-risk karyotype [
42].
MiR-143/145 differentially modulate HSC and progenitor activity via suppression of canonical tumor growth factor (TGF)-β signaling and loss of expression of these miRNAs can lead to MDS development [
43]. The interaction between HSC, progenitor cells and bone marrow stromal cells is modulated by CXCL12, a chemokine that is regulated by several different miRNAs [
44]. Among them, miR-23a may have a critical role in MDS pathogenesis by regulating the functional properties of the hematopoietic niche [
44]. MiR-10a and miR-10b were found to be overexpressed in CD34+ cells leading to the up-regulation of TWIST-1 leading to reduced sensitivity to apoptosis [
45]. High levels of miR-21 expression in MDS have been reported to mediate hematopoietic suppression by over-activation of TGF-β signaling [
46]. Several tumor suppressor miRNAs, including several let-7 family members, miR-423, and miR-103a were down-regulated in MDS samples with
SF3B1,
SRSF2, and
U2AF1 (
U2AF35) mutations compared to wild type samples, indicating their role in MDS development [
47]. In another study, it was shown that up-regulation of miR-125a in MDS CD34+ cells modulates NF-kB activation and inhibits erythroid differentiation, rendering miR-125a a potential therapeutic target [
48]. This miRNA is supposed to control the size of the stem cells pool by modulating their apoptosis [
28]. Finally, mutations in the epigenetic modifier
TET2 are involved in the development of myeloid malignancies [
49] and is a target of miR-22, a miRNA that is up-regulated in MDS [
50].
1.2. miRNA deregulation and cytogenetic abnormalities in MDS
Cytogenetic abnormalities are very common in both de novo and secondary MDS [
7,
51]. The deregulation of several miRNAs has been associated with specific cytogenetic abnormalities. In particular, miR-595 is localized in chromosome 7 and targets RPL27A. It has been found to be down-regulated in MDS patients with monosomy 7/isolated loss of 7q (7q-) leading to RPL27A down-regulation, p53 activation, apoptosis, and inhibition of proliferation [
52]. MiR-205-5p is encoded by chromosome 1 and its up-regulation contributes to MDS development via PTEN suppression causing MDS cells proliferation [
53]. Another miRNA that is located in chromosome 1 and its deregulation is involved in MDS pathogenesis is miR-194-5p, in MDS patients with trisomy 1 [
54].
MDS with isolated del(5q) is characterized by anemia and thrombocytosis [
39]. Investigators examined the role of miRNAs that are in this region of chromosome 5 and found that the knockdown of miR-145 and miR-146a resulted in thrombocytosis, mild neutropenia and megakaryocytic dysplasia [
55]. As discussed above, miR-145 affects megakaryocyte and erythroid differentiation by targeting Fli-1, a megakaryocyte and erythroid regulatory transcription factor [
39]. Patients with del(5q) MDS were found to have decreased expression of miR-145 and increased expression of Fli-1 [
39]. Overexpression of miR-150 was also associated with del(5q) MDS contributing to thrombocytosis [
56,
57]. In another study, investigators identified 21 different miRNAs that had aberrant expression in del(5q) MDS patients including miR-34a (up-regulated), miR-378 and miR-146a (downregulated) [
58].
The t(2;11)(p21;q23) translocation has been associated with the overexpression of miR-125b, while trisomy 8 was correlated to miR-383 overexpression in MDS patients [
59,
60]. Kang et al, reported increased expression of miR-661, which is encoded by chromosome 8, in MDS patients via p53 activation [
61]. Another miRNA located on the same chromosome, miR-597, induces apoptosis through down-regulation of FOS Like 2 (FOSL2) and was found to be overexpressed in patients with MDS compared to controls, indicating a possible role in MDS pathogenesis [
62].
1.3. miRNAs as potential prognostic biomarkers in MDS
Many studies have investigated the potential prognostic value of several miRNAs in MDS (
Table 1). In one of the first relevant studies, Sokol et al identified a miRNA signature of ten different miRNAs that was associated with the IPSS risk category and noted the prognostic significance of miR-181 family members in lower-risk MDS patients [
63]. Recently, miR-181a-2-3p was shown to be an independent prognostic biomarker in MDS patients in terms of OS [
64]. Overexpression of miR-125a was associated with shorter OS and it was found to inhibit erythroid differentiation in leukemia and MDS cell lines [
48]. Additionally, miR-22 targets the TET2 tumor suppressor gene and its overexpression, both in plasma and in CD34+ progenitor cells, was associated with high-risk subtypes of MDS and decreased OS [
50,
65].
Deregulation of many miRNAs is associated with the progression of MDS and transformation to AML, which is a synonym for poor prognosis [
66]. Specifically, the up-regulation of miR-196b-5p and down-regulation of miR-29b have been associated with increased risk of AML transformation [
67,
68]. Similarly, Kirimura et al found that the down-regulation of miR-29b in MDS bone marrow cells could play a role in the transformation to AML via the up-regulation of the anti-apoptotic protein myeloid cell leukaemia 1 (MCL-1) [
68]. Expression levels of miR-422a and miR-617 have also been correlated with disease progression in MDS patients [
69]. All members of miR-320 family (miR-320a, miR-320b, miR-320c, miR-320d, and miR-320e) have been reported to be overexpressed in MDS patients and in a series of 82 patients high levels of miR-320c and miR-320d were related to shorter OS, while the up-regulation of miR-320d was found to be an independent prognostic factor [
70].
Furthermore, low levels of miR-194-5p and miR-661 expression have been associated with decreased OS in MDS patients [
54,
61]. In a cohort of 41 patients, miR-125b-5p, miR-155-5p and miR-181a-2-3p bone marrow transcript levels were found elevated in higher-risk patients [
71] and, likewise, low expression levels of miR-21, miR-126 and miR-146b-5p have been detected in lower-risk compared to higher-risk MDS patients. Among them, elevated levels of miR-126 and miR-155 were associated with shorter OS and leukemia-free survival (LFS), while elevated levels of miR-124a tended to be associated with reduced survival rates [
72].
Peripheral blood circulating-microRNA profiles have also emerged as useful diagnostic and prognostic biomarkers for MDS patients [
73,
74]. In particular, the expression levels of miR-27a-3p, miR-150-5p, miR-199a-5p, miR-223-3p and miR-451a were found reduced in higher-risk MDS patients and the decreased levels of miR-451a and miR-223-3p were independently associated with a lower progression-free survival (PFS) and OS, respectively [
74]. Zuo et al, identified and validated a 7-microRNA plasma signature (let-7a, miR-144, miR-16, miR-25, miR-451, miR-651, and miR-655) as an independent predictor of survival in patients with MDS and normal karyotype [
73]. Finally, Hrustincova et al incorporated the expression levels of miR-1237-3p and miR-548av-5p from extracellular vesicles in a prognostic risk score, based on data from 42 patients, as they exhibited the strongest prognostic value in terms of OS [
75].
1.4. miRNAs as potential predictive biomarkers in MDS
Several studies have attempted to investigate the potential role of miRNAs as predictors of treatment response in patients with MDS (
Table 2). Lenalidomide is an immunomodulatory agent that selectively suppresses the del(5q) clone and is used for the treatment of lower-risk MDS with del(5q) [
6,
76]. Down-regulation of miR-145 and miR-146, which are encoded by chromosome 5, plays a crucial role in the development of del(5q) MDS via increased expression of their target genes, TIRAP and TRAF6, respectively, leading to inappropriate activation of innate immune signaling [
77]. In a phase II single arm study in lower-risk MDS patients with anemia, miR-145 and miR-146 were decreased at baseline in patients with del(5q) MDS and significantly up-regulated after 3 and 6 months of treatment with lenalidomide [
78]. In another study investigators found that the expression levels of miR-143 and miR-145 were increased during treatment and lenalidomide selectively abrogated progenitor activity in cells depleted of miR-143 and miR-145 rendering them potential predictive biomarkers [
79]. Similarly, expression of miRNAs clustering to the 14q32 region and pro-apoptotic miR-34a and miR-34a* was reduced following lenalidomide administration [
80,
81].
HMAs are nucleoside analogs used for the treatment of higher-risk MDS and the prediction of HMA responsiveness is deemed of critical importance [
6]. In a study of 27 patients with higher-risk MDS or AML with myelodysplasia-related changes, the investigators examined the predictive value of specific miRNAs, expressed in bone marrow CD34
+ cells before and after the administration of azacytidine [
82]. Up-regulation of miR-17-3p and down-regulation of miR-100-5p and miR-133b at baseline was associated with higher overall response rate (ORR) while increased levels of miR-100-5p were associated with shorter OS [
82]. Furthermore, deregulation of 30 different miRNAs was observed after the administration of azacytidine in responders. Specifically, miR-10b-5p, miR-15a-5p/b-5p, miR-24-3p, and miR-148b-3p were down-regulated in responders after azacytidine treatment while they remained at the same levels in non-responders, thus rendering them potential predictive biomarkers [
82]. Mongiorgi et al recently showed that miR-192-5p specifically targets and inhibits BCL2 and its overexpression in bone marrow mononuclear cells was correlated to increased OS and leukemia-free survival (LFS) in MDS patients responding to combination of azacytidine and lenalidomide [
83]. In a recent study, investigators evaluated the predictive value of miR-22 in MDS patients after HMAs, however, they concluded that it is not an appropriate predictive biomarker [
84].
Regarding circulating miRNAs in the peripheral blood, miR-21 is a potential predictive biomarker for response to HMA therapy in patients with MDS, since the baseline level of serum miR-21 was found significantly decreased in responders compared to non-responders [
85]. MiR-124 is involved in MDS pathogenesis via targeting the cyclin-dependent kinase 6 (CDK6) gene and was up-regulated in response to epigenetic treatments, azacytidine or the histone deacetylase inhibitor panobinostat, in peripheral blood and bone marrow mononuclear cells [
86,
87]. In another study of 42 MDS patients, investigators identified five circulating miRNAs, namely miR-423-5p, miR-126-3p, miR-151a-3p, miR-125a-5p and miR-199a-3p whose combined expression levels in plasma could predict response to azacytidine therapy [
75]. Finally, beyond HMAs, in a recent study investigators found that overexpression of exosomal miR-92a (member of miR17-92 cluster) in plasma promoted cytarabine resistance in MDS/AML by activating Wnt/β-catenin signaling pathway, rendering miR-92a both a potential predictive biomarker and a therapeutic target for patients with MDS [
88].