Next Article in Journal
Behavioral and Cognitive Comorbidities in Genetic Rat Models of Absence Epilepsy (Focusing on GAERS and WAG/Rij Rats)
Next Article in Special Issue
Gemtuzumab Ozogamicin in Acute Myeloid Leukemia: Efficacy, Toxicity, and Resistance Mechanisms—A Systematic Review
Previous Article in Journal
Quantitative Computed Tomography Lung COVID Scores with Laboratory Markers: Utilization to Predict Rapid Progression and Monitor Longitudinal Changes in Patients with Coronavirus 2019 (COVID-19) Pneumonia
Previous Article in Special Issue
Case Report of a DDX41 Germline Mutation in a Family with Multiple Relatives Suffering from Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs Associated with a Bad Prognosis in Acute Myeloid Leukemia and Their Impact on Macrophage Polarization

by
Laura Jimbu
1,2,*,
Oana Mesaros
1,2,
Corina Joldes
1,
Alexandra Neaga
1,
Laura Zaharie
1,2 and
Mihnea Zdrenghea
1,2
1
Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania
2
Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(1), 121; https://doi.org/10.3390/biomedicines12010121
Submission received: 29 October 2023 / Revised: 24 December 2023 / Accepted: 3 January 2024 / Published: 7 January 2024
(This article belongs to the Special Issue Advances in the Pathogenesis and Treatment of Acute Myeloid Leukemia)

Abstract

:
MicroRNAs (miRNAs) are short, non-coding ribonucleic acids (RNAs) associated with gene expression regulation. Since the discovery of the first miRNA in 1993, thousands of miRNAs have been studied and they have been associated not only with physiological processes, but also with various diseases such as cancer and inflammatory conditions. MiRNAs have proven to be not only significant biomarkers but also an interesting therapeutic target in various diseases, including cancer. In acute myeloid leukemia (AML), miRNAs have been regarded as a welcome addition to the limited therapeutic armamentarium, and there is a vast amount of data on miRNAs and their dysregulation. Macrophages are innate immune cells, present in various tissues involved in both tissue repair and phagocytosis. Based on their polarization, macrophages can be classified into two groups: M1 macrophages with pro-inflammatory functions and M2 macrophages with an anti-inflammatory action. In cancer, M2 macrophages are associated with tumor evasion, metastasis, and a poor outcome. Several miRNAs have been associated with a poor prognosis in AML and with either the M1 or M2 macrophage phenotype. In the present paper, we review miRNAs with a reported negative prognostic significance in cancer with a focus on AML and analyze their potential impact on macrophage polarization.

1. Introduction

The central dogma of molecular biology was first presented by Francis Crick in a lecture in 1957, where he stated that deoxyribonucleic acid (DNA) is transcribed into ribonucleic acid (RNA) and RNA is translated into proteins [1]. Of course, at that time, the picture was far less sharp than it is today, but Crick was the first to understand the DNA–RNA–protein sequence.
The current understanding of nucleic acids recognizes two main categories of RNAs: coding RNAs, represented by messenger RNA (mRNA) and corresponding to the function described above, namely an intermediate template for the translation of genetic information into amino acid sequences, and non-coding RNAs (ncRNA), whose functions have only more recently been partly understood. According to their functions, ncRNAs are further divided into housekeeping ncRNAs and regulatory ncRNAs (Table 1).
MiRNAs are single-stranded short fragments (19–24 nucleotides) of RNA and their main role is the posttranscriptional regulation of gene expression by mRNA silencing; thus, they have many varied physiological roles [5]. Interestingly, one miRNA is able to regulate more than one gene, sometimes even hundreds, due to the fact that they attach with incomplete complementarity to the mRNA [6]. MiRNAs have not only been associated with physiologic processes, but their dysregulation was also associated with cancer and various nonmalignant diseases [7,8,9,10].
The biogenesis of miRNA can either be canonic or non-canonic [11]. The canonic pathway starts in the nucleus with the primary transcript (pri-microRNA) that is transcribed by either polymerase II or polymerase III [12]. These precursors are usually found in introns or in intragenic regions. An endonuclease called Drosha, together with the double-stranded RNA-binding protein DiGeorge syndrome critical region gene 8 (DGCR8), will cleave this transcript, resulting in a pre-microRNA [6,12]. Via the exportin 5/RanGTP complex, the pre-microRNA will exit the nucleus and travel to the cytoplasm, where a second endonuclease called Dicer will cleave the loop of the hairpin of the pre-microRNA and a mature microRNA duplex will be formed. Argonaute 2 (Ago 2) will cleave the double-stranded RNA into single-stranded RNA. Later, one of the single strands of RNA will be coupled with the RNA-induced silencing complex (RISC) protein complex (formed by transactivating response RNA-binding protein, Ago2, and Dicer), while the other will be degraded. If the single-stranded RNA is complementary to an mRNA, the mRNA will be degraded via the RISC protein complex by cleavage [12]. There are several non-canonical pathways, which are either Drosha- or Dicer-independent. These pathways also lead to inhibition of translation [11].
The history of miRNAs started in the 1980s but peaked in 1993 with the discovery made in Ambros and Ruvnkun’s labs, who described the first microRNAs encoded by lin-4 [13,14]. Their discovery was made by working with Caenorhabditis (C.) elegans, a type of nematode. In order for C. Elegans to progress from stage L1 to L2, the lin-14 protein (p) has to decrease. The regulation of the lin-14 gene is performed in a posttranscriptional manner via lin-4, which encodes for two small RNA molecules which will bind, via complementarity, to the lin-14 gene and will downregulate the production of lin-14, thus allowing the larvae to pass from L1 to L2, a more mature state [13,14]. Later, other miRNAs were described, mounting to presently over a thousand, some present only in one species while others, such as lin-17 [15], are ubiquitous and present in flies to humans [16]. Posttranscriptional gene silencing was also described in plants [17] in 1990, and in 1992, in fungi [18].
The main focus of the present paper is to investigate the association of miRNA expression and macrophage polarization in the setting of cancer with focus on AML. In particular, we will focus on miRNAs reported as being associated with a bad prognosis in this group of diseases and how they correlate with macrophage polarization. The goal of this approach is to assess a potential mechanism of miRNA expression affecting the outcome of AML, by either promoting an M2- or an M1-type microenvironment. In a previous paper, we described the miRNAs associated with a good prognosis in AML and their known effects on macrophages [19].

2. MiRNAs and Cancer

After scientists caught a glimpse of miRNAs in physiologic processes, they started to notice an association between them and different diseases. The first report suggesting the association between miRNAs and cancer, specifically chronic lymphatic leukemia (CLL), was published in 2002 by Croce and colleagues [8]. In their paper, they demonstrated that microRNA (miR)-15 and miR-16 are downregulated in CLL. Both miRNAs were found on chromosome 13q14, a region frequently deleted in several other cancers [20,21,22,23]. Apparently, more than 50% of the miRNAs are situated in ‘fragile sites’ of the genome that are associated with malignancies. The miRNAs that were associated with the development and progression of cancer were named ‘oncomiRs’ [24]. Of course, miRNAs are not only associated with cancer but also with protecting the host against cancer, and thus acting as tumor-suppressors [24]. After Croce’s discovery, a substantial amount of information on miRNAs and cancer emerged.
Based on the information that miRNAs have the ability to act both as an oncomiR and as a tumor suppressor, clinical trials targeting them have been developed [25]. A quick search on www.clinicaltrials.gov (accessed on 2 January 2024) using ‘cancer’ and ‘miR’ showed more than 400 results, mostly focused on miRNAs as biomarkers.
AML is a hematological malignancy with a dismal prognosis. Many reports presented the association between miRNA expression and specific chromosomal abnormalities [26,27] or mutations in AML [28,29,30]. On the other hand, miRNAs were also associated with prognosis, disease progression, or proliferation [31,32,33,34]. Based on these findings, miRNAs are now regarded as important biomarkers that might predict the response to treatment, the risk of progression, or the rate of complete remission (CR) and overall survival (OS) in both solid cancers of hematological malignancies [35,36,37,38]. Only a few studies have focused on the role and association between miRNAs and macrophage polarization in cancer [19,39,40,41].
In AML, several miRNA targets have already been addressed. One of them, miR-126, is highly expressed in patients with inv(16)(p13q22). In a mouse model of inv(16)(p13q22) AML, Kuo et al. showed that adding an miR-126 inhibitor improved survival compared to the control group. Another study showed that adding an miR-10a inhibitor to cytarabine + nutlin-3a, a murine double minute 2 homolog (MDM2) inhibitor, improved OS [42]. MiR-193b is a tumor suppressor in AML, inhibiting excessive proliferation. In AML, miR-193b is downregulated. An miR-193b mimic showed promising results in AML xenografts compared to controls, improving survival [43]. Also downregulated in AML is miR-146 [44]. A study published in 2020 reported that an miR-146 mimic successfully targeted the NF-κB pathway in mice by inhibiting it and also inhibiting TRAF6 and IRAK1, thus inhibiting progression. The same study showed that the miR-146 mimic inhibited proliferation in different cell lines [45]. Another targeted miRNA was miR-9, which is overexpressed in mixed lineage leukemia (MLL). Adding an miR-9 inhibitor in the Mono-Mac-6 cell line has been proven efficacious, decreasing proliferation [46].

3. Macrophages and Their Polarization

Macrophages are part of the innate immunity, playing an important role in phagocytosis, thus eradicating infections and tumor cells, and also in tissue repair [47]. A simplistic yet commonly used classification divides macrophages into two categories based on their polarization: M1 macrophages, classically activated with a pro-inflammatory activity, and M2 macrophages, alternatively activated and anti-inflammatory [48]. Polarization refers to the ability of macrophages to exhibit different functions and phenotypes based on stimuli from the microenvironment. Cytokines such as lipopolysaccharide, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α will tilt the neutral, non-polarized M0 phenotype towards M1 polarization, while apoptotic cells, IL-4, IL-13, IL-10, IL-33, fungi, parasites, and transforming growth factor (TGF)-β will promote M2 macrophages [49,50]. Based on their phenotypes, M1 macrophages express on surface markers such as CD64 and CD86, while M2 macrophages express CD206 and CD163 [51]. The imbalance between M1 and M2 macrophages can either lead to inflammatory states or to cancer due to a very ‘tolerant’ state [51,52]. In cancer, macrophages are very abundant in the tumor environment (TME) and they are frequently called tumor-associated macrophages (TAMs) [53]. In early stages, M1/M2 ratios favor the M1 subtype, but in later stages, the M2 subtype is more common and is associated with a poorer prognosis [54]. A high M2/M1 ratio is associated with proliferation, tumor evasion, angioneogenesis, metastasis, immune suppression, and, overall, a poor prognosis [51].

4. MicroRNAs Associated with a Bad Prognosis of AML and M1 Polarization

The miR-17-92 cluster (miR-17-5p, miR-17-3p, miR-18a, miR-19a, miR-19b, miR-20a, and miR-92-1), or oncomiR-1, is highly dysregulated in several solid and hematological cancers [33]. This cluster regulates a plethora of transcription factors such as TP53, c-MYC, n-MYC, STAT3, MXI1, E2F1, E2F2, E2F3, and NKX3.1. Furthermore, the miR-17-92 cluster regulates TGF-β receptor II, Smad2 and Smad4, BCL2L11, anti-angiogenic factors thrombospondin-1 (TSP-1), connective tissue growth factor (CTGF), insulin gene enhancer protein (Isl-1), and the T-box 1 protein (Tbx1) [55]. In AML, the miR-17-92 cluster is overexpressed and is associated with a poor prognosis. MiR-92a-1-5p was increased in mouse bone-marrow-derived M1 macrophages [56]. On the other hand, downregulation of miR-17-92 is essential for myeloid differentiation [57]. Thus, in MLL, where the abovementioned miRNAs are overexpressed, targeting both miR-17-5p and miR-19a-3p by antagomiRs reduces the ability of MLL cells to form colonies compared to non-MLL AML controls [58]. Several other studies suggested that the miR-17 cluster is associated with a poor prognosis [33,59] through several mechanisms, including the promotion of leukemic blast proliferation [60]. With regard to macrophage polarization, miR-17 and miR-20a promote an M1 phenotype by inhibiting signal-regulatory protein a (SIRPa) [60].
MiR-20a is part of the miR-17-92 cluster and is associated with several cancers. It has been reported that miR-20a interferes with signaling pathways such as ENH1/Id1, MAPK1/c-Myc, PTEN/PI3K/AKT, FBXL5/BTG3, or the Sonic hedgehog pathways [61]. In cancers, the overexpression of miR-20a has been associated both with a poor [62,63] or a good prognosis [64]. Also, its downregulation was associated with a poor prognosis [65]. In a cohort of 61 patients with AML, downregulated miR-20a was associated with a poorer prognosis, compared to those with a high expression [66]. In cell lines, overexpression of miR-20a stimulated apoptosis and inhibited proliferation of AML cells [66], thus suggesting its potential as a therapeutic target. MiR-20a promotes M1 macrophage polarization by inhibiting SIRPa [60,67].
MiR-125b is associated with apoptosis, differentiation, and proliferation by regulating important pathways such as NF-κB, p53, PI3K/Akt/mTOR, ErbB2, and Wnt [68]. Interestingly, miR-125b also targets DICER1, suggesting that its alteration could affect the entire biogenesis of miRNAs. Similar to other miRNAs, miR-125b behaves either as an oncogene or as a tumor suppressor, being either upregulated in certain cancers or downregulated in others. In both AML and acute lymphoblastic leukemia (ALL), miR-125b is generally upregulated [68]. In AML and myelodysplastic syndrome (MDS) patients with t(2;11)(p21;q23) translocation, miR-125b was 90 times more upregulated compared to normal controls. Several subtypes of AML, like fms-related receptor tyrosine kinase 3 (FLT3)-mutated AML, AML harboring the translocation AML-ETO, acute promyelocytic leukemia, and trisomy 21-acute megakaryocytic leukemia, are associated with high levels of miR-125b [69]. Another study showed that miR-125b is highly implicated in myeloid differentiation and erythroid and megakaryocytic progenitor proliferation. In acute megakaryocytic leukemia associated with Down’s syndrome, myeloid differentiation was severely impaired [70]. Interestingly, miR-125b is highly expressed on macrophages, and the studies performed by Chaudhuri et al. showed that overexpression of miR-125b in mice injected with EL4-Fluc thymoma cells increased the macrophages’ capacity for killing and led to tumor shrinkage. MiR-125b regulates macrophage activation via IRF4 [71]. There are conflicting results about miR-125b. Another study showed that mice which overexpressed miR-125b in transplanted liver cells developed various hematological malignancies such as B-ALL, T-ALL, and myeloid neoplasms [72]. In mice, overexpression of miR-125b was associated with the development of myeloproliferative neoplasms and with transformation to AML [73]. In AML, miR-125b is associated with refractoriness to daunorubicin and, thus, with a poorer prognosis, altering apoptosis via decreasing PUMA and GRK2 [74]. Concerning macrophage polarization, miR-125b is associated with the M1 phenotype [75].
MiR-146a and b are situated on chromosomes 5 and 10, respectively, and they are dysregulated in several cancers [76], promoting proliferation and metastasis [77]. A study in miR-146b-knockout mice showed that they developed AML or B-cell lymphoma, probably due to the alteration of the NF-kB pathway by inhibiting TNF receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1) [78]. In AML, downregulation of miR-146a was associated with progression by targeting the NF-κB pathway [79]. On the other hand, a study of 53 AML patients showed that miR-146a is overexpressed and is associated with a poorer prognosis [80]. In pediatric AML with a normal karyotype, hsa-miR-146b was associated with a poor prognosis [81]. Lower levels of miR-146b were detected in low- and intermediate-I-risk MDS compared to intermediate-II- and high-risk MDS [82]. MiR-146b targets IRF5, a transcription factor, promoting the M1 macrophage phenotype [83].
MiR-155 is encoded by a sequence located on the 21st chromosome in the non-coding B cell integration cluster (BIC) gene [84], which is heavily dysregulated in different types of cancers. MiR-155 is also involved in regulating both innate and adaptive immune responses, having an important role in myeloid progenitor differentiation by targeting the transcription factor PU.1. Apart from PU.1, miR-155 also regulates SHIP1, which acts as a negative regulator of the PI3K/Akt pathway involved in several biological processes such as differentiation, apoptosis, transcription, and translation [85]. In lung cancer, a meta-analysis showed that miR-155 could be useful in diagnosis, but could not predict the response to treatment [86]. On the other hand, high levels of miR-155 in breast cancer were associated with a good prognosis and with a good response to immunotherapy [87]. In patients with diffuse large B-cell lymphoma, cobomarsen, an miR-155 inhibitor, showed promising results in preclinical studies by reducing the tumor burden and stimulating apoptosis [88]. Also, in B-cell lymphoma, melanoma, and breast, gastric, ovarian, colon, nasopharyngeal, and pancreatic cancer, overexpression of miR-155 was associated with a better prognosis, acting as a tumor suppressor by unleashing the immune system against the tumor [89]. At the other end of the spectrum, in AML, several studies have proven that upregulated miR-155 is associated with a poor prognosis [90,91]. A study which included 363 patients with AML showed that patients with low levels of miR-155 had a better OS than normal karyotype patients [92]. Similar results have been seen in an AML pediatric cohort of 196 patients with a normal karyotype [93]. From a subtype point of view, high levels of miR-155 were associated with FLT3-internal tandem duplication (ITD) positive AML [94], and MLL [95]. Another inhibitor of miR-155, MLN4924, has been used in the AML setting, improving survival in mice. Its mechanism is based on the upregulation of SHIP1, a phosphatase highly expressed on hematopoietic cells which inhibits survival and proliferation [96]. Furthermore, miR-155 upregulates PU.1, a transcription factor which promotes differentiation [97]. Another study showed that silvestrol, a compound extracted from a plant called Aglaia foveolata, improved survival in mice with FLT3-ITD-mutated AML, decreasing both miR-155 and the expression of FLT3-ITD [98]. In addition, silvestrol showed promising results in combination with cytarabine, daunorubicine, and etoposide [99]. MiR-155 also regulates aerobic glycolysis. In cell lines with knockout miR-155, it has been observed that the treatment sensitivity increased for both FLT3 inhibitors and adriamycin by inhibiting aerobic glycolysis via PIK3R1, a gene also associated with insulin resistance [100]. In cancer, overexpression of miR-155 was associated with the M1 macrophage subtype [84].
Another miRNA associated with a dismal prognosis is miR-210. Higher levels of miR-210 were associated with a low OS [101]. As with other miRNAs, miR-210 is associated with cell proliferation, angioneogenesis, and DNA repair, and its overexpression is associated with a poor prognosis not only in AML, but also in different solid cancers. Interestingly, in ALL, patients with low levels of miR-210 were associated with relapse and with a lower response to treatment [102]. Also, in MDS, miR-210 and miR-155 downregulate SHIP1, and thus tyrosine-protein kinase Tec is upregulated, promoting MDS cell survival [103]. MiR-210 switches macrophages to M1 polarization [104].

5. MicroRNAs Associated with a Bad Prognosis in AML and M2 Polarization

MiR-19a dysregulation has been reported in several cancers [105] and is associated with the upregulation of the NF-κB signaling pathway [106]. MiR-19a was found to be expressed in many human cancers with contradictory consequences, being reported to both promote or inhibit cancer progression in different type of neoplasms [107]. A study by Zhang et al. showed that miR-19a/b was upregulated in AML patients compared to controls. Moreover, overexpression of miR-19a/b was associated with the female gender, elderly patients, several mutations such as U2AF1, C-KIT, CEBPA, and IDH1/2, and a poor prognosis (lower CR rates and OS) [107]. MiR-19a inhibits the M1 subtype by targeting STAT1 and interferon regulatory factor 1 (IRF1) [108] and promotes the M2 macrophage phenotype by activating STAT3 [109].
MiR-21 has regulatory roles in several biological processes and is expressed in all types of cells. MiR-21 is situated on chromosome 17 [110]. This miRNA is dysregulated not only in cancer, but also in several nonmalignant conditions including cardiovascular and pulmonary diseases [111] and autoimmune conditions [112,113]. Its expression is ubiquitous but with varying levels, being highly expressed in immune cells such as dendritic cell, monocytes, and macrophages [110]. Due to its abundant expression in different cells, miR-21 is not really suitable as a biomarker in any disease.
In AML, miR-21 downregulates several genes involved in apoptosis, such as programmed cell death 4 (PDCD4), BTG2, SPRY1, and PTEN [114,115]. Another study showed that miR-21 was overexpressed in AML, while Krüppel-like factor 5 (KLF5) was downregulated. KLF5 is a transcription factor which acts as a tumor suppressor in AML. In nucleophosmin-1 (NPM-1)-mutated AML, miR-21 levels were higher compared to wild-type NPM1, suggesting its role in the diseases pathogenesis [114]. Overexpression of miR-21 was associated with a poorer prognosis [116] and chemo-resistance [115]. Another study showed that in homeobox (HOX)-associated AML, targeting miR-21 and miR-196b improved the prognosis and response to treatment [117]. MiR-21 was shown to promote an M2 macrophage phenotype in different settings either in cancer or in sepsis [118,119].
MiR-23a is part of the miR-23a–27a–24-2 cluster and it is encoded on chromosome 19. MiR-23a is not only associated with cancer but also with other conditions such as cardiac [120] or autoimmune diseases, playing an important role in apoptosis, proliferation, and differentiation [121]. With the exception of erythroid leukemia, where miR-23a is upregulated, in other hematological malignancies such as chronic myeloid leukemia or CLL and in other subtypes of AML, miR-23a is downregulated. The same is true in solid cancers, where in most subtypes, miR-23a is downregulated, while in some cancers like head and neck cancer, it is upregulated [120]. Moreover, miR-23a was associated in several studies with advanced stages, metastasis, and a dismal prognosis or resistance to treatment [120]. Interestingly, the miR-23a-27a-24-2 cluster also regulates macrophage polarization. MiR-23a stimulates M1 macrophage polarization in different settings [122,123], while miR-27a and miR-24-2 promote M2 polarization [122]. MiR-27a is also encoded on chromosome 19 and it is associated with apoptosis, proliferation, differentiation, metastasis, angioneogenesis, and treatment response, playing the role of both a tumor suppressor and an oncogene in different types of cancer [124]. In most cancer patients, miR-27a is overexpressed.
In AML, overexpression of miR-23a is associated with chemo-resistance to cytarabine, lowering the expression of TOP2B, a gene that encodes a DNA topoisomerase involved in different genetic processes [125]. A high miR-24 expression was reported in AML patients with t(8;21), but with no impact on OS and relapse-free survival (RFS) compared to those with a low miR-24 expression [126]. A study which included 147 patients with acute leukemia demonstrated that miR-24 was overexpressed in both AML and ALL compared to healthy controls and was associated with a dismal prognosis [127]. On the other hand, another study showed that miR-24 has a higher expression on AML cells compared to ALL cells [128].
MiR-221/miR-222 are encoded on the Xp11.3 chromosome and they are highly overexpressed in certain types of cancers, such as glioma, bladder, pancreatic, gastric, or colorectal cancer, and in some hematological malignancies, such as CLL, ALL, multiple myeloma (MM), or AML [129,130]. They are associated with tumorigenesis, angiogenesis, metastasis, and a worse prognosis or chemo-resistance in certain types of cancers [129]. In AML, miR-221 and miR-222 are overexpressed and downregulate TP53 via YOD1, a deubiquitinase. A lower level of YOD1 is associated with downregulation of TP53. MiR-221 and miR-222 are inversely correlated with YOD1 [131]. Targeting miR-221 has been shown to be beneficial in a study where an anti-miR-221 and gold nanoparticles co-carrying AS1411, an actamer, inhibited leukemic growth by targeting the NCL/miR-221/NFκB/DNMT1 pathway [132]. In another study, miR-222 and miR-181 were studied in the AML setting. MiR-222 was highly downregulated but was not associated with the response to treatment or FAB classification [133]. On the contrary, other studies suggest that miR-222 is overexpressed in AML and upregulates the Wnt/β-catenin pathway by inhibiting Axin2, a tumor suppressor [134]. Also, miR-222 and miR-181 had a higher expression in AML than in MDS both in peripheral blood and bone marrow [135].
In ovarian cancer, miR-222 induces M2 macrophage proliferation [136]. The same relationship was reported in a study in mice with burn injuries [137].
MiR-126 has been associated with a poor prognosis, treatment refractoriness, and chemo-resistance, targeting the PI3K/AKT/MTOR pathway and thus stimulating proliferation of leukemic cells. In humans, under normal conditions, miR-126 is involved in maintaining the hematopoietic stem cells in quiescence, while in leukemia, the malignant cells are similarly kept in a dormant state. In AML, this miRNA is overexpressed [138]. As expected, inhibiting miR-126 in acute leukemia improved survival by eliminating the malignant cells, while in normal bone marrow, its inhibition stimulated the proliferation of hematopoietic stem cells [139]. Similar results were published by using an antagomiR-126 in AML [140]. High levels of miR-126 were also encountered in the FAB M4Eo AML subtype, and promising results have been shown when using an miR-126 inhibitor [141]. Interestingly, a study from 2015 showed that both overexpression and knockout of miR-126 leads to leukemia by affecting different signaling pathways [142]. Based on these data, miR-126 could be a valuable target in the treatment of AML. In different settings, miR-126 has been associated with the M2 phenotype [143,144,145], but information in cancer settings is lacking.
Figure 1 summarizes the association of all the aforementioned miRNAs with macrophage polarization.
Table 2 presents the miRNAs and their targets (pathways or transcription factors).

6. MicroRNAs Associated with a Bad Prognosis in AML and Unknown Macrophage Polarization

MiR-3151 is encoded in the BAALC gene in intron 1. A study which included 179 patients with de novo, cytogenetically normal AML of ≥60 years old showed that miR-3151 is overexpressed and is associated with mutations such as RUNX1 and MN1, with wild-type NPM1, and with a high expression of the BAALC gene. Even if it was associated with a lower percentage of peripheral blast cells, the prognosis was poorer in this category of patients [146]. In younger AML patients, with intermediate-risk AML, similar results have been reported, suggesting that miR-3151 is a potential biomarker and target in AML [147]. Even after allogeneic hematopoietic stem cell transplantation, overexpression of miR-3151 or the BAALC gene was associated with a poorer prognosis [148]. No convincing evidence of its association with macrophage polarization exists.
MiR-4262 is also associated with a poor prognosis. Patients with a high expression of miR-4262 had a lower OS and lower relapse-free survival compared to low expressors [149]. MiR-4262 targets KLF6, which in the AML setting is downregulated and thus stimulates proliferation and invasion [150]. We have not found any association between macrophage polarization and the abovementioned miRNA.

7. Discussion

AML is a hematological malignancy with a poor prognosis, despite major improvements having been made not only in the management but also in the understanding of the genetic landscape of this disease. Starting in the 1990s, an explosion of data regarding miRNAs emerged, mostly in research proposing their use as biomarkers, both in diagnosis and in the assessment of treatment efficacy. In acute leukemias, as with several other hematologic malignancies, the obtention of diagnostic material is relatively straightforward because the tumor is present in a circulating form and thus can be obtained by drawing blood or by bone marrow aspiration. In contrast, in solid tumors, where the tumor material is sometimes far less easily accessible, circulating plasma miRNAs and liquid biopsies are regarded as promising tools for diagnosis and prognosis. From a therapeutic viewpoint, in preclinical studies, either miRNA inhibitors or miRNA mimics have shown promising results. However, very few have been translated into the clinic. In AML, there are several miRNAs associated with a bad prognosis, most of them upregulated, and thus their inhibition holds promise to improve disease outcomes. On the other hand, macrophages and their M1 or M2 polarization have been associated with the prognosis of both solid cancers and hematological malignancies. In most cases, M2 macrophages are associated with a dismal prognosis, with advanced disease and metastasis. Their involvement in the TME is highly important and thus targeting them could further improve survival. We found that miRNAs associated with a dismal prognosis in AML are not mainly associated with an M2 phenotype, which, in AML too, is linked to progression and a low OS. Thus, miRNAs are likely to influence the prognosis through a number of mechanisms, and not mainly via macrophage polarization.

Author Contributions

Conceptualization, L.J. and M.Z.; investigation, C.J., L.J., O.M., L.Z. and A.N.; writing—original draft preparation, O.M., L.J., C.J., A.N. and L.Z.; writing—review and editing, L.J. and M.Z.; supervision, M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cobb, M. 60 years ago, Francis Crick changed the logic of biology. PLoS Biol. 2017, 15, e2003243. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, P.; Wu, W.; Chen, Q.; Chen, M. Non-Coding RNAs and their Integrated Networks. J. Integr. Bioinform. 2019, 16, 20190027. [Google Scholar] [CrossRef] [PubMed]
  3. Sana, J.; Faltejskova, P.; Svoboda, M.; Slaby, O. Novel classes of non-coding RNAs and cancer. J. Transl. Med. 2012, 10, 103. [Google Scholar] [CrossRef] [PubMed]
  4. Wei, J.-W.; Huang, K.; Yang, C.; Kang, C.-S. Non-coding RNAs as regulators in epigenetics (Review). Oncol. Rep. 2017, 37, 3–9. [Google Scholar] [CrossRef] [PubMed]
  5. Bhaskaran, M.; Mohan, M. MicroRNAs: History, biogenesis, and their evolving role in animal development and disease. Vet. Pathol. 2014, 51, 759–774. [Google Scholar] [CrossRef] [PubMed]
  6. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 2006, 6, 259–269. [Google Scholar] [CrossRef]
  7. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef]
  8. 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]
  9. Zhou, S.-S.; Jin, J.-P.; Wang, J.-Q.; Zhang, Z.-G.; Freedman, J.H.; Zheng, Y.; Cai, L. miRNAS in cardiovascular diseases: Potential biomarkers, therapeutic targets and challenges. Acta Pharmacol. Sin. 2018, 39, 1073–1084. [Google Scholar] [CrossRef]
  10. Tang, X.; Tang, G.; Ozcan, S. Role of microRNAs in diabetes. Biochim. Biophys. Acta 2008, 1779, 697–701. [Google Scholar] [CrossRef]
  11. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  12. Macfarlane, L.A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, R.C.; Ambros, V. An extensive class of small RNAs in Caenorhabditis elegans. Science 2001, 294, 862–864. [Google Scholar] [CrossRef] [PubMed]
  14. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef] [PubMed]
  15. Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquinelli, A.E.; Bettinger, J.C.; Rougvie, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 2000, 403, 901–906. [Google Scholar] [CrossRef] [PubMed]
  16. Pasquinelli, A.E.; Reinhart, B.J.; Slack, F.; Martindale, M.Q.; Kuroda, M.I.; Maller, B.; Hayward, D.C.; Ball, E.E.; Degnan, B.; Müller, P.; et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000, 408, 86–89. [Google Scholar] [CrossRef]
  17. Napoli, C.; Lemieux, C.; Jorgensen, R. Introduction of a Chimeric Chalcone Synthase Gene into Petunia Results in Reversible Co-Suppression of Homologous Genes in trans. Plant Cell 1990, 2, 279–289. [Google Scholar] [CrossRef]
  18. Romano, N.; Macino, G. Quelling: Transient inactivation of gene expression in Neurospora crassa by transformation with homologous sequences. Mol. Microbiol. 1992, 6, 3343–3353. [Google Scholar] [CrossRef]
  19. Neaga, A.; Bagacean, C.; Tempescul, A.; Jimbu, L.; Mesaros, O.; Blag, C.; Tomuleasa, C.; Bocsan, C.; Gaman, M.; Zdrenghea, M. MicroRNAs Associated with a Good Prognosis of Acute Myeloid Leukemia and Their Effect on Macrophage Polarization. Front. Immunol. 2020, 11, 582915. [Google Scholar] [CrossRef]
  20. Zojer, N.; Königsberg, R.; Ackermann, J.; Fritz, E.; Dallinger, S.; Krömer, E.; Kaufmann, H.; Riedl, L.; Gisslinger, H.; Schreiber, S.; et al. Deletion of 13q14 remains an independent adverse prognostic variable in multiple myeloma despite its frequent detection by interphase fluorescence in situ hybridization. Blood 2000, 95, 1925–1930. [Google Scholar] [CrossRef]
  21. Chung, C.-Y.; Kantarjian, H.; Haidar, M.; Starostik, P.; Manshouri, T.; Gidel, C.; Freireich, E.; Keating, M.; Albitar, M. Deletions in the 13q14 locus in adult lymphoblastic leukemia. Cancer 2000, 88, 1359–1364. [Google Scholar] [CrossRef]
  22. Rosenwald, A.; Ott, G.; Krumdiek, A.K.; Dreyling, M.H.; Katzenberger, T.; Kalla, J.; Roth, S.; Ott, M.M.; Müller-Hermelink, H.K. A biological role for deletions in chromosomal band 13q14 in mantle cell and peripheral t-cell lymphomas? Genes Chromosomes Cancer 1999, 26, 210–214. [Google Scholar] [CrossRef]
  23. Rouault, A.; Banneau, G.; MacGrogan, G.; Jones, N.; Elarouci, N.; Barouk-Simonet, E.; Venat, L.; Coupier, I.; Letouzé, E.; de Reyniès, A.; et al. Deletion of Chromosomes 13q and 14q Is a Common Feature of Tumors with BRCA2 Mutations. PLoS ONE 2012, 7, e52079. [Google Scholar] [CrossRef] [PubMed]
  24. Svoronos, A.A.; Engelman, D.M.; Slack, F.J. OncomiR or Tumor Suppressor? The Duplicity of MicroRNAs in Cancer. Cancer Res. 2016, 76, 3666–3670. [Google Scholar] [CrossRef] [PubMed]
  25. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.-S. Therapeutic advances of miRNAs: A preclinical and clinical update. J. Adv. Res. 2021, 28, 127–138. [Google Scholar] [CrossRef] [PubMed]
  26. Dixon-McIver, A.; East, P.; Mein, C.A.; Cazier, J.B.; Molloy, G.; Chaplin, T.; Andrew Lister, T.; Young, B.D.; Debernardi, S. Distinctive patterns of microRNA expression associated with karyotype in acute myeloid leukaemia. PLoS ONE 2008, 3, e2141. [Google Scholar] [CrossRef] [PubMed]
  27. Jongen-Lavrencic, M.; Sun, S.M.; Dijkstra, M.K.; Valk, P.J.M.; Löwenberg, B. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood 2008, 111, 5078–5085. [Google Scholar] [CrossRef]
  28. Liao, Q.; Wang, B.; Li, X.; Jiang, G. miRNAs in acute myeloid leukemia. Oncotarget 2017, 8, 3666–3682. [Google Scholar] [CrossRef]
  29. Hoang, D.H.; Zhao, D.; Branciamore, S.; Maestrini, D.; Rodriguez, I.R.; Kuo, Y.H.; Rockne, R.; Khaled, S.K.; Zhang, B.; Nguyen, L.X.T.; et al. MicroRNA networks in FLT3-ITD acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2022, 119, e2112482119. [Google Scholar] [CrossRef]
  30. Gadewal, N.; Kumar, R.; Aher, S.; Gardane, A.; Gaur, T.; Varma, A.K.; Khattry, N.; Hasan, S.K. miRNA-mRNA Profiling Reveals Prognostic Impact of SMC1A Expression in Acute Myeloid Leukemia. Oncol. Res. 2020, 28, 321–330. [Google Scholar] [CrossRef]
  31. Ghazaryan, A.; Wallace, J.A.; Tang, W.W.; Barba, C.; Lee, S.-H.; Bauer, K.M.; Nelson, M.C.; Kim, C.N.; Stubben, C.; Voth, W.P.; et al. miRNA-1 promotes acute myeloid leukemia cell pathogenesis through metabolic regulation. Front. Genet. 2023, 14, 1192799. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, L.; Chen, R.a.; Zhang, Y.; Fan, W.; Xiao, F.; Yan, X. Low expression of circulating microRNA-328 is associated with poor prognosis in patients with acute myeloid leukemia. Diagn. Pathol. 2015, 10, 109. [Google Scholar] [CrossRef] [PubMed]
  33. Cao, Y.; Liu, Y.; Shang, L.; Chen, H.; Yue, Y.; Dong, W.; Guo, Y.; Yang, H.; Yang, X.; Liu, Y.; et al. Overexpression of miR-17 predicts adverse prognosis and disease recurrence for acute myeloid leukemia. Int. J. Clin. Oncol. 2022, 27, 1222–1232. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, Y.; Cheng, Z.; Pang, Y.; Cui, L.; Qian, T.; Quan, L.; Zhao, H.; Shi, J.; Ke, X.; Fu, L. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J. Hematol. Oncol. 2019, 12, 51. [Google Scholar] [CrossRef] [PubMed]
  35. Macha, M.A.; Seshacharyulu, P.; Krishn, S.R.; Pai, P.; Rachagani, S.; Jain, M.; Batra, S.K. MicroRNAs (miRNAs) as biomarker(s) for prognosis and diagnosis of gastrointestinal (GI) cancers. Curr. Pharm. Des. 2014, 20, 5287–5297. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, J.; Xiao, D.; Wang, J. A 16-miRNA Prognostic Model to Predict Overall Survival in Neuroblastoma. Front. Genet. 2022, 13, 827842. [Google Scholar] [CrossRef]
  37. Gahlawat, A.W.; Fahed, L.; Witte, T.; Schott, S. Total circulating microRNA level as an independent prognostic marker for risk stratification in breast cancer. Br. J. Cancer 2022, 127, 156–162. [Google Scholar] [CrossRef]
  38. Sanchez-Espiridion, B.; Montalban, C.; Guisado, P.; Rodriguez, M.E.; Canales, M.; Alves, J.; Vega, F.; Morente, M.; Tomás, J.F.; Piris, M.A.; et al. Mirna EXPRESSION SIGNATURES as PROGNOSTIC Markers IN ADVANCED Classical HODGKIN LYMPHOMA. Blood 2010, 116, 4157. [Google Scholar] [CrossRef]
  39. Li, L.; Sun, P.; Zhang, C.; Li, Z.; Cui, K.; Zhou, W. MiR-98 modulates macrophage polarization and suppresses the effects of tumor-associated macrophages on promoting invasion and epithelial–mesenchymal transition of hepatocellular carcinoma. Cancer Cell Int. 2018, 18, 95. [Google Scholar] [CrossRef]
  40. Wang, Z.; Xu, L.; Hu, Y.; Huang, Y.; Zhang, Y.; Zheng, X.; Wang, S.; Wang, Y.; Yu, Y.; Zhang, M.; et al. miRNA let-7b modulates macrophage polarization and enhances tumor-associated macrophages to promote angiogenesis and mobility in prostate cancer. Sci. Rep. 2016, 6, 25602. [Google Scholar] [CrossRef]
  41. Iurca, I.; Tirpe, A.; Zimta, A.-A.; Moldovan, C.; Gulei, D.; Slabý, O.; Condorelli, G.; Berindan-Neagoe, I. Macrophages Interaction and MicroRNA Interplay in the Modulation of Cancer Development and Metastasis. Front. Immunol. 2020, 11, 870. [Google Scholar] [CrossRef] [PubMed]
  42. Vu, T.T.; Stölzel, F.; Wang, K.W.; Röllig, C.; Tursky, M.L.; Molloy, T.J.; Ma, D.D. miR-10a as a therapeutic target and predictive biomarker for MDM2 inhibition in acute myeloid leukemia. Leukemia 2021, 35, 1933–1948. [Google Scholar] [CrossRef] [PubMed]
  43. Issa, H.; Bhayadia, R.; Winkler, R.; Swart, L.E.; Heckl, D.; Klusmann, J.-H. Preclinical testing of miRNA-193b-3p mimic in acute myeloid leukemias. Leukemia 2023, 37, 1583–1587. [Google Scholar] [CrossRef] [PubMed]
  44. Shahid, S.; Shahid, W.; Shaheen, J.; Akhtar, M.W.; Sadaf, S. Circulating miR-146a expression as a non-invasive predictive biomarker for acute lymphoblastic leukemia. Sci. Rep. 2021, 11, 22783. [Google Scholar] [CrossRef] [PubMed]
  45. Su, Y.-L.; Wang, X.; Mann, M.; Adamus, T.P.; Wang, D.; Moreira, D.F.; Zhang, Z.; Ouyang, C.; He, X.; Zhang, B.; et al. Myeloid cell–targeted miR-146a mimic inhibits NF-κB–driven inflammation and leukemia progression in vivo. Blood 2020, 135, 167–180. [Google Scholar] [CrossRef]
  46. Chen, P.; Price, C.; Li, Z.; Li, Y.; Cao, D.; Wiley, A.; He, C.; Gurbuxani, S.; Kunjamma, R.B.; Huang, H.; et al. miR-9 is an essential oncogenic microRNA specifically overexpressed in mixed lineage leukemia-rearranged leukemia. Proc. Natl. Acad. Sci. USA 2013, 110, 11511–11516. [Google Scholar] [CrossRef]
  47. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
  48. Mesaros, O.; Jimbu, L.; Neaga, A.; Popescu, C.; Berceanu, I.; Tomuleasa, C.; Fetica, B.; Zdrenghea, M. Macrophage Polarization in Chronic Lymphocytic Leukemia: Nurse-Like Cells Are the Caretakers of Leukemic Cells. Biomedicines 2020, 8, 516. [Google Scholar] [CrossRef]
  49. Yao, Y.; Xu, X.H.; Jin, L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front. Immunol. 2019, 10, 792. [Google Scholar] [CrossRef]
  50. Rőszer, T. Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediat. Inflamm. 2015, 2015, 816460. [Google Scholar] [CrossRef]
  51. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, W.; Guan, N.; Zhang, X.; Liu, Y.; Gao, X.; Wang, L. Study on the imbalance of M1/M2 macrophage polarization in severe chronic periodontitis. Technol. Health Care 2023, 31, 117–124. [Google Scholar] [CrossRef] [PubMed]
  53. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef] [PubMed]
  54. Jayasingam, S.D.; Citartan, M.; Thang, T.H.; Mat Zin, A.A.; Ang, K.C.; Ch’ng, E.S. Evaluating the Polarization of Tumor-Associated Macrophages Into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front. Oncol. 2019, 9, 1512. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. Lu, L.; McCurdy, S.; Huang, S.; Zhu, X.; Peplowska, K.; Tiirikainen, M.; Boisvert, W.A.; Garmire, L.X. Time Series miRNA-mRNA integrated analysis reveals critical miRNAs and targets in macrophage polarization. Sci. Rep. 2016, 6, 37446. [Google Scholar] [CrossRef]
  57. Feketea, G.; Bocsan, C.I.; Popescu, C.; Gaman, M.; Stanciu, L.A.; Zdrenghea, M.T. A Review of Macrophage MicroRNAs’ Role in Human Asthma. Cells 2019, 8, 420. [Google Scholar] [CrossRef]
  58. Mian, Y.A.; Zeleznik-Le, N.J. The miR-17∼92 cluster contributes to MLL leukemia through the repression of MEIS1 competitor PKNOX1. Leuk. Res. 2016, 46, 51–60. [Google Scholar] [CrossRef]
  59. Liu, Y.; Cao, Y.; Yang, X.; Chen, H.; Yang, H.; Liu, Y.; Gu, W. High expression of miR-107 and miR-17 predicts poor prognosis and guides treatment selection in acute myeloid leukemia. Transl. Cancer Res. 2023, 12, 913–927. [Google Scholar] [CrossRef]
  60. Kuo, G.; Wu, C.-Y.; Yang, H.-Y. MiR-17-92 cluster and immunity. J. Formos. Med. Assoc. 2019, 118, 2–6. [Google Scholar] [CrossRef]
  61. Huang, D.; Peng, Y.; Ma, K.; Deng, X.; Tang, L.; Jing, D.; Shao, Z. MiR-20a, a novel promising biomarker to predict prognosis in human cancer: A meta-analysis. BMC Cancer 2018, 18, 1189. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, M.; Gu, H.; Wang, S.; Qian, H.; Zhu, W.; Zhang, L.; Zhao, C.; Tao, Y.; Xu, W. Circulating miR-17-5p and miR-20a: Molecular markers for gastric cancer. Mol. Med. Rep. 2012, 5, 1514–1520. [Google Scholar] [CrossRef] [PubMed]
  63. Cheng, D.; Zhao, S.; Tang, H.; Zhang, D.; Sun, H.; Yu, F.; Jiang, W.; Yue, B.; Wang, J.; Zhang, M.; et al. MicroRNA-20a-5p promotes colorectal cancer invasion and metastasis by downregulating Smad4. Oncotarget 2016, 7, 45199–45213. [Google Scholar] [CrossRef] [PubMed]
  64. Selven, H.; Andersen, S.; Pedersen, M.I.; Lombardi, A.P.G.; Busund, L.-T.R.; Kilvær, T.K. High expression of miR-17-5p and miR-20a-5p predicts favorable disease-specific survival in stage I-III colon cancer. Sci. Rep. 2022, 12, 7080. [Google Scholar] [CrossRef]
  65. Fan, M.-Q.; Huang, C.-B.; Gu, Y.; Xiao, Y.; Sheng, J.-X.; Zhong, L. Decrease expression of microRNA-20a promotes cancer cell proliferation and predicts poor survival of hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2013, 32, 21. [Google Scholar] [CrossRef] [PubMed]
  66. Bao, F.; Zhang, L.; Pei, X.; Lian, C.; Liu, Y.; Tan, H.; Lei, P. MiR-20a-5p functions as a potent tumor suppressor by targeting PPP6C in acute myeloid leukemia. PLoS ONE 2021, 16, e0256995. [Google Scholar] [CrossRef] [PubMed]
  67. Zhu, D.; Pan, C.; Li, L.; Bian, Z.; Lv, Z.; Shi, L.; Zhang, J.; Li, D.; Gu, H.; Zhang, C.-Y.; et al. MicroRNA-17/20a/106a modulate macrophage inflammatory responses through targeting signal-regulatory protein α. J. Allergy Clin. Immunol. 2013, 132, 426–436. [Google Scholar] [CrossRef]
  68. Wang, Y.; Zeng, G.; Jiang, Y. The Emerging Roles of miR-125b in Cancers. Cancer Manag. Res. 2020, 12, 1079–1088. [Google Scholar] [CrossRef]
  69. Shaham, L.; Binder, V.; Gefen, N.; Borkhardt, A.; Izraeli, S. MiR-125 in normal and malignant hematopoiesis. Leukemia 2012, 26, 2011–2018. [Google Scholar] [CrossRef]
  70. Klusmann, J.H.; Li, Z.; Böhmer, K.; Maroz, A.; Koch, M.L.; Emmrich, S.; Godinho, F.J.; Orkin, S.H.; Reinhardt, D. miR-125b-2 is a potential oncomiR on human chromosome 21 in megakaryoblastic leukemia. Genes Dev. 2010, 24, 478–490. [Google Scholar] [CrossRef]
  71. Chaudhuri, A.A.; So, A.Y.; Sinha, N.; Gibson, W.S.; Taganov, K.D.; O’Connell, R.M.; Baltimore, D. MicroRNA-125b potentiates macrophage activation. J. Immunol. 2011, 187, 5062–5068. [Google Scholar] [CrossRef] [PubMed]
  72. Bousquet, M.; Harris, M.H.; Zhou, B.; Lodish, H.F. MicroRNA miR-125b causes leukemia. Proc. Natl. Acad. Sci. USA 2010, 107, 21558–21563. [Google Scholar] [CrossRef]
  73. O’Connell, R.M.; Chaudhuri, A.A.; Rao, D.S.; Gibson, W.S.; Balazs, A.B.; Baltimore, D. MicroRNAs enriched in hematopoietic stem cells differentially regulate long-term hematopoietic output. Proc. Natl. Acad. Sci. USA 2010, 107, 14235–14240. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, L.; Bai, H.; Wang, C.; Wei, D.; Qin, Y.; Xu, X. microRNA-125b promotes leukemia cell resistance to daunorubicin by inhibiting apoptosis. Mol. Med. Rep. 2014, 9, 1909–1916. [Google Scholar] [CrossRef] [PubMed]
  75. Essandoh, K.; Li, Y.; Huo, J.; Fan, G.C. MiRNA-Mediated Macrophage Polarization and its Potential Role in the Regulation of Inflammatory Response. Shock 2016, 46, 122–131. [Google Scholar] [CrossRef] [PubMed]
  76. Shi, L.; Su, Y.; Zheng, Z.; Qi, J.; Wang, W.; Wang, C. miR-146b-5p promotes colorectal cancer progression by targeting TRAF6. Exp. Ther. Med. 2022, 23, 231. [Google Scholar] [CrossRef] [PubMed]
  77. Li, K.; Zhou, Z.; Li, J.; Xiang, R. miR-146b Functions as an Oncogene in Oral Squamous Cell Carcinoma by Targeting HBP1. Technol. Cancer Res. Treat. 2020, 19, 1533033820959404. [Google Scholar] [CrossRef]
  78. Mitsumura, T.; Ito, Y.; Chiba, T.; Matsushima, T.; Kurimoto, R.; Tanaka, Y.; Kato, T.; Uchida, K.; Ito, T.; Yamamoto, K.; et al. Ablation of miR-146b in mice causes hematopoietic malignancy. Blood Adv. 2018, 2, 3483–3491. [Google Scholar] [CrossRef]
  79. Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef]
  80. Wang, Y.; Li, Z.; He, C.; Wang, D.; Yuan, X.; Chen, J.; Jin, J. MicroRNAs expression signatures are associated with lineage and survival in acute leukemias. Blood Cells Mol. Dis. 2010, 44, 191–197. [Google Scholar] [CrossRef]
  81. Zhu, R.; Zhao, W.; Fan, F.; Tang, L.; Liu, J.; Luo, T.; Deng, J.; Hu, Y. A 3-miRNA signature predicts prognosis of pediatric and adolescent cytogenetically normal acute myeloid leukemia. Oncotarget 2017, 8, 38902–38913. [Google Scholar] [CrossRef] [PubMed]
  82. Choi, Y.; Hur, E.H.; Moon, J.H.; Goo, B.K.; Choi, D.R.; Lee, J.H. Expression and prognostic significance of microRNAs in Korean patients with myelodysplastic syndrome. Korean J. Intern. Med. 2019, 34, 390–400. [Google Scholar] [CrossRef] [PubMed]
  83. Peng, L.; Zhang, H.; Hao, Y.; Xu, F.; Yang, J.; Zhang, R.; Lu, G.; Zheng, Z.; Cui, M.; Qi, C.F.; et al. Reprogramming macrophage orientation by microRNA 146b targeting transcription factor IRF5. EBioMedicine 2016, 14, 83–96. [Google Scholar] [CrossRef] [PubMed]
  84. Kalkusova, K.; Taborska, P.; Stakheev, D.; Smrz, D. The Role of miR-155 in Antitumor Immunity. Cancers 2022, 14, 5414. [Google Scholar] [CrossRef] [PubMed]
  85. Xue, H.; Hua, L.-M.; Guo, M.; Luo, J.-M. SHIP1 is targeted by miR-155 in acute myeloid leukemia. Oncol. Rep. 2014, 32, 2253–2259. [Google Scholar] [CrossRef] [PubMed]
  86. Shao, C.; Yang, F.; Qin, Z.; Jing, X.; Shu, Y.; Shen, H. The value of miR-155 as a biomarker for the diagnosis and prognosis of lung cancer: A systematic review with meta-analysis. BMC Cancer 2019, 19, 1103. [Google Scholar] [CrossRef]
  87. Wang, J.; Wang, Q.; Guan, Y.; Sun, Y.; Wang, X.; Lively, K.; Wang, Y.; Luo, M.; Kim, J.A.; Murphy, E.A.; et al. Breast cancer cell–derived microRNA-155 suppresses tumor progression via enhancing immune cell recruitment and antitumor function. J. Clin. Investig. 2022, 132, e157248. [Google Scholar] [CrossRef]
  88. Anastasiadou, E.; Seto, A.G.; Beatty, X.; Hermreck, M.; Gilles, M.E.; Stroopinsky, D.; Pinter-Brown, L.C.; Pestano, L.; Marchese, C.; Avigan, D.; et al. Cobomarsen, an Oligonucleotide Inhibitor of miR-155, Slows DLBCL Tumor Cell Growth In Vitro and In Vivo. Clin. Cancer Res. 2021, 27, 1139–1149. [Google Scholar] [CrossRef]
  89. Nguyen, M.-H.T.; Luo, Y.-H.; Li, A.-L.; Tsai, J.-C.; Wu, K.-L.; Chung, P.-J.; Ma, N. miRNA as a Modulator of Immunotherapy and Immune Response in Melanoma. Biomolecules 2021, 11, 1648. [Google Scholar] [CrossRef]
  90. Hu, X.L.; Tang, A.P. Expression of miR-155 in Acute Myeloid Leukemia and Its Clinical Significance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2016, 24, 980–984. [Google Scholar] [CrossRef]
  91. Tang, L.; Peng, Y.Z.; Li, C.G.; Jiang, H.W.; Mei, H.; Hu, Y. Prognostic and Clinicopathological Significance of MiR-155 in Hematologic Malignancies: A Systematic Review and Meta-analysis. J. Cancer 2019, 10, 654–664. [Google Scholar] [CrossRef] [PubMed]
  92. Marcucci, G.; Maharry, K.S.; Metzeler, K.H.; Volinia, S.; Wu, Y.Z.; Mrózek, K.; Nicolet, D.; Kohlschmidt, J.; Whitman, S.P.; Mendler, J.H.; et al. Clinical role of microRNAs in cytogenetically normal acute myeloid leukemia: miR-155 upregulation independently identifies high-risk patients. J. Clin. Oncol. 2013, 31, 2086–2093. [Google Scholar] [CrossRef] [PubMed]
  93. Ramamurthy, R.; Hughes, M.; Morris, V.; Bolouri, H.; Gerbing, R.B.; Wang, Y.-C.; Loken, M.R.; Raimondi, S.C.; Hirsch, B.A.; Gamis, A.S.; et al. miR-155 expression and correlation with clinical outcome in pediatric AML: A report from Children’s Oncology Group. Pediatr. Blood Cancer 2016, 63, 2096–2103. [Google Scholar] [CrossRef] [PubMed]
  94. Wallace, J.A.; Kagele, D.A.; Eiring, A.M.; Kim, C.N.; Hu, R.; Runtsch, M.C.; Alexander, M.; Huffaker, T.B.; Lee, S.H.; Patel, A.B.; et al. miR-155 promotes FLT3-ITD-induced myeloproliferative disease through inhibition of the interferon response. Blood 2017, 129, 3074–3086. [Google Scholar] [CrossRef] [PubMed]
  95. 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]
  96. Rauh, M.J.; Sly, L.M.; Kalesnikoff, J.; Hughes, M.R.; Cao, L.P.; Lam, V.; Krystal, G. The role of SHIP1 in macrophage programming and activation. Biochem. Soc. Trans. 2004, 32, 785–788. [Google Scholar] [CrossRef]
  97. Li, G.; Hao, W.; Hu, W. Transcription factor PU.1 and immune cell differentiation (Review). Int. J. Mol. Med. 2020, 46, 1943–1950. [Google Scholar] [CrossRef]
  98. Alachkar, H.; Santhanam, R.; Harb, J.G.; Lucas, D.M.; Oaks, J.J.; Hickey, C.J.; Pan, L.; Kinghorn, A.D.; Caligiuri, M.A.; Perrotti, D.; et al. Silvestrol exhibits significant in vivo and in vitro antileukemic activities and inhibits FLT3 and miR-155 expressions in acute myeloid leukemia. J. Hematol. Oncol. 2013, 6, 21. [Google Scholar] [CrossRef]
  99. Cencic, R.; Carrier, M.; Trnkus, A.; Porco, J.A., Jr.; Minden, M.; Pelletier, J. Synergistic effect of inhibiting translation initiation in combination with cytotoxic agents in acute myelogenous leukemia cells. Leuk. Res. 2010, 34, 535–541. [Google Scholar] [CrossRef]
  100. Wang, L.; Jiang, P.; Li, J.; Huang, Y.; Wen, J.; Wu, Z.; Chen, Y.; Hu, J. Loss of MiR-155 Sensitizes FLT3-ITD(+)AML to Chemotherapy and FLT3 Inhibitors via Glycolysis Blocking by Targeting PIK3R1. J. Cancer 2023, 14, 99–113. [Google Scholar] [CrossRef]
  101. Moussa Agha, D.; Rouas, R.; Najar, M.; Bouhtit, F.; Naamane, N.; Fayyad-Kazan, H.; Bron, D.; Meuleman, N.; Lewalle, P.; Merimi, M. Identification of Acute Myeloid Leukemia Bone Marrow Circulating MicroRNAs. Int. J. Mol. Sci. 2020, 21, 7065. [Google Scholar] [CrossRef] [PubMed]
  102. Tang, X.; Chen, L.; Yan, X.; Li, Y.; Xiong, Y.; Zhou, X. Overexpression of miR-210 is Associated with Poor Prognosis of Acute Myeloid Leukemia. Med. Sci. Monit. 2015, 21, 3427–3433. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, D.W.; Futami, M.; Carroll, M.; Feng, Y.; Wang, Z.; Fernandez, M.; Whichard, Z.; Chen, Y.; Kornblau, S.; Shpall, E.J.; et al. Loss of SHIP-1 protein expression in high-risk myelodysplastic syndromes is associated with miR-210 and miR-155. Oncogene 2012, 31, 4085–4094. [Google Scholar] [CrossRef] [PubMed]
  104. Virga, F.; Cappellesso, F.; Stijlemans, B.; Henze, A.-T.; Trotta, R.; Van Audenaerde, J.; Mirchandani, A.S.; Sanchez-Garcia, M.A.; Vandewalle, J.; Orso, F.; et al. Macrophage miR-210 induction and metabolic reprogramming in response to pathogen interaction boost life-threatening inflammation. Sci. Adv. 2021, 7, eabf0466. [Google Scholar] [CrossRef] [PubMed]
  105. Ardizzone, A.; Calabrese, G.; Campolo, M.; Filippone, A.; Giuffrida, D.; Esposito, F.; Colarossi, C.; Cuzzocrea, S.; Esposito, E.; Paterniti, I. Role of miRNA-19a in Cancer Diagnosis and Poor Prognosis. Int. J. Mol. Sci. 2021, 22, 4697. [Google Scholar] [CrossRef]
  106. Gantier, M.P.; Stunden, H.J.; McCoy, C.E.; Behlke, M.A.; Wang, D.; Kaparakis-Liaskos, M.; Sarvestani, S.T.; Yang, Y.H.; Xu, D.; Corr, S.C.; et al. A miR-19 regulon that controls NF-κB signaling. Nucleic Acids Res. 2012, 40, 8048–8058. [Google Scholar] [CrossRef]
  107. Zhang, T.-j.; Lin, J.; Zhou, J.-d.; Li, X.-x.; Zhang, W.; Guo, H.; Xu, Z.-j.; Yan, Y.; Ma, J.-c.; Qian, J. High bone marrow miR-19b level predicts poor prognosis and disease recurrence in de novo acute myeloid leukemia. Gene 2018, 640, 79–85. [Google Scholar] [CrossRef]
  108. Zhu, X.; Guo, Q.; Zou, J.; Wang, B.; Zhang, Z.; Wei, R.; Zhao, L.; Zhang, Y.; Chu, C.; Fu, X.; et al. MiR-19a-3p Suppresses M1 Macrophage Polarization by Inhibiting STAT1/IRF1 Pathway. Front. Pharmacol. 2021, 12, 614044. [Google Scholar] [CrossRef]
  109. Wang, C.; Wang, X.; Zhang, D.; Sun, X.; Wu, Y.; Wang, J.; Li, Q.; Jiang, G. The macrophage polarization by miRNAs and its potential role in the treatment of tumor and inflammation (Review). Oncol. Rep. 2023, 50, 190. [Google Scholar] [CrossRef]
  110. Jenike, A.E.; Halushka, M.K. miR-21: A non-specific biomarker of all maladies. Biomark. Res. 2021, 9, 18. [Google Scholar] [CrossRef]
  111. Kumarswamy, R.; Volkmann, I.; Thum, T. Regulation and function of miRNA-21 in health and disease. RNA Biol. 2011, 8, 706–713. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, S.; Wan, X.; Ruan, Q. The MicroRNA-21 in Autoimmune Diseases. Int. J. Mol. Sci. 2016, 17, 864. [Google Scholar] [CrossRef] [PubMed]
  113. Zahm, A.M.; Thayu, M.; Hand, N.J.; Horner, A.; Leonard, M.B.; Friedman, J.R. Circulating microRNA is a biomarker of pediatric Crohn disease. J. Pediatr. Gastroenterol. Nutr. 2011, 53, 26–33. [Google Scholar] [CrossRef] [PubMed]
  114. Riccioni, R.; Lulli, V.; Castelli, G.; Biffoni, M.; Tiberio, R.; Pelosi, E.; Lo-Coco, F.; Testa, U. miR-21 is overexpressed in NPM1-mutant acute myeloid leukemias. Leuk. Res. 2015, 39, 221–228. [Google Scholar] [CrossRef]
  115. Vandewalle, V.; Essaghir, A.; Bollaert, E.; Lenglez, S.; Graux, C.; Schoemans, H.; Saussoy, P.; Michaux, L.; Valk, P.J.M.; Demoulin, J.B.; et al. miR-15a-5p and miR-21-5p contribute to chemoresistance in cytogenetically normal acute myeloid leukaemia by targeting PDCD4, ARL2 and BTG2. J. Cell Mol. Med. 2021, 25, 575–585. [Google Scholar] [CrossRef] [PubMed]
  116. Li, X.; Zhang, X.; Ma, H.; Liu, Y.; Cheng, S.; Wang, H.; Sun, J. Upregulation of serum exosomal miR-21 was associated with poor prognosis of acute myeloid leukemia patients. Food Sci. Technol. 2022, 42, e51621. [Google Scholar] [CrossRef]
  117. Velu, C.S.; Chaubey, A.; Phelan, J.D.; Horman, S.R.; Wunderlich, M.; Guzman, M.L.; Jegga, A.G.; Zeleznik-Le, N.J.; Chen, J.; Mulloy, J.C.; et al. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J. Clin. Investig. 2014, 124, 222–236. [Google Scholar] [CrossRef]
  118. Ren, W.; Hou, J.; Yang, C.; Wang, H.; Wu, S.; Wu, Y.; Zhao, X.; Lu, C. Extracellular vesicles secreted by hypoxia pre-challenged mesenchymal stem cells promote non-small cell lung cancer cell growth and mobility as well as macrophage M2 polarization via miR-21-5p delivery. J. Exp. Clin. Cancer Res. 2019, 38, 62. [Google Scholar] [CrossRef]
  119. Yao, M.; Cui, B.; Zhang, W.; Ma, W.; Zhao, G.; Xing, L. Exosomal miR-21 secreted by IL-1β-primed-mesenchymal stem cells induces macrophage M2 polarization and ameliorates sepsis. Life Sci. 2021, 264, 118658. [Google Scholar] [CrossRef]
  120. Wang, N.; Tan, H.Y.; Feng, Y.G.; Zhang, C.; Chen, F.; Feng, Y. microRNA-23a in Human Cancer: Its Roles, Mechanisms and Therapeutic Relevance. Cancers 2018, 11, 7. [Google Scholar] [CrossRef]
  121. Guo, Y.X.; Wang, N.; Wu, W.C.; Li, C.Q.; Chen, R.H.; Zhang, Y.; Li, X. The Role of miR-23b in Cancer and Autoimmune Disease. J. Oncol. 2021, 2021, 6473038. [Google Scholar] [CrossRef] [PubMed]
  122. Boucher, A.; Klopfenstein, N.; Hallas, W.M.; Skibbe, J.; Appert, A.; Jang, S.H.; Pulakanti, K.; Rao, S.; Cowden Dahl, K.D.; Dahl, R. The miR-23a~27a~24-2 microRNA Cluster Promotes Inflammatory Polarization of Macrophages. J. Immunol. 2021, 206, 540–553. [Google Scholar] [CrossRef] [PubMed]
  123. Jiang, T.; Sun, L.; Zhu, J.; Li, N.; Gu, H.; Zhang, Y.; Li, M.; Xu, J. MicroRNA-23a-3p promotes macrophage M1 polarization and aggravates lipopolysaccharide-induced acute lung injury by regulating PLK1/STAT1/STAT3 signalling. Int. J. Exp. Pathol. 2022, 103, 198–207. [Google Scholar] [CrossRef] [PubMed]
  124. Li, X.; Xu, M.; Ding, L.; Tang, J. MiR-27a: A Novel Biomarker and Potential Therapeutic Target in Tumors. J. Cancer 2019, 10, 2836–2848. [Google Scholar] [CrossRef] [PubMed]
  125. Hatzl, S.; Perfler, B.; Wurm, S.; Uhl, B.; Quehenberger, F.; Ebner, S.; Troppmair, J.; Reinisch, A.; Wölfler, A.; Sill, H.; et al. Increased Expression of Micro-RNA-23a Mediates Chemoresistance to Cytarabine in Acute Myeloid Leukemia. Cancers 2020, 12, 496. [Google Scholar] [CrossRef] [PubMed]
  126. Yin, J.Y.; Tang, Q.; Qian, W.; Qian, J.; Lin, J.; Wen, X.M.; Zhou, J.D.; Zhang, Y.Y.; Zhu, X.W.; Deng, Z.Q. Increased expression of miR-24 is associated with acute myeloid leukemia with t(8;21). Int. J. Clin. Exp. Pathol. 2014, 7, 8032–8038. [Google Scholar] [PubMed]
  127. 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]
  128. Mi, S.; Lu, J.; Sun, M.; Li, Z.; Zhang, H.; Neilly, M.B.; Wang, Y.; Qian, Z.; Jin, J.; Zhang, Y.; et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2007, 104, 19971–19976. [Google Scholar] [CrossRef]
  129. Abak, A.; Amini, S.; Sakhinia, E.; Abhari, A. MicroRNA-221: Biogenesis, function and signatures in human cancers. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3094–3117. [Google Scholar] [CrossRef]
  130. Song, J.; Ouyang, Y.; Che, J.; Li, X.; Zhao, Y.; Yang, K.; Zhao, X.; Chen, Y.; Fan, C.; Yuan, W. Potential Value of miR-221/222 as Diagnostic, Prognostic, and Therapeutic Biomarkers for Diseases. Front. Immunol. 2017, 8, 56. [Google Scholar] [CrossRef]
  131. Pei, H.Z.; Peng, Z.; Zhuang, X.; Wang, X.; Lu, B.; Guo, Y.; Zhao, Y.; Zhang, D.; Xiao, Y.; Gao, T.; et al. miR-221/222 induce instability of p53 By downregulating deubiquitinase YOD1 in acute myeloid leukemia. Cell Death Discov. 2023, 9, 249. [Google Scholar] [CrossRef] [PubMed]
  132. Deng, R.; Shen, N.; Yang, Y.; Yu, H.; Xu, S.; Yang, Y.-W.; Liu, S.; Meguellati, K.; Yan, F. Targeting epigenetic pathway with gold nanoparticles for acute myeloid leukemia therapy. Biomaterials 2018, 167, 80–90. [Google Scholar] [CrossRef] [PubMed]
  133. Iravani Saadi, M.; Arandi, N.; Yaghobi, R.; Azarpira, N.; Geramizadeh, B.; Ramzi, M. Aberrant Expression of the miR-181b/miR-222 after Hematopoietic Stem Cell Transplantation in Patients with Acute Myeloid Leukemia. Indian J. Hematol. Blood Transfus. 2019, 35, 446–450. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, Z.; Zhong, L.; Dan, W.; Chu, X.; Liu, C.; Luo, X.; Zhang, Z.; Lu, Y.; Wan, P.; Wang, X.; et al. miRNA-222-3p enhances the proliferation and suppresses the apoptosis of acute myeloid leukemia cells by targeting Axin2 and modulating the Wnt/β-catenin pathway. Biochem. Biophys. Res. Commun. 2022, 620, 83–91. [Google Scholar] [CrossRef]
  135. Pons, A.; Nomdedeu, B.; Navarro, A.; Gaya, A.; Gel, B.; Diaz, T.; Valera, S.; Rozman, M.; Belkaid, M.; Montserrat, E.; et al. Hematopoiesis-related microRNA expression in myelodysplastic syndromes. Leuk. Lymphoma 2009, 50, 1854–1859. [Google Scholar] [CrossRef]
  136. Ying, X.; Wu, Q.; Wu, X.; Zhu, Q.; Wang, X.; Jiang, L.; Chen, X.; Wang, X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 2016, 7, 43076–43087. [Google Scholar] [CrossRef]
  137. Ito, I.; Finnerty, C.C.; Herndon, D.N.; Kobayashi, M.; Suzuki, F. miR-222 stimulates M2b macrophage polarization in severely burned mice through the degradation of long noncoding RNA GAS5. J. Immunol. 2019, 202, 187.25. [Google Scholar] [CrossRef]
  138. Lechman, E.R.; Gentner, B.; Ng, S.W.; Schoof, E.M.; van Galen, P.; Kennedy, J.A.; Nucera, S.; Ciceri, F.; Kaufmann, K.B.; Takayama, N.; et al. miR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells. Cancer Cell 2016, 29, 214–228. [Google Scholar] [CrossRef]
  139. de Leeuw, D.C.; Denkers, F.; Olthof, M.C.; Rutten, A.P.; Pouwels, W.; Jan Schuurhuis, G.; Ossenkoppele, G.J.; Smit, L. Attenuation of microRNA-126 Expression That Drives CD34+38− Stem/Progenitor Cells in Acute Myeloid Leukemia Leads to Tumor Eradication. Cancer Res. 2014, 74, 2094–2105. [Google Scholar] [CrossRef]
  140. Dorrance, A.M.; Neviani, P.; Ferenchak, G.J.; Huang, X.; Nicolet, D.; Maharry, K.S.; Ozer, H.G.; Hoellarbauer, P.; Khalife, J.; Hill, E.B.; et al. Targeting leukemia stem cells in vivo with antagomiR-126 nanoparticles in acute myeloid leukemia. Leukemia 2015, 29, 2143–2153. [Google Scholar] [CrossRef]
  141. Zhang, L.; Nguyen, L.X.T.; Chen, Y.-C.; Wu, D.; Cook, G.J.; Hoang, D.H.; Brewer, C.J.; He, X.; Dong, H.; Li, S.; et al. Targeting miR-126 in inv(16) acute myeloid leukemia inhibits leukemia development and leukemia stem cell maintenance. Nat. Commun. 2021, 12, 6154. [Google Scholar] [CrossRef] [PubMed]
  142. Li, Z.; Chen, P.; Su, R.; Li, Y.; Hu, C.; Wang, Y.; Arnovitz, S.; He, M.; Gurbuxani, S.; Zuo, Z.; et al. Overexpression and knockout of miR-126 both promote leukemogenesis. Blood 2015, 126, 2005–2015. [Google Scholar] [CrossRef] [PubMed]
  143. Shou, X.; Wang, Y.; Jiang, Q.; Chen, J.; Liu, Q. miR-126 promotes M1 to M2 macrophage phenotype switching via VEGFA and KLF4. PeerJ 2023, 11, e15180. [Google Scholar] [CrossRef] [PubMed]
  144. Li, J.; Liu, Y.; Lai, W.; Song, L.; Deng, J.; Li, C.; Jiang, S. MicroRNA-126 regulates macrophage polarization to prevent the resorption of alveolar bone in diabetic periodontitis. Arch. Oral Biol. 2023, 150, 105686. [Google Scholar] [CrossRef]
  145. Wei, Y.; Zhu, M.; Schober, A. Macrophage MicroRNAs as Therapeutic Targets for Atherosclerosis, Metabolic Syndrome, and Cancer. Int. J. Mol. Sci. 2018, 19, 1756. [Google Scholar] [CrossRef]
  146. Eisfeld, A.-K.; Marcucci, G.; Maharry, K.; Schwind, S.; Radmacher, M.D.; Nicolet, D.; Becker, H.; Mrózek, K.; Whitman, S.P.; Metzeler, K.H.; et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood 2012, 120, 249–258. [Google Scholar] [CrossRef]
  147. Díaz-Beyá, M.; Brunet, S.; Nomdedéu, J.; Cordeiro, A.; Tormo, M.; Escoda, L.; Ribera, J.M.; Arnan, M.; Heras, I.; Gallardo, D.; et al. The expression level of BAALC-associated microRNA miR-3151 is an independent prognostic factor in younger patients with cytogenetic intermediate-risk acute myeloid leukemia. Blood Cancer J. 2015, 5, e352. [Google Scholar] [CrossRef]
  148. Shakirova, A.I.; Barkhatov, I.M.; Churkina, A.I.; Mamaev, N.N.; Zubarovskaya, L.S.; Afanas’ev, B.V. Clinical Value of miR-3151 Overexpression in Synergistic Interaction with BAALC Host Gene in Patients with Acute Myeloid Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation. Clin. Oncohematol. 2019, 12, 303–308. [Google Scholar] [CrossRef]
  149. Han, G.T.; Sun, Z.L. Up-regulation of serum miR-4262 predicts clinical outcome of patients with acute myeloid leukemia. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2172–2176. [Google Scholar]
  150. Qu, H.; Chen, Y.; Zeng, W.; Huang, X.; Cheng, S. Boosting effects of MiR-4262 on acute myeloid leukemia advancement via governing KLF6. Res. Sq. 2020, in press. [Google Scholar] [CrossRef]
Figure 1. MiRNAs associated with M1 or M2 polarization. MiR-17, 20a, 23a, 125b, 146b, 155, 210 are associated with M1 macrophage polarization, while miR-19a, 21, 27a, 24-2, 222, 126 are associated with the M2 phenotype.
Figure 1. MiRNAs associated with M1 or M2 polarization. MiR-17, 20a, 23a, 125b, 146b, 155, 210 are associated with M1 macrophage polarization, while miR-19a, 21, 27a, 24-2, 222, 126 are associated with the M2 phenotype.
Biomedicines 12 00121 g001
Table 1. Types of non-coding RNAs. Adapted from [2,3] and from [4] with permission.
Table 1. Types of non-coding RNAs. Adapted from [2,3] and from [4] with permission.
NameAbbreviation
housekeeping ncRNAsribosomal RNArRNA
transfer RNAtRNA
small nuclear RNAsnRNA
small nucleolar RNAsnoRNA
telomerase RNATERC
tRNA halvestiRNA
tRNA-derived fragmentst-RF
regulatory ncRNAsmicroRNAmiRNA
small interfering RNAsiRNA
piwi-interfering RNApiRNA
enhancer RNAeRNA
long non-coding RNAslncRNA
circular RNAcircRNA
YRNAYRNA
Table 2. miRNAs and their targets.
Table 2. miRNAs and their targets.
miRNAPathway/Transcription Factor Targeted
miR-17-92 clusterTP53, c-MYC, n-MYC, STAT3, MXI1, E2F1, E2F2, E2F3, NKX3.1, TGF-β receptor II, Smad2, Smad4, BCL2L11, TSP-1, CTGF, Isl-1 and Tbx1
miR-20aENH1/Id1, MAPK/c-Myc, PTEN/PI3K/AKT, FBXL5/BTG3, the Sonic hedgehog pathways, and SIRPa
miR-125bNF-κB, p53, PI3K/Akt/mTOR, ErbB2, Wnt, DICER1, IRF4, PUMA and GRK2
miR-146NF-kB pathway, TRAF6, IRAK1, IRF5
miR-155PI3K/Akt, PU.1, SHIP1
miR-210SHIP1
miR-19aNF-κB, STAT1, IRF1, STAT3
miR-21PDCD4, BTG2, KLF5
miR-23aTOP2B
miR-221TP53
miR-222Wnt/β-catenin pathway, Axin2,
miR-126PI3K/AKT/MTOR
miR-4262KLF6
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

Jimbu, L.; Mesaros, O.; Joldes, C.; Neaga, A.; Zaharie, L.; Zdrenghea, M. MicroRNAs Associated with a Bad Prognosis in Acute Myeloid Leukemia and Their Impact on Macrophage Polarization. Biomedicines 2024, 12, 121. https://doi.org/10.3390/biomedicines12010121

AMA Style

Jimbu L, Mesaros O, Joldes C, Neaga A, Zaharie L, Zdrenghea M. MicroRNAs Associated with a Bad Prognosis in Acute Myeloid Leukemia and Their Impact on Macrophage Polarization. Biomedicines. 2024; 12(1):121. https://doi.org/10.3390/biomedicines12010121

Chicago/Turabian Style

Jimbu, Laura, Oana Mesaros, Corina Joldes, Alexandra Neaga, Laura Zaharie, and Mihnea Zdrenghea. 2024. "MicroRNAs Associated with a Bad Prognosis in Acute Myeloid Leukemia and Their Impact on Macrophage Polarization" Biomedicines 12, no. 1: 121. https://doi.org/10.3390/biomedicines12010121

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