Next Article in Journal
Atraric Acid Exhibits Anti-Inflammatory Effect in Lipopolysaccharide-Stimulated RAW264.7 Cells and Mouse Models
Previous Article in Journal
Biphasic Force-Regulated Phosphorylation Site Exposure and Unligation of ERM Bound with PSGL-1: A Novel Insight into PSGL-1 Signaling via Steered Molecular Dynamics Simulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification of Acute Myeloid Leukemia Bone Marrow Circulating MicroRNAs

1
Laboratory of Experimental Hematology, Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
2
Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada
3
Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
4
Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
5
Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(19), 7065; https://doi.org/10.3390/ijms21197065
Submission received: 2 September 2020 / Revised: 21 September 2020 / Accepted: 22 September 2020 / Published: 25 September 2020
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Background: In addition to their roles in different biological processes, microRNAs in the tumor microenvironment appear to be potential diagnostic and prognostic biomarkers for various malignant diseases, including acute myeloid leukemia (AML). To date, no screening of circulating miRNAs has been carried out in the bone marrow compartment of AML. Accordingly, we investigated the circulating miRNA profile in AML bone marrow at diagnosis (AMLD) and first complete remission post treatment (AMLPT) in comparison to healthy donors (HD). Methods: Circulating miRNAs were isolated from AML bone marrow aspirations, and a low-density TaqMan miRNA array was performed to identify deregulated miRNAs followed by quantitative RT-PCR to validate the results. Bioinformatic analysis was conducted to evaluate the diagnostic and prognostic accuracy of the highly and significantly identified deregulated miRNA(s) as potential candidate biomarker(s). Results: We found several deregulated miRNAs between the AMLD vs. HD vs. AMLPT groups, which were involved in tumor progression and immune suppression pathways. We also identified significant diagnostic and prognostic signatures with the ability to predict AML patient treatment response. Conclusions: This study provides a possible role of enriched circulating bone marrow miRNAs in the initiation and progression of AML and highlights new markers for prognosis and treatment monitoring.

Graphical Abstract

1. Introduction

Acute myeloid leukemia (AML) is a common myelogenous malignancy in adults that is often characterized by disease relapse. It is caused by the acquisition of cytogenetic and molecular abnormalities by a hematopoietic stem cell, which transforms it into a leukemic stem cell that self-renews and proliferates [1]. Although many researchers are interested in improving our understanding of AML disease evolution [2], its pathogenesis, resistance to treatments and escape from immune surveillance have not yet been fully elucidated [3]. Despite recent advances in understanding the molecular basis of this cancer, allowing the development of target-specific therapies, more than 50% of patients with AML relapse and die [4]. They develop resistance to treatment caused by different genetic and immunosuppressive mechanisms. These mechanisms include changes in the expression levels of both intracellular and circulating microRNAs (miRNAs) [3,5,6,7]. Moreover, AML has been proven to be a heterogeneous disease, and to date, the cytogenetic classification fails to predict the clinical outcome for a large group of patients and therefore fails to guide the continuation of treatment in almost one-third of the patients [8]. The prognostic factors currently used remain unsatisfactory, and it is necessary to explore new biomarkers for the diagnosis, prognosis, and therapeutic targets of AML to develop more effective surveillance and treatment programs [3].
Small (19- to 22-nucleotide long) noncoding RNAs called microRNAs are crucial for the posttranscriptional regulation of gene expression. They play an integral role in numerous biological processes, including the immune response, cell-cycle control, metabolism, stem cell self-renewal and differentiation [9]. Aberrant miRNA expression is associated with many diseases, including cancer, and miRNA-based drugs represent a novel and potentially powerful therapeutic approach [10,11,12]. Several studies have proposed using miRNAs as biomarkers for diagnosis, prediction and prognosis in cancer diseases, including AML [13,14]. Extracellular miRNAs, namely, those circulating in peripheral blood, seem to be attracting increasing attention from researchers. Given the ease with which miRNAs can be isolated and their structural stability under different conditions of treatment and sample isolation, they are also proposed as diagnostic, prognostic or predictive biomarkers in several cancers [15,16].
In AML, peripheral blood circulating miRNAs have been detected and could be useful as noninvasive biomarkers for the detection of leukemia at the time of diagnosis and prognosis [3,7,14,17]. In our laboratory, we previously identified eight plasma miRs differentially expressed between healthy donors and AML patients, showing that miR-150 and miR-342 are downregulated in the plasma of AML patients at diagnosis, and their expression levels in complete remission AML patients resembled those in healthy controls [18]. Zhi et al. performed an analysis using AML serum samples and showed a significant increase in six miRNAs (miR-10a-5p, miR-93-5p, miR-129-5p, miR-155-5p, miR-181b-5p and miR-320d) [19]. The level of miR-181b-5p in serum, on the other hand, is significantly associated with an increased survival rate [19]. Yan J. and his team have shown that miR-217, a tumor suppressor, [17] is deregulated in patients with AML and more importantly in patients with low genetic risk. Other circulating miRNAs also seem of great interest for AML prognosis, namely, miR-210, whose deregulation was observed in patients in complete remission (CR) [20]. Patients expressing miR-210 have a significantly worse overall survival (OS), and those with weak expression of miR-328 have a shorter OS [20,21]. A significantly reduced expression of miR-638 has been shown in AML patients and increased in CR [22]. Another study carried out by Tian C et al. showed that the plasma expression of miR-192 is also increased in AML patients with favorable cytogenetic risk and high OS [23].
Several studies have focused on the profile of peripheral blood circulating miRNAs in acute myeloblastic leukemia. To date, no study has been published on miRNAs in the main microenvironment of AML development, namely, the bone marrow. It remains unknown how miRNAs released in the bone marrow niche in which AML arises are subverted to support leukemic cells and protect them from immunosurveillance. The contribution of the tumor microenvironment (TME) to the survival, spread, and relapse of AML in the bone marrow is crucial in the development, progression, immune response escape, and therapy failure in leukemia. Therefore, the aim of this study was to investigate, for the first time, whether circulating miRNAs display differential expression profiles in the bone marrow of AML patients at diagnosis and first complete remission after treatment in comparison with healthy donors. Such screening will highlight the crucial role of enriched circulating bone marrow miRNAs in AML diagnosis and prognosis and their impact on both disease progression and response to treatment.

2. Results

2.1. Bone Marrow Circulating miRNAs Expression Profile in Newly Diagnosed AML Patients

A few studies have focused on circulating peripheral blood miRNAs, but at this time, no studies have characterized them in the principal compartment of AML disease, namely, the bone marrow. We detected miRNA expression in bone marrow body fluid of AML patients at diagnosis (AMLD) (n = 27), and we compared their profile to that in healthy donors (HD) (n = 11). Moreover, miRNAs with a Ct value > 35 in either the AML group or the control group were excluded from further analysis. Differentially expressed (DE) miRNAs between AMLD and HD samples, of which 61 were upregulated and 37 were downregulated, were identified with a threshold of p < 0.05 and a 1,5-fold change (Table 1).
The clustering of the pretreated AML patients and HD based on the highly significant DE miRNAs (21 up and 18 down p < 0.005 and 2 FC) showed a high separation between the two groups (Figure 1A,B). Interestingly, 16 of these DE miRNAs were undetectable or expressed at very low levels in one of the two groups (miR-520a-3p, miR-548d, miR-449b, miR-49-3p, miR-520f, miR-330-5p, and miR-34c-5p in HD patients and miR-326, miR-198, miR-miR-518d, miR-107, miR-215, miR-504, miR-890, and miR-448 in AML), suggesting their important role in the progression of AML disease.
To validate our data analysis, the levels of the top 46 differentially expressed miRNAs with very high statistical significance were quantified by RT-qPCR in a second independent cohort including 15 AML patient bone marrow aspiration samples. The results showed that 30 of the 46 miRNAs tested and identified as DE in the first cohort were confirmed in the second cohort (Table 2), while 14 were also DE in the same way as in the first cohort but were statistically insignificant. Only two miRNAs out of the 46 tested showed a different profile between the first and second cohorts, suggesting the need to increase the number of patients in our second cohort.
Our results permit us to establish an AML bone marrow circulating miRNA signature consisting of downregulated miR-448, miR-890, miR-504, miR-145, and miR-365 (Figure 2A) and upregulated miR-9, miR-21, miR-629 and miR-449b (Figure 2B), with highly significant AUCs (Figure 2C) and highly differentially expressed fold changes (p-value < 0.005; FC > 2). The combination of these miRNAs has diagnostic value, enabling the identification of AML with high sensitivity and specificity (AUC = 0.98; p-value < 0.0001) (Figure 2D).
This miRNAs signature was also confirmed in the second cohort (Figure 3A–C).

2.2. Functional Analysis of the Pathways of AML Bone Marrow Circulating miRNAs

GO classification and KEGG pathway analyses were used to identify the biological functions of miRNA pathways, including oncogenic processes, cell death pathways, immune functions and proliferation signaling pathways. The threshold of GO and KEGG classification was set as p < 0.05. MicroRNAs in cancer (hsa:05206, p = 6.05 × 10−11), acute myeloid leukemia apoptosis (hsa:05221, p = 3.98 × 10−5), Ras signaling pathway (hsa:05221, p = 2.29 × 10−5) and PI3K-Akt signaling pathway (hsa:04151, p = 1.82×10−4) were the most significantly enriched miRNA target gene pathways. We also observed miRNAs implicated in apoptosis, chemokine and T cell receptor signaling pathways (Table 3A). GO classification analysis of DE miRNAs also showed several biological processes, cellular components, and molecular functions implicated in the regulation of macromolecule metabolic processes, positive cell proliferation, negative regulation of cell death, regulation of apoptosis and immune processes (Table 3B). This suggests the implication of these deregulated circulating miRNAs both in malignant cell proliferation and the immune tumor microenvironment of AML.
The analysis of signaling pathways related to the published data (see the discussion section) showed the involvement of several of these confirmed DE miRNAs, such as miR-21, miR-24 and miR-9, in T cell activation, proliferation and Th2 and Treg polarization. Some of these DE miRNAs are considered oncogenes and are expressed in AML tumor cells, suggesting their role in tumor protection and T cell dysfunction.

2.3. Prognosis Value of Bone Marrow Circulating miRNAs in AML Based on Genetic Risk.

We compared the miRNA profiles of newly diagnosed patients based on their genetic risk classification to highlight the correlation between circulating bone marrow miRNAs and the prognostic value of classical genetic risk. Given the experimental limits and the limited number of patients, we compared two groups of patients, those considered favorable (FAV) (n = 9) and those considered adverse (ADV) (n = 10), according to their prognosis of molecular and genetic risk. The list of miRNAs was filtered with a p-value < 0.05 and fold change ≥ 1.5. Our analysis identified 19 downregulated miRNAs in the favorable group compared to the adverse group, and only miRNA-222 was upregulated (FC = 30; p = 0.04) (Table 4). Interestingly, six of these downregulated miRNAs in the FAV group were also downregulated in all AML patients at diagnosis compared to HD. However, miR-196b and 490-3p were downregulated in both the FAV group and the HD group compared to all AML patients (Table 4). This suggests a link between some DE BM circulating miRNAs and classical genetic risk classification.

2.4. Correlation between Bone Marrow Circulating miRNA Expression Profiles and Overall Survival Rate of Patients with AML

Regarding cytogenetic risk, the AML circulating bone marrow miRNA profile revealed a set of differentially expressed miRNAs between the unfavorable and favorable groups. Regardless of the classic classification factors, we compared patients only on the basis of their overall survival 4 years after diagnosis. Comparison of the two groups of patients at the time of diagnosis showed that 4 miRNAs (miR-328; miR-671-3p; miR-636 and miR-363) were downregulated in patients who remained in remission 4 years after treatment (n = 12) compared to those who died during the first three years after treatment (n = 15) (Figure 4A). We confirmed these downregulated miRNAs in a small second cohort (12 AML patients at diagnosis: 6 AMLDCR vs 6 AMLDR) (Figure 4B). These miRNAs could be used as independent prognostic factors for overall survival. In particular, the prognostic relevance of miR-328 and miR-671-3p expression was more statistically significant in separating the patient group based on their OS than based on their genetic risk, and the combined ROC analyses revealed an AUC of 0.7 in first and 0.92 in second cohort (p < 0.0001), indicating a prognostic value of these 2 miRNAs (Figure 4C). In conclusion, our analysis showed that the expression of circulating bone marrow miRNAs appears to be associated with overall survival risk.

2.5. Bone Marrow Circulating miRNAs Profile of First Complete Remission AML Patients (AMLPT)

To understand the importance of these DE miRNAs described previously in defining the response to treatment, we analyzed and compared the miRNA profiles of twelve paired patients at diagnosis to their profiles during first complete remission (CR1) three months post treatment (PT). Our results showed that 19 miRNAs were upregulated and 69 were downregulated in CR1 AML patients after treatment (AMLPT) compared to newly diagnosed patients (AMLD) (p < 0.05 and 1,5-fold change) (Table 5).
To determine the prognostic value of these miRNAs in determining posttreatment remission, we compared the profiles of miRNAs differentiating AML patients in CR1 (AMLPT) and healthy donors (HD) from those of newly diagnosed patients (AMLD). The results showed that 14 miRNAs are upregulated, and four miRNAs were downregulated in AMLD compared to both AMLPT patients and healthy donors.
Interestingly, the results showed that the bone marrow expression level of miR-518d-3p was also still undetectable in CR1 patients after treatment similar to HD (Figure 5). Overall, this correlation allowed us to establish promising bone marrow circulating miRNA biomarkers for AML remission post treatment, suggesting their possible role in the response of AML patients to chemotherapy treatment.

3. Discussion

Despite advances in the search for new optimal therapeutic approaches, including targeted immunotherapy, only a minority of patients are cured, especially in the elderly population. The identification of new therapeutic targets is necessary to design new or improve existing therapeutic protocols. This is only possible through understanding the different players that drive the pathogenesis of this disease, especially those related to TME. In addition, the search for new diagnostic and prognostic markers of AML is necessary, given the heterogeneity of the disease and the inadequacy of current genetic markers in predicting survival. MiRNAs circulating free or contained in microvesicles (MVs) can be secreted by a wide variety of cells in the TME and are increasingly seen as potential future markers for the diagnosis and prognosis of various malignant diseases.
In this work, we have for the first time analyzed the profile of enriched miRNAs circulating in the bone marrow, which is considered the main location of the initiation and development of acute myeloid leukemia. The results demonstrated a consistent number of differentially expressed miRNAs between AML patients and healthy donors. This allowed us to establish a miRNA signature at the time of diagnosis that provided good separation between the patients and healthy donors. More than thirty of these miRNAs were validated in a second independent cohort of patients. However, two DE miRNAs in the first cohort did not give similar results in the second cohort, of which 14 were similarly DE but statistically insignificant. This could be due to the limited number of patients in the second cohort, which suggests the need to increase the number of patients in the future. Interestingly, some of these DE miRNAs were undetectable or expressed at very low levels either in patients or in HD, suggesting their role in the development of AML. Among the miRNAs that were expressed in HD but undetectable in patients, we found miR-448, miR-890 and miR-504. These miRNAs have been shown to be downregulated and function as tumor suppressors in several types of cancer [24,25]. Recently, miR-504 was shown to be significantly downregulated in the serum of AML patients and cell lines, and its overexpression inhibits cell growth and induces apoptosis in leukemic cells [26]. MiR-448 is downregulated in T-cell acute lymphoblastic leukemia (T-ALL) and seems to target the principal proto-oncogene (TAL1) of this disease [27]. To date, no publication has shown a link between miR-448 and miR-890 in AML, suggesting that their deregulation in bone marrow fluid may be associated with cells other than leukemic blasts. Their deregulation in the TME following the development of the disease must be elucidated for their direct impact not only on the proliferation of malignant cells but also on immune TME cells. Interestingly, it was reported that in human colon cancer, miR-448 could suppress CD8+ T-cell apoptosis and enhance the CD8+ T-cell response by inhibiting indoleamine 2,3-dioxygenase 1 enzyme function [28]. In our results, we also observed other differentially expressed miRNAs, including miR-520a, 651, 520f and 449, that were upregulated in patients and undetectable or expressed at very low levels in HD. Among these, only miR-520a was described to be DE in AML blasts and was associated with a poor prognosis in AML [29], and miR-520f has been reported as an oncogene in human melanoma and promotes malignant cell proliferation [30].
Our results also allowed us to detect miRNAs that are strongly DE but are expressed in both the AML and HD groups. Some, such as miR-9, miR-548d and miR-21, are highly deregulated and are known to be oncogenes in AML or other types of cancer. An increased level of miR-9 has been observed in the bone marrow and peripheral blood of 200 AML patients compared to HD [31]. Chen Tian et al. showed that the knockdown of miR-9 suppressed the proliferation of AML cells by the induction of G0 arrest and apoptosis in vitro, resulting in decreased circulating leukemic cells and prolonged survival in a xenotransplant mouse model [32]. In addition, overexpression of miR-9 enhances the suppressive function of MDSCs, while its knockdown impairs them and inhibits the tumor growth of Lewis lung carcinoma in mice. [33]. MiRNA-548d-5p was shown to be upregulated in metastatic colorectal cancer tumor-derived exosomes [34]. There was no association between these miRNAs and AML in the literature, suggesting that upregulation of AML in bone marrow fluid is due to a cell origin other than leukemic cells. Certain studies have shown its role in promoting osteogenic differentiation of mesenchymal stem cells [35,36]. Surprisingly, we also observed that some AML-upregulated bone marrow circulating miRNAs are reported in the literature as tumor suppressors, including miR-34a, miR-375, miR-576-3p, and miR-597. For example, miR-34a is strongly expressed in AML patient bone marrow fluid and is known to be downregulated in leukemia cells in the marrow and peripheral blood serum of patients [37,38] and appears to play an important role as a tumor suppressor and immunotherapeutic agent [39,40]. The expression of miR-375 was also downregulated in leukemic cell lines and primary AML blasts compared to normal controls, and its overexpression decreased proliferation and colony formation, reduced xenograft tumor size and prolonged the survival time in a leukemia xenograft mouse model [41]. MiR-576-3p was shown to be downregulated in colorectal [42] and bladder cancer [43], to inhibit the migration and invasion of lung adenocarcinoma [44] and to significantly inhibit the migration and pro-angiogenic abilities of glioma cells under hypoxic conditions [45]. Additionally, miR-597 inhibits breast and colon cancer cell proliferation, migration and invasion [46,47]. These findings strengthen our previous hypothesis for a cell origin other than leukemia cells for these miRNAs and suggest their dual function according to the type of cancer or the tumor focus. Interestingly, recent analyses of the regulatory network of CD19-CAR-T immunotherapy for B acute lymphocyte leukemia revealed that microRNA miR-375 could regulate the crosstalk between the genes encoding transcription factors and histones involved in CD19-CAR-T therapy [48].
Our results also showed that some miRNAs were expressed in both the AML and HD groups but were significantly downregulated in AML patients compared to HD, among which we found miR-145, 150, 204, 365 and 518b. MiR-145 has been increasingly identified as a critical suppressor of carcinogenesis and therapeutic resistance in several cancers [49]. Recent data showed that the level of this miRNA in serum and bone marrow mononuclear cells of AML patients was significantly lower than that of HD and was related to poor prognosis [50]. MiR-145 seems to play an important role in the immune response against leukemic cells. It was reported to enhance host antitumor immunity by altering the cytokine milieu, metastatic microenvironment and reprogramming tumor-associated myeloid cells [51]. Recent work revealed that miR-145, which is known to be downregulated by cisplatin in cisplatin-resistant ovarian cancer cells, also represses PD-L1 gene expression and induces T cell apoptosis in vitro [52]. It is known that the level of miR-150 is decreased in the plasma and blast cells of AML patients at diagnosis and that reintroducing miR-150 expression induces myeloid differentiation and inhibits the proliferation of AML cells [18,53]. Xi et al. developed a novel targeted therapeutic strategy using FLT3L-guided miR-150-based nanoparticles to treat FLT3-overexpressing AML in an animal model with high efficacy and minimal side effects [54]. MiR-204 expression in AML patients was decreased and was associated with shorter patient survival. Higher expression of this miR was observed in patients after induction therapy and was correlated with complete remission [55], and its targeting promotes the viability and invasion of AML cells [56]. Wang et al. demonstrated that overexpression of these miRNAs potentiates the sensitivity of AML cells to arsenic trioxide [57]. In addition, recent work showed that in breast cancer, miR-204 regulated the expression of key cytokines in tumor cells and reprogrammed immune cells by shifting myeloid and lymphocyte populations, suggesting that this miRNA suppresses tumor metastasis and remodeling the immune microenvironment [58]. MiR-518b was also reported to function as a tumor suppressor in glioblastoma [56], esophageal and squamous cell carcinoma [59].
Based on these results, we also established an AML signature at diagnosis, consisting of nine miRNAs with an AUC greater than 0.77. Combined ROC analyses using these nine miRNAs revealed an elevated AUC of 0.98 (p < 0.0001), indicating an additive effect on the diagnostic value of these miRNAs. This signature, as well as more than thirty DE miRNAs, was validated in a second cohort of patients and provided good separation between AML patients and HD. Our patient group comparison at diagnosis based on their overall survival showed that miR-328, miR-671-3p, miR-636 and miR-363 are upregulated in relapsed patients. These miRs are described in the literature as promising prognostic biomarkers in AML or other malignant diseases [21,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75]. Interestingly, all the patients involved in this comparison were not grafted, which suggests that these four miRs could constitute a signature to guide the choice of the best treatment strategy, especially the utility of grafting patients at diagnosis. A recent study using a TCGA dataset identified a set of miRNAs, including miR-363, that could predict clinical outcome in a heterogeneous AML population [76]. This study confirms our results showing the high expression of circulating bone marrow miR-363 in nontransplanted patients with shorter OS and suggests that its high expression may help to identify patients recommended for an early allo-HSCT regimen. Interestingly, this miRNA seems to play an important role in the immune response against leukemic cells. An enrichment of specific cellular miRNAs, including miR-363, in EVs derived from CD40/IL-4-stimulated chronic lymphoblastic leukemia cells was observed, and autologous patient CD4(+) T cells were found to be capable of internalizing the CLL-EVs that target the immunomodulatory molecule CD69 [77].
Our comparison of patients before and after three months of treatment (first complete remission (CR1)) allowed us to identify a set of miRNAs that were differentially expressed (4 downregulated and 14 upregulated) in the same way as when comparing patients at diagnosis with HD. However, other analyses are underway on a larger cohort of patients to study the long-term prognostic value (four years after treatment) of these miRNAs. It should also be noted that an analysis of the profile of relapsing patients during the first months is also necessary to validate these miRNAs, which we could not obtain in our study. These miRNAs, in addition to their value in monitoring posttreatment remission, may also play a role in the elimination of blasts directly or indirectly by improving the antileukemia response. Among these DE in AML CR1, miR-484, miR-518d and miR-99b are upregulated in addition to the previously described miR-328. MiR-99b was also showed as being responsible for the conversion of monocytes into MDSCs and to be associated with resistance to treatment with immune checkpoint inhibitors in melanoma patients [78]. MiR-518d may be an interesting target due to its low or undetectable level in HD and post treatment. It was reported to be implicated in the inhibition of cervical cancer cell proliferation, migration and invasion [79] and in the regulation of advanced small cell lung cancer cell proliferation, migration and chemotherapy response [80]. To date, little work has focused on this miRNA, and none of the studies have reported its involvement in AML, which makes it a good avenue for future investigation. For the downregulated miRNAs in AML CR1, miR-24 appear to be involved in AML evolution an immune response. High expression of miR-24 has been observed in AML leukemia patients and is associated with the risk of relapse and poor survival [81], and its role in nasopharyngeal carcinoma pathogenesis by mediating T-cell suppression has been demonstrated [82]. In a previous study, we demonstrated that this miRNA negatively regulates IFN-γ expression in T lymphocytes [83], and recently, we showed its direct impact on AML T lymphocyte fragility and dysfunction (in preparation). Furthermore, other downregulated miRNAs in CR1, such as miR-15b [84], miR-27a [85], miR-29c [86], miR-106a [87] and miR-181a [88], have been described as being implicated in AML pathogenesis or drug resistance.
As reported, high expression of miR-181a in AML blasts, cell lines and serum has been observed [89,90], and its overexpression significantly enhanced cell proliferation and increased the ratio of S-phase cells by regulating the tumor suppressor ATM [91,92]. Inhibition of this miR in mice implanted with AML CD34+ hematopoietic progenitor cells (HSPCs) increased myeloid differentiation, inhibited engraftment and infiltration of leukemic CD34+ cells into the bone marrow and spleen, and finally reduced leukemic symptoms [93]. However, miR-181a also plays a role in the sensitization of leukemic resistant cells to daunorubicin and NK cells and may provide a promising option for AML immunotherapy treatment of chemoresistant blasts [94]. MiR-181a appears to play an important role in T cell activation [95] and CAR-T cell immunotherapy [96]. A recent study showed that inhibition of DOT1L reduces the expression of miR-181a, which in turn selectively enhances low-avidity T cell responses and attenuates graft versus host development, suggesting that this would allow the safe and effective use of allogeneic antitumor T cells [97]. In melanoma patients, the plasma levels of miR-181a were higher in patients at diagnosis compared to controls, and the development of metastasis has been associated with changes in immune effector and regulatory cells with changes in plasma and cellular levels of immune regulatory miRNA [98]. Melanoma-derived exosomes decrease TNFα secretion in CD8+ cells and are enriched for hsa-miR-181a, which is capable of interacting directly with the 3′-UTR sequence of TNFα and driving immune escape in melanoma [99]. We have previously shown in our laboratory that overexpression of miR-181a negatively regulates IFN-γ expression in activated PB CD4+ T cells by directly binding to its target sites in the mRNA and increases the expression of IL4 [83]. MiR-181c also seems to play a dual role similar to miR-181a in both malignant and immune cells. A study conducted by R Su et al. compared patients with AML blasts with normal controls, and most AML patients showed significantly increased expression levels of all miR-181 members, including miR-181c [100]. MiR-181c is important in T cell activation, and the overexpression of miR-181c results in inhibition of T cell activation and actin polymerization coupled with defective immunological synapse formation [101]. Interestingly, R. Le Dieu et al. showed that the ability of AML T cells to form immune synapses was significantly impaired [102]. It was identified as a novel miRNA that promotes Th17 cell differentiation and autoimmunity [103]. In addition, circulating miR-181c can be of a cell origin other than cancer cells. Xiao Li et al. showed that exosomes derived from human umbilical cord mesenchymal stem cells are enriched for miR-181c, which in turn attenuates burn-induced excessive inflammation [104].
MiR-532 was reported as an oncogene in breast [105], colorectal [106] and human gastric cancers [107]. In recent published work, Lin et al. concluded that plasma exosome-derived miR-532 can be used as a novel survival predictor for acute myeloid leukemia [108]. This miRNA has also been implicated in the immune inflammatory response [109]. In addition to its function as a tumor suppressor and/or oncogene [110], miR-146a was shown to play a critical role in regulating T cell functions by providing a general brake on proliferation and activation of immune cells and was able to limit immune responses linked to the TH1 and TH17 subsets. Mice lacking this miRNA developed more severe TH17 responses in murine models of autoimmunity, with increased production of IFN-γ and IL-17 and reduced secretion of IL-4 [111]. Furthermore, miR-146a is also a key regulator of Treg cell biology, as deficiency of miR-146a leads to impaired function of Treg cells and consequently to the breakdown of immunological tolerance and to the development of fatal immune-mediated lesions depending on IFN-γ. Indeed, in addition to their role in the proliferation of cancer cells, many of these downregulated miRNAs, including miR-15 [112,113], miR-27a [114,115] and miR-29c [116,117,118], appear to play a role in immune regulation, T regulatory and inflammatory responses and TIL dysfunction.
Together, these results suggest that enriched circulating bone marrow miRNAs are key regulators of AML blast proliferation and affect immune cell functions in the TME. Our GO classification and KEGG pathway analyses confirmed that these DE circulating miRNAs could play a role not only in the oncogenic process, cell death and proliferation signaling pathways but also in immune function. Further research may elucidate their role in the malignant cells and TME. Studies are currently underway in our laboratory to investigate the impact of these miRNAs on the antileukemic immune response with the aim of establishing new immunotherapeutic targets of AML.

4. Materials and Methods

4.1. Patients

Patients used in this study had a newly diagnosed AML in addition to being in complete remission after the first treatment (three months after first induction) as determined by bone marrow test. A total of 46 patients at the time of diagnosis, 12 of them in complete remission post treatment, provided bone marrow samples. Healthy subjects (n =11) were enrolled as negative controls. None of these controls had been previously diagnosed with any type of malignancy or other benign disease. Informed consent, approved by the Clinical Research Ethics Committee of Jules Bordet Institute (approval code: B079201214544, approval date: 5 July 2012) was obtained from each participant. Details of the clinical data are provided in Table 6.

4.2. Bone Marrow Aspiration Sampling and RNA Extraction.

At presentation, bone marrow aspiration samples for miRNA detection were collected in EDTA-K2 tubes and processed within 1 h of collection. Bone marrow body fluid samples were centrifuged at 1200× g for 10 min at 4 °C to pellet the hematopoietic cells, and the supernatant was transferred into microcentrifuge tubes, followed by a second centrifugation at 12,000× g for 10 min at 4 °C. The supernatant was transferred to RNase/DNase-free tubes and stored at −80 °C. Total RNA was isolated from the plasma using a mirVana PARIS isolation kit (Ambion, Austin, TX, USA) according to the manufacturer’s instructions for plasma samples. Briefly, 400 μL of human bone marrow plasma was used to extract total RNA. Each sample was eluted in 100 μL of RNase-free water and concentrated to a final volume of 20 μL by using an Eppendorf Concentrator Plus 5301 (Eppendorf, Aarschot, Belgium). The RNA sample concentration was quantified by a NanoDrop ND-1000 (Thermo Fisher Scientific, Waltham, MA, USA).

4.3. miRNA Expression Profile

Thirty nanograms of total bone marrow fluid extracted RNA was used for cDNA synthesis from the miRNAs using a TaqMan® microRNA Reverse Transcription Kit (#4366596; Applied Biosystems, Waltham, MA, USA) and Megaplex RT primers (Human Pool A, #4399966; Applied Biosystems) following the manufacturer’s protocol described previously [18]. After the preamplification step, the products were amplified and then loaded into TaqMan Human MicroRNA Array A (#4398965; Applied Biosystems), which contains the TaqMan primers and probes in each well for the 380 different mature human miRNAs; miR-425 transcript was used as a normalization signal. The relative expression levels of miRNAs were calculated using the comparative ΔΔCt method as described previously [18]. The fold changes in miRNAs were calculated by the equation 2−ΔΔCt.

4.4. TaqMan miRNA Assay for Individual miRNAs

The expression of individual miRs was determined using the TaqMan miRNA assay as described previously [18]. The expression levels of miRNAs were calculated as described previously, and miR-425 was used as an internal reference. The predesigned TaqMan assay primers and probes corresponding to the ones used in TLDA card A were purchased from Thermo Fisher Scientific. The calculated delta Ct ± SD for the patients was compared with the delta Ct ± SD (SD stands for the standard deviation of the average delta Ct of the group for the healthy control group and tested for statistical significance).

4.5. Statistical Analysis

Data analysis was performed in R version 3.5.3 using packages of the Bioconductor project (DOI: 10.1186/gb-2004-5-10-r80). Raw cycle threshold (Ct) values were loaded into R using the HTqPRC package (doi:10.1093/bioinformatics/btp578) and flagged as “Undetermined” if they were above 35. miRNAs that are expressed in at least one sample group (i.e., Ct > 35 in at least X samples) were retained for further analysis. To normalize the data, delta Ct (ΔCt) values were calculated for each sample by subtracting the Ct value of miR-425 from its other values. miR-425 was selected as the best reference candidate by both the geNorm and NormFinder methods implemented in the NormqPCR package (https://rdrr.io/bioc/NormqPCR/man/NormqPCR-package.html) accessed on 1 August 2020. Testing for differential expression between sample groups was performed using empirical Bayes moderated t-statistics computed by the limma package (https://bioconductor.org/packages/release/bioc/html/limma.html). miRNAs with an absolute fold change > 1.5 and a p value < 0.05 were considered differentially expressed (DEmiRNAs). The ability of the selected signatures and miRNAs to accurately discriminate between different patient groups was estimated by constructing and assessing the performance of ensemble-based classifiers (DOI 10.1007/s10462-009-9124-7). These are stacked classifiers that combine the predictions of several classification algorithms in order to predict the outcome. The Area Under Receiver Operating Characteristic Curve (AUC) was calculated by a bootstrapping procedure with 100 iterations. Enrichment analysis of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (https://www.genome.jp/kegg/pathway.html) was performed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) tool (http://david.abcc.ncifcrf.gov/) accessed on 13 August 2020.

5. Conclusions

In our study, for the first time, we highlight circulating miRNAs in the bone marrow of AML patients and provide important insight into the possible role of these miRNAs in the TME. We have shown that the main TME of AML is well enriched in molecules that have variable expression according to the progression of the disease as well as the predisposition to the treatment response. In addition to the value that these circulating miRNAs can provide for the diagnosis of AML or its confirmation, their contribution could extend to the prognostic level by guiding, at diagnosis, the choice of transplants for certain patients. Some of these miRNAs appear to be of malignant origin, and their impact on resistance to treatment or evasion of immune system surveillance has been described in other types of cancer. Other miRNAs that to date have never been described in AML could have another cellular origin (immune cells, MSCS and stromal cells) and could be at the origin of the promotion of the proliferation of blasts while directing the immune response to a protumoral phenotype. Further in vitro and in vivo investigations are necessary to confirm these hypotheses, which open a wide door to new immunotherapeutic targets for the eradication of AML.

Author Contributions

Conceptualization, D.M.A., R.R., M.N., P.L. and M.M.; investigation, D.M.A., R.R., M.N., N.N., H.F.-K., and F.B.; resources, P.L., and M.M.; writing-original/draft preparation, D.M.A., M.N., N.N., H.F.-K., F.B. and M.M.; review and editing, D.M.A., M.N., and M.M.; supervision, M.M.; project administration, D.B., N.M., P.L., and M.M.; funding acquisition, P.L. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Les Amis de l’Institut Bordet, Le Fonds National de la Recherche Scientifique (FNRS, Télévie).

Conflicts of Interest

The authors have no conflicts of interest to declare. They confirm that there are no conflicts of interest associated with this publication. The manuscript has been read and approved by all named authors.

References

  1. Thomas, D.; Majeti, R. Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017, 129, 1577–1585. [Google Scholar] [CrossRef]
  2. Shysh, A.C.; Nguyen, L.T.; Guo, M.; Vaska, M.; Naugler, C.; Rashid-Kolvear, F. The incidence of acute myeloid leukemia in Calgary, Alberta, Canada: A retrospective cohort study. BMC Public Health 2017, 18, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. 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] [Green Version]
  4. Watts, J.; Nimer, S. Recent advances in the understanding and treatment of acute myeloid leukemia. F1000Research 2018, 7, 1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Marcucci, G.; Mrozek, K.; Radmacher, M.D.; Garzon, R.; Bloomfield, C.D. The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood 2011, 117, 1121–1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. He, C.; Luo, B.; Jiang, N.; Liang, Y.; He, Y.; Zeng, J.; Liu, J.; Zheng, X. OncomiR or antioncomiR: Role of miRNAs in Acute Myeloid Leukemia. Leuk. Lymphoma 2019, 60, 284–294. [Google Scholar] [CrossRef]
  7. Trino, S.; Lamorte, D.; Caivano, A.; Laurenzana, I.; Tagliaferri, D.; Falco, G.; Del Vecchio, L.; Musto, P.; De Luca, L. MicroRNAs as New Biomarkers for Diagnosis and Prognosis, and as Potential Therapeutic Targets in Acute Myeloid Leukemia. Int. J. Mol. Sci 2018, 19, 460. [Google Scholar] [CrossRef] [Green Version]
  8. Dohner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Buchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar]
  9. Christopher, A.F.; Kaur, R.P.; Kaur, G.; Kaur, A.; Gupta, V.; Bansal, P. MicroRNA therapeutics: Discovering novel targets and developing specific therapy. Perspect. Clin. Res. 2016, 7, 68–74. [Google Scholar]
  10. Ling, H.; Fabbri, M.; Calin, G.A. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat. Rev. Drug Discov. 2013, 12, 847. [Google Scholar]
  11. Si, W.; Shen, J.; Zheng, H.; Fan, W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin. Epigenet. 2019, 11, 25. [Google Scholar]
  12. Slaby, O.; Laga, R.; Sedlacek, O. Therapeutic targeting of non-coding RNAs in cancer. Biochem. J. 2017, 474, 4219–4251. [Google Scholar] [PubMed]
  13. Ferreira, P.; Roela, R.A.; Lopez, R.V.M.; Del Pilar Estevez-Diz, M. The prognostic role of microRNA in epithelial ovarian cancer: A systematic review of literature with an overall survival. Oncotarget 2020, 11, 1085–1095. [Google Scholar]
  14. Buhagiar, A.; Borg, J.; Ayers, D. Overview of current microRNA biomarker signatures as potential diagnostic tools for leukaemic conditions. Noncoding RNA Res. 2020, 5, 22–26. [Google Scholar]
  15. Hamam, R.; Hamam, D.; Alsaleh, K.A.; Kassem, M.; Zaher, W.; Alfayez, M.; Aldahmash, A.; Alajez, N.M. Circulating microRNAs in breast cancer: Novel diagnostic and prognostic biomarkers. Cell Death Dis. 2017, 8, e3045. [Google Scholar]
  16. Dufresne, S.; Rebillard, A.; Muti, P.; Friedenreich, C.M.; Brenner, D.R. A Review of Physical Activity and Circulating miRNA Expression: Implications in Cancer Risk and Progression. Cancer Epidemiol. Biomark. Prev. 2018, 27, 11–24. [Google Scholar]
  17. Yan, J.; Wu, G.; Chen, J.; Xiong, L.; Chen, G.; Li, P. Downregulated miR-217 expression predicts a poor outcome in acute myeloid leukemia. Cancer Biomark. 2018, 22, 73–78. [Google Scholar] [CrossRef]
  18. Fayyad-Kazan, H.; Bitar, N.; Najar, M.; Lewalle, P.; Fayyad-Kazan, M.; Badran, R.; Hamade, E.; Daher, A.; Hussein, N.; ElDirani, R.; et al. Circulating miR-150 and miR-342 in plasma are novel potential biomarkers for acute myeloid leukemia. J. Transl. Med. 2013, 11, 31. [Google Scholar] [CrossRef] [Green Version]
  19. Zhi, F.; Cao, X.; Xie, X.; Wang, B.; Dong, W.; Gu, W.; Ling, Y.; Wang, R.; Yang, Y.; Liu, Y. Identification of circulating microRNAs as potential biomarkers for detecting acute myeloid leukemia. PLoS ONE 2013, 8, e56718. [Google Scholar] [CrossRef] [Green Version]
  20. 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. [Google Scholar]
  21. Liu, L.; Chen, R.; 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] [Green Version]
  22. Liu, X.; Li, P.; Yun, Y.; Zhang, D.; Ma, H.; He, Q.; Jia, G.; Peng, J. Prognostic value of plasma miR-638 in patients with acute myeloid leukemia. Int. J. Clin. Exp. Pathol. 2017, 10, 550–555. [Google Scholar]
  23. Tian, C.; Zhang, L.; Li, X.; Zhang, Y.; Li, J.; Chen, L. Low miR-192 expression predicts poor prognosis in pediatric acute myeloid leukemia. Cancer Biomark. 2018, 22, 209–215. [Google Scholar]
  24. Li, B.; Ge, L.; Li, M.; Wang, L.; Li, Z. miR-448 suppresses proliferation and invasion by regulating IGF1R in colorectal cancer cells. Am. J. Transl. Res. 2016, 8, 3013–3022. [Google Scholar] [PubMed]
  25. Wang, C.; Xu, C.; Niu, R.; Hu, G.; Gu, Z.; Zhuang, Z. MiR-890 inhibits proliferation and invasion and induces apoptosis in triple-negative breast cancer cells by targeting CD147. BMC Cancer 2019, 19, 577. [Google Scholar]
  26. Li, S.M.; Zhao, Y.Q.; Hao, Y.L.; Liang, Y.Y. Upregulation of miR-504-3p is associated with favorable prognosis of acute myeloid leukemia and may serve as a tumor suppressor by targeting MTHFD2. Eur. Rev. Med. Pharm. Sci. 2019, 23, 1203–1213. [Google Scholar]
  27. Correia, N.C.; Melao, A.; Povoa, V.; Sarmento, L.; Gomez de Cedron, M.; Malumbres, M.; Enguita, F.J.; Barata, J.T. microRNAs regulate TAL1 expression in T-cell acute lymphoblastic leukemia. Oncotarget 2016, 7, 8268–8281. [Google Scholar]
  28. Lou, Q.; Liu, R.; Yang, X.; Li, W.; Huang, L.; Wei, L.; Tan, H.; Xiang, N.; Chan, K.; Chen, J.; et al. miR-448 targets IDO1 and regulates CD8(+) T cell response in human colon cancer. J. Immunother. Cancer 2019, 7, 210–214. [Google Scholar]
  29. Gao, H.Y.; Wang, W.; Luo, X.G.; Jiang, Y.F.; He, X.; Xu, P.; Chen, X.; Li, X.Y. Screening of prognostic risk microRNAs for acute myeloid leukemia. Hematology 2018, 23, 747–755. [Google Scholar]
  30. Sun, M.X.; An, Q.; Chen, L.M.; Guo, L. MIR-520f Regulated Itch Expression and Promoted Cell Proliferation in Human Melanoma. Cells. Dose Response 2020, 18, 1559325820918450. [Google Scholar]
  31. Zheng, H.Z.; Sun, Y.B.; Zhan, T.; Zeng, K.D.; Liu, C.Z.; Zhang, J.L. miR-9-5p prompts malignancies of acute myeloid leukemia cells mainly by targeting p27. Int. J. Clin. Exp. Med. 2018, 11, 2076–2083. [Google Scholar]
  32. Chen, L.; Hu, W.; Li, G.; Guo, Y.; Wan, Z.; Yu, J. Inhibition of miR-9-5p suppresses prostate cancer progress by targeting StarD13. Cell Mol. Biol. Lett. 2019, 24, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tian, J.; Rui, K.; Tang, X.; Ma, J.; Wang, Y.; Tian, X.; Zhang, Y.; Xu, H.; Lu, L.; Wang, S. MicroRNA-9 Regulates the Differentiation and Function of Myeloid-Derived Suppressor Cells via Targeting Runx1. J. Immunol. 2015, 195, 1301–1311. [Google Scholar] [CrossRef] [Green Version]
  34. Tang, Y.; Zhao, Y.; Song, X.; Song, X.; Niu, L.; Xie, L. Tumor-derived exosomal miRNA-320d as a biomarker for metastatic colorectal cancer. J. Clin. Lab. Anal. 2019, 33, e23004. [Google Scholar] [CrossRef] [Green Version]
  35. Zhang, W.; Zhang, L.; Zhou, Y.; Ji, X.; Liu, J.; Liu, D.; Yin, P.; Peng, Y.; Hao, M.; Zhang, L.; et al. Synergistic Effects of BMP9 and miR-548d-5p on Promoting Osteogenic Differentiation of Mesenchymal Stem Cells. Biomed. Res. Int. 2015, 2015, 309747. [Google Scholar] [CrossRef] [Green Version]
  36. Sun, J.; Wang, Y.; Li, Y.; Zhao, G. Downregulation of PPARgamma by miR-548d-5p suppresses the adipogenic differentiation of human bone marrow mesenchymal stem cells and enhances their osteogenic potential. J. Transl Med. 2014, 12, 168. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, X.; Li, H. Diagnostic Value of miR-34a in Bone Marrow Mononuclear Cells of Acute Myeloid Leukemia Patients. Clin. Lab. 2020, 66. [Google Scholar] [CrossRef]
  38. Huang, Y.; Zou, Y.; Lin, L.; Ma, X.; Chen, H. Identification of serum miR-34a as a potential biomarker in acute myeloid leukemia. Cancer Biomark. 2018, 22, 799–805. [Google Scholar] [CrossRef]
  39. Liu, L.; Ren, W.; Chen, K. MiR-34a Promotes Apoptosis and Inhibits Autophagy by Targeting HMGB1 in Acute Myeloid Leukemia Cells. Cell Physiol. Biochem. 2017, 41, 1981–1992. [Google Scholar] [CrossRef]
  40. Wang, X.; Li, J.; Dong, K.; Lin, F.; Long, M.; Ouyang, Y.; Wei, J.; Chen, X.; Weng, Y.; He, T.; et al. Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cell Signal. 2015, 27, 443–452. [Google Scholar] [CrossRef]
  41. Bi, L.; Zhou, B.; Li, H.; He, L.; Wang, C.; Wang, Z.; Zhu, L.; Chen, M.; Gao, S. A novel miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry contributes to leukemogenesis in acute myeloid leukemia. BMC Cancer 2018, 18, 182. [Google Scholar]
  42. Eldaly, M.N.; Metwally, F.M.; Shousha, W.G.; El-Saiid, A.S.; Ramadan, S.S. Clinical Potentials of miR-576-3p, miR-613, NDRG2 and YKL40 in Colorectal Cancer Patients. Asian Pac. J. Cancer Prev. 2020, 21, 1689–1695. [Google Scholar] [PubMed]
  43. Meng, F.M.; Meng, F.M.; Song, X.L. MiR-576-3p is a novel marker correlated with poor clinical outcome in bladder cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 973–977. [Google Scholar]
  44. Greenawalt, E.J.; Edmonds, M.D.; Jain, N.; Adams, C.M.; Mitra, R.; Eischen, C.M. Targeting of SGK1 by miR-576-3p Inhibits Lung Adenocarcinoma Migration and Invasion. Mol. Cancer Res. 2019, 17, 289–298. [Google Scholar] [PubMed] [Green Version]
  45. Hu, Q.; Liu, F.; Yan, T.; Wu, M.; Ye, M.; Shi, G.; Lv, S.; Zhu, X. MicroRNA5763p inhibits the migration and proangiogenic abilities of hypoxiatreated glioma cells through hypoxiainducible factor1alpha. Int. J. Mol. Med. 2019, 43, 2387–2397. [Google Scholar]
  46. He, J.; Mai, J.; Li, Y.; Chen, L.; Xu, H.; Zhu, X.; Pan, Q. miR-597 inhibits breast cancer cell proliferation, migration and invasion through FOSL2. Oncol Rep. 2017, 37, 2672–2678. [Google Scholar]
  47. Li, S.; Liu, Z.; Fang, X.D.; Wang, X.Y.; Fei, B.Y. MicroRNA (miR)-597-5p Inhibits Colon Cancer Cell Migration and Invasion by Targeting FOS-Like Antigen 2 (FOSL2). Front. Oncol. 2019, 9, 495. [Google Scholar] [PubMed] [Green Version]
  48. Zhang, Q.; Hu, H.; Chen, S.Y.; Liu, C.J.; Hu, F.F.; Yu, J.; Wu, Y.; Guo, A.Y. Transcriptome and Regulatory Network Analyses of CD19-CAR-T Immunotherapy for B-ALL. Genom. Proteom. Bioinform. 2019, 17, 190–200. [Google Scholar]
  49. Xu, W.; Hua, Y.; Deng, F.; Wang, D.; Wu, Y.; Zhang, W.; Tang, J. MiR-145 in cancer therapy resistance and sensitivity: A comprehensive review. Cancer Sci. 2020, 111, 3122–3131. [Google Scholar] [CrossRef]
  50. Shi, Q.; Xing, G.; Qi, M.; Xing, Y. Lower Serum miR-145 Predicts Poor Prognosis in Patients with Acute Myeloid Leukemia. Clin. Lab. 2020, 66, 7754. [Google Scholar]
  51. Ishii, H.; Vodnala, S.K.; Achyut, B.R.; So, J.Y.; Hollander, M.C.; Greten, T.F.; Lal, A.; Yang, L. miR-130a and miR-145 reprogram Gr-1+CD11b+ myeloid cells and inhibit tumor metastasis through improved host immunity. Nat. Commun. 2018, 9, 2611. [Google Scholar] [CrossRef] [PubMed]
  52. Sheng, Q.; Zhang, Y.; Wang, Z.; Ding, J.; Song, Y.; Zhao, W. Cisplatin-mediated down-regulation of miR-145 contributes to up-regulation of PD-L1 via the c-Myc transcription factor in cisplatin-resistant ovarian carcinoma cells. Clin. Exp. Immunol. 2020, 200, 45–52. [Google Scholar] [CrossRef] [PubMed]
  53. Morris, V.A.; Cummings, C.L.; Korb, B.; Boaglio, S.; Oehler, V.G. Deregulated KLF4 Expression in Myeloid Leukemias Alters Cell Proliferation and Differentiation through MicroRNA and Gene Targets. Mol. Cell. Boil. 2016, 36, 559–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jiang, X.; Bugno, J.; Hu, C.; Yang, Y.; Herold, T.; Qi, J.; Chen, P.; Gurbuxani, S.; Arnovitz, S.; Strong, J.; et al. Eradication of acute myeloid leukemia with FLT3 ligand-targeted miR-150 nanoparticles. Cancer Res. 2016, 76, 4470–4480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Butrym, A.; Rybka, J.; Baczynska, D.; Tukiendorf, A.; Kuliczkowski, K.; Mazur, G. Low expression of microRNA-204 (miR-204) is associated with poor clinical outcome of acute myeloid leukemia (AML) patients. J. Exp. Clin. Cancer Res. 2015, 34, 68. [Google Scholar] [CrossRef] [Green Version]
  56. Xue, F.; Che, H. The long non-coding RNA LOC285758 promotes invasion of acute myeloid leukemia cells by down-regulating miR-204-5p. FEBS Open Bio. 2020, 10, 734–743. [Google Scholar] [PubMed] [Green Version]
  57. Xu, X.; Zhang, F.; Chen, X.; Ying, Q. MicroRNA-518b functions as a tumor suppressor in glioblastoma by targeting PDGFRB. Mol. Med. Rep. 2017, 16, 5326–5332. [Google Scholar] [CrossRef] [PubMed]
  58. Hong, B.S.; Ryu, H.S.; Kim, N.; Kim, J.; Lee, E.; Moon, H.; Kim, K.H.; Jin, M.-S.; Kwon, N.H.; Kim, S.; et al. Tumor suppressor microRNA-204-5p regulates growth, metastasis, and immune microenvironment remodeling in breast cancer. Cancer Res. 2019, 79, 1520–1534. [Google Scholar] [CrossRef] [PubMed]
  59. Zhang, M.; Zhou, S.; Zhang, L.; Zhang, J.; Cai, H.; Zhu, J.; Huang, C.; Wang, J. miR-518b is down-regulated, and involved in cell proliferation and invasion by targeting Rap1b in esophageal squamous cell carcinoma. FEBS Lett. 2012, 586, 3508–3521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Di Meo, A.; Brown, M.D.; Finelli, A.; Jewett, M.A.; Diamandis, E.P.; Yousef, G.M. Prognostic urinary miRNAs for the assessment of small renal masses. Clin. Biochem. 2020, 75, 15–22. [Google Scholar]
  61. Xia, W.; Gong, D.; Qin, X.; Cai, Z. MicroRNA-671-3p suppresses proliferation and invasion of breast cancer cells by targeting DEPTOR. Nan Fang Yi Ke Da Xue Xue Bao 2020, 40, 42–48. [Google Scholar] [PubMed]
  62. Yao, Y.; Zhou, Y.; Fu, X. miR-671-3p is downregulated in non-small cell lung cancer and inhibits cancer progression by directly targeting CCND2. Mol. Med. Rep. 2019, 19, 2407–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Li, Z.; Zhang, Z.; Bi, H.; Zhang, Q.; Zhang, S.; Zhou, L.; Zhu, X.; Zhou, J. Upregulated microRNA-671-3p promotes tumor progression by suppressing forkhead box P2 expression in non-small-cell lung cancer. Mol. Med. Rep. 2019, 20, 3149–3159. [Google Scholar] [CrossRef] [PubMed]
  64. Lu, G.-F.; You, C.-Y.; Chen, Y.-S.; Jiang, H.; Zheng, X.; Tang, W.-W.; Wang, X.-Y.; Xu, H.-Y.; Geng, F. MicroRNA-671-3p promotes proliferation and migration of glioma cells via targeting CKAP4. OncoTargets Ther. 2018, 11, 6217–6226. [Google Scholar] [CrossRef] [Green Version]
  65. Zhu, Z.; Wen, Y.; Xuan, C.; Chen, Q.; Xiang, Q.; Wang, J.; Liu, Y.; Luo, L.; Zhao, S.; Deng, Y.; et al. Identifying the key genes and microRNAs in prostate cancer bone metastasis by bioinformatics analysis. FEBS Open Bio. 2020, 10, 674–688. [Google Scholar] [CrossRef]
  66. Hu, Q.L.; Xu, Z.P.; Lan, Y.F.; Li, B. miR-636 represses cell survival by targeting CDK6/Bcl-2 in cervical cancer. Kaohsiung J. Med. Sci. 2020, 36, 328–335. [Google Scholar] [CrossRef] [Green Version]
  67. Erdogan, B.; Facey, C.; Qualtieri, J.; Tedesco, J.; Rinker, E.; Isett, R.B.; Tobias, J.; Baldwin, D.A.; Thompson, J.E.; Carroll, M.; et al. Diagnostic microRNAs in myelodysplastic syndrome. Exp. Hematol. 2011, 39, 915–926. [Google Scholar] [CrossRef]
  68. Zhao, Y.J.; Song, X.; Niu, L.; Tang, Y.; Song, X.; Xie, L. Circulating Exosomal miR-150-5p and miR-99b-5p as Diagnostic Biomarkers for Colorectal Cancer. Front. Oncol. 2019, 9, 1129. [Google Scholar] [CrossRef] [Green Version]
  69. Wang, Z.; Zhao, Z.; Yang, Y.; Luo, M.; Zhang, M.; Wang, X.; Liu, L.; Hou, N.; Guo, Q.; Song, T.; et al. MiR-99b-5p and miR-203a-3p Function as Tumor Suppressors by Targeting IGF-1R in Gastric Cancer. Sci. Rep. 2018, 8, 10119. [Google Scholar] [CrossRef] [Green Version]
  70. Liu, C.-J.; Huang, F.-Z.; Yang, J.-H.; Liu, C.-P.; Mao, X.-H.; Yi, W.-M.; Shen, X.-B.; Peng, C.; Chen, M.-F.; Jiang, B.; et al. The role of miR-99b in mediating hepatocellular carcinoma invasion and migration. Eur. Rev. Med Pharmacol. Sci. 2018, 22, 2273–2281. [Google Scholar]
  71. Chen, S.; Chen, Y.; Zhu, Z.; Tan, H.; Lu, J.; Qin, P.; Xu, L. Identification of the key genes and microRNAs in adult acute myeloid leukemia with FLT3 mutation by bioinformatics analysis. Int. J. Med Sci. 2020, 17, 1269–1280. [Google Scholar] [PubMed]
  72. Ma, X.; Jin, L.; Lei, X.; Tong, J.; Wang, R. MicroRNA-363-3p inhibits cell proliferation and induces apoptosis in retinoblastoma cells via the Akt/mTOR signaling pathway by targeting PIK3CA. Oncol. Rep. 2020, 43, 1365–1374. [Google Scholar] [PubMed]
  73. Zhang, L.; Wang, L.; Lu, N.; Wang, J.; Yan, R.; Yan, H.; Zhang, J.; Zhang, M. Micro RNA-363 inhibits esophageal squamous cell carcinoma progression by directly targeting sperm-associated antigen 5. J. Int. Med Res. 2020, 48. [Google Scholar] [CrossRef]
  74. Xie, Y.; Chen, L.; Gao, Y.; Ma, X.; He, W.; Zhang, Y.; Zhang, F.; Fan, Y.; Gu, L.; Li, P.; et al. miR-363 suppresses the proliferation, migration and invasion of clear cell renal cell carcinoma by downregulating S1PR1. Cancer Cell Int. 2020, 20, 227. [Google Scholar] [PubMed]
  75. Zhang, R.; Li, Y.; Dong, X.; Peng, L.; Nie, X. MiR-363 sensitizes cisplatin-induced apoptosis targeting in Mcl-1 in breast cancer. Med Oncol. 2014, 31, 347. [Google Scholar] [PubMed]
  76. Zhang, H.; Zhang, N.; Wang, R.; Shao, T.; Feng, Y.; Yao, Y.; Wu, Q.; Zhu, S.; Cao, J.; Zhang, H.; et al. High expression of miR-363 predicts poor prognosis and guides treatment selection in acute myeloid leukemia. J. Transl. Med. 2019, 17, 106. [Google Scholar]
  77. Smallwood, D.T.; Apollonio, B.; Willimott, S.; Lezina, L.; Alharthi, A.; Ambrose, A.R.; De Rossi, G.; Ramsay, A.G.; Wagner, S.D. Extracellular vesicles released by CD40/IL-4-stimulated CLL cells confer altered functional properties to CD4+ T cells. Blood 2016, 128, 542–552. [Google Scholar]
  78. Huber, V.; Vallacchi, V.; Fleming, V.; Hu, X.; Cova, A.; Dugo, M.; Shahaj, E.; Sulsenti, R.; Vergani, E.; Filipazzi, P.; et al. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J. Clin. Investig. 2018, 128, 5505–5516. [Google Scholar]
  79. Liu, J.; Wang, D.; Long, Z.; Liu, J.; Li, W. CircRNA8924 Promotes Cervical Cancer Cell Proliferation, Migration and Invasion by Competitively Binding to MiR-518d-5p /519-5p Family and Modulating the Expression of CBX8. Cell Physiol. Biochem. 2018, 48, 173–184. [Google Scholar]
  80. Huang, J.; Cao, D.; Sha, J.; Zhu, X.; Han, S. DLL3 is regulated by LIN28B and miR-518d-5p and regulates cell proliferation, migration and chemotherapy response in advanced small cell lung cancer. Biochem. Biophys. Res. Commun. 2019, 514, 853–860. [Google Scholar]
  81. Organista-Nava, J.; Gómez-Gómez, Y.; Illades-Aguiar, B.; Alarcón-Romero, L.D.C.; 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] [PubMed] [Green Version]
  82. Ye, S.; Zhang, H.; Cai, T.-T.; Liu, Y.-N.; Ni, J.-J.; He, J.; Peng, J.-Y.; Chen, Q.-Y.; Mo, H.-Y.; Cui, J.; et al. Exosomal miR-24-3p impedes T-cell function by targetingFGF11and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016, 240, 329–340. [Google Scholar] [CrossRef] [PubMed]
  83. Fayyad-Kazan, H.; Hamade, E.; Rouas, R.; Najar, M.; Fayyad-Kazan, M.; El Zein, N.; ElDirani, R.; Hussein, N.; Fakhry, M.; Al-Akoum, C.; et al. Downregulation of microRNA-24 and -181 parallels the upregulation of IFN-gamma secreted by activated human CD4 lymphocytes. Hum. Immunol. 2014, 75, 677–685. [Google Scholar] [CrossRef]
  84. Lovat, F.; Nigita, G.; Distefano, R.; Nakamura, T.; Gasparini, P.; Tomasello, L.; Fadda, P.; Ibrahimova, N.; Catricala, S.; Palamarchuk, A.; et al. Combined loss of function of two different loci of miR-15/16 drives the pathogenesis of acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2020, 117, 12332–12340. [Google Scholar] [CrossRef]
  85. Sun, Y.-P.; Lu, F.; Han, X.-Y.; Ji, M.; Zhou, Y.; Zhang, A.-M.; Wang, H.-C.; Ma, D.-X.; Ji, C.-Y. MiR-424 and miR-27a increase TRAIL sensitivity of acute myeloid leukemia by targeting PLAG1. Oncotarget 2016, 7, 25276–25290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tang, L.-J.; Sun, G.-K.; Zhang, T.-J.; Wu, D.-H.; Zhou, J.-D.; Ma, B.-B.; Xu, Z.-J.; Wen, X.-M.; Chen, Q.; Yao, D.-M.; et al. Down-regulation of miR-29c is a prognostic biomarker in acute myeloid leukemia and can reduce the sensitivity of leukemic cells to decitabine. Cancer Cell Int. 2019, 19, 177. [Google Scholar] [CrossRef]
  87. Lim, E.L.; Trinh, D.L.; Ries, R.E.; Wang, J.; Gerbing, R.B.; Ma, Y.; Topham, J.; Hughes, M.; Pleasance, E.; Mungall, A.J.; et al. MicroRNA Expression-Based Model Indicates Event-Free Survival in Pediatric Acute Myeloid Leukemia. J. Clin. Oncol. 2017, 35, 3964–3977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Han, Y.-X.; You, L.-S.; Liu, H.; Mao, L.-P.; Ye, X.; Qian, W.-B. Apoptosis of acute myeloid leukemia HL-60 cells induced by CDK inhibitor SNS-032 and its molecular mechanisms. Zhejiang Da Xue Xue Bao Yi Xue Ban 2015, 44, 174–178. [Google Scholar] [PubMed]
  89. Xu, P.; Zhou, D.; Yan, G.; Ouyang, J.; Chen, B. Correlation of miR-181a and three HOXA genes as useful biomarkers in acute myeloid leukemia. Int. J. Lab. Hematol. 2020, 42, 16–22. [Google Scholar] [CrossRef]
  90. Ma, Y.; Yan, M.-X.; Luo, Z.-Y. Expression of miR-181a in Acute Myeloid Leukaemia and Its Effect on Cell Proliferation. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2016, 24, 985–989. [Google Scholar]
  91. Liu, X.; Liao, W.; Peng, H.; Luo, X.; Luo, Z.; Jiang, H.; Xu, L. miR-181a promotes G1/S transition and cell proliferation in pediatric acute myeloid leukemia by targeting ATM. J. Cancer Res. Clin. Oncol. 2016, 142, 77–87. [Google Scholar] [CrossRef] [PubMed]
  92. Hua, J.-Y.; Feng, Y.; Pang, Y.; Zhou, X.-H.; Xu, B.; Yan, M.-X. MiR-181a Promotes Proliferation of Human Acute Myeloid Leukemia Cells by Targeting ATM. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2016, 24, 347–351. [Google Scholar] [PubMed]
  93. Guo, Q.; Luan, J.; Li, N.; Zhang, Z.; Zhu, X.; Zhao, L.; Wei, R.; Sun, L.; Shi, Y.; Yin, X.; et al. MicroRNA-181 as a prognostic biomarker for survival in acute myeloid leukemia: A meta-analysis. Oncogene 2017, 8, 89130–89141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Nanbakhsh, A.; Visentin, G.; Olive, D.; Janji, B.; Mussard, E.; Dessen, P.; Meurice, G.; Zhang, Y.; Louache, F.; Bourhis, J.-H.; et al. miR-181a modulates acute myeloid leukemia susceptibility to natural killer cells. OncoImmunology 2015, 4, e996475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Okada, H.; Kohanbash, G.; Lotze, M.T. MicroRNAs in immune regulation—Opportunities for cancer immunotherapy. Int. J. Biochem. Cell Boil. 2010, 42, 1256–1261. [Google Scholar] [CrossRef] [Green Version]
  96. Papapetrou, E.P.; Kovalovsky, D.; Beloeil, L.; Sant’Angelo, D.; Sadelain, M. Harnessing endogenous miR-181a to segregate transgenic antigen receptor expression in developing versus post-thymic T cells in murine hematopoietic chimeras. J. Clin. Investig. 2009, 119, 157–168. [Google Scholar] [CrossRef]
  97. Kagoya, Y.; Nakatsugawa, M.; Saso, K.; Guo, T.; Anczurowski, M.; Wang, C.-H.; Butler, M.O.; Arrowsmith, C.H.; Hirano, N. DOT1L inhibition attenuates graft-versus-host disease by allogeneic T cells in adoptive immunotherapy models. Nat. Commun. 2018, 9, 1915. [Google Scholar] [CrossRef]
  98. Achberger, S.; Aldrich, W.; Tubbs, R.; Crabb, J.W.; Singh, A.D.; Triozzi, P.L. Circulating immune cell and microRNA in patients with uveal melanoma developing metastatic disease. Mol. Immunol. 2014, 58, 182–186. [Google Scholar] [CrossRef] [Green Version]
  99. Vignard, V.; Labbé, M.; Marec, N.; André-Grégoire, G.; Jouand, N.; Fonteneau, J.-F.; Labarrière, N.; Fradin, D. MicroRNAs in Tumor Exosomes Drive Immune Escape in Melanoma. Cancer Immunol. Res. 2020, 8, 255–267. [Google Scholar] [CrossRef] [Green Version]
  100. Su, R.; Lin, H.-S.; Zhang, X.-H.; Yin, X.-L.; Ning, H.-M.; Liu, B.; Zhai, P.-F.; Gong, J.-N.; Shen, C.; Song, L.; et al. MiR-181 family: Regulators of myeloid differentiation and acute myeloid leukemia as well as potential therapeutic targets. Oncogene 2014, 34, 3226–3239. [Google Scholar] [CrossRef]
  101. Lim, S.P.; Ioannou, N.; Ramsay, A.G.; Darling, D.; Gäken, J.; Mufti, G.J. miR-181c -BRK1 axis plays a key role in actin cytoskeleton-dependent T cell function. J. Leukoc. Boil. 2018, 103, 855–866. [Google Scholar]
  102. Le Dieu, R.; Taussig, D.C.; Ramsay, A.G.; Mitter, R.; Miraki-Moud, F.; Fatah, R.; Lee, A.M.; Lister, T.A.; Gribben, J.G. Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood 2009, 114, 3909–3916. [Google Scholar] [PubMed] [Green Version]
  103. Zhang, Z.; Xue, Z.; Liu, Y.; Liu, H.; Guo, X.; Li, Y.; Yang, H.; Zhang, L.; Da, Y.; Yao, Z.; et al. MicroRNA-181c promotes Th17 cell differentiation and mediates experimental autoimmune encephalomyelitis. Brain Behav. Immun. 2018, 70, 305–314. [Google Scholar]
  104. Li, X.; Liu, L.; Yang, J.; Yu, Y.; Chai, J.; Wang, L.; Ma, L.; Yin, H. Exosome Derived From Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation. EBioMedicine 2016, 8, 72–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Huang, L.; Tang, X.; Shi, X.; Su, L. miR-532-5p promotes breast cancer proliferation and migration by targeting RERG. Exp. Ther. Med. 2019, 19, 400–408. [Google Scholar]
  106. Zhang, J.; Zhou, W.; Liu, Y.; Liu, T.; Li, C.; Wang, L. Oncogenic role of microRNA-532-5p in human colorectal cancer via targeting of the 5’UTR of RUNX3. Oncol. Lett. 2018, 15, 7215–7220. [Google Scholar] [CrossRef] [Green Version]
  107. Xu, X.; Zhang, Y.; Liu, Z.; Zhang, X.; Jia, J. miRNA-532-5p functions as an oncogenic microRNA in human gastric cancer by directly targeting RUNX3. J. Cell. Mol. Med. 2016, 20, 95–103. [Google Scholar]
  108. Lin, X.; Ling, Q.; Lv, Y.; Ye, W.; Huang, J.; Guo, Q.; Wang, J.; Li, Z.; Jin, J. Plasma exosome-derived microRNA-532 as a novel predictor for acute myeloid leukemia. Cancer Biomarkers 2020, 28, 151–158. [Google Scholar]
  109. Ibrahim, S.; Szóstek-Mioduchowska, A.; Skarzynski, D.J. Expression profiling of selected miRNAs in equine endometrium in response to LPS challenge in vitro: A new understanding of the inflammatory immune response. Veter. Immunol. Immunopathol. 2019, 209, 37–44. [Google Scholar] [CrossRef]
  110. Shomali, N.; Mansoori, B.; Mohammadi, A.; Shirafkan, N.; Ghasabi, M.; Baradaran, B. MiR-146a functions as a small silent player in gastric cancer. Biomed. Pharmacother. 2017, 96, 238–245. [Google Scholar] [CrossRef]
  111. Emming, S.; Chirichella, M.; Monticelli, S. MicroRNAs as modulators of T cell functions in cancer. Cancer Lett. 2018, 430, 172–178. [Google Scholar] [CrossRef] [PubMed]
  112. Gagnon, J.D.; Kageyama, R.; Shehata, H.M.; Fassett, M.S.; Mar, D.J.; Wigton, E.J.; Johansson, K.; Litterman, A.J.; Odorizzi, P.; Simeonov, D.; et al. miR-15/16 Restrain Memory T Cell Differentiation, Cell Cycle, and Survival. Cell Rep. 2019, 28, 2169–2181.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Singh, Y.; Garden, O.A.; Lang, F.; Cobb, B.S. MicroRNA-15b/16 Enhances the Induction of Regulatory T Cells by Regulating the Expression of Rictor and mTOR. J. Immunol. 2015, 195, 5667–5677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Yao, X.; Tu, Y.; Xu, Y.; Guo, Y.; Yao, F.; Zhang, X. Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J. Cell. Mol. Med. 2020. [Google Scholar] [CrossRef]
  115. Fan, X.; Wang, J.; Qin, T.; Zhang, Y.; Liu, W.; Jiang, K.; Huang, D. Exosome miR-27a-3p secreted from adipocytes targets ICOS to promote antitumor immunity in lung adenocarcinoma. Thorac. Cancer 2020, 11, 1453–1464. [Google Scholar] [CrossRef]
  116. Wang, Y.; Zhang, X.; Li, H.; Yu, J.; Ren, X. The role of miRNA-29 family in cancer. Eur. J. Cell Boil. 2013, 92, 123–128. [Google Scholar] [CrossRef]
  117. Schmitt, M.; Margue, C.; Behrmann, I.; Kreis, S. MiRNA-29: A microRNA Family with Tumor-Suppressing and Immune-Modulating Properties. Curr. Mol. Med. 2013, 13, 572–585. [Google Scholar] [CrossRef]
  118. Chandiran, K.; Lawlor, R.; Pannuti, A.; Perez, G.G.; Srinivasan, J.; Golde, T.E.; Miele, L.; Osborne, B.A.; Minter, L.M. Notch1 primes CD4 T cells for T helper type I differentiation through its early effects on miR-29. Mol. Immunol. 2018, 99, 191–198. [Google Scholar] [CrossRef]
Figure 1. Differentially expressed bone marrow circulating miRNAs in newly diagnosed AML patients compared to healthy donors. (A) Hierarchical clustering and heatmap of microRNA (miRNA) expression profile in acute myeloid leukemia (AML) (n = 27) versus healthy donors (n = 11). The list of miRNAs was filtered with a p-value < 0.005 and fold change ≤ 0.5 or ≥ 2. A total of 39 miRNAs (21 up- and 18 downregulated) were significant differentially expressed. The color key indicates centred and standardized miRNA expression levels (Z-scores). Upper color bar labels samples by baseline diagnosis. (B) Principal Component Analysis (PCA) showing the grouping of AML patients (red) and HD (blue) based on the 39 DEmiRNAs profile.
Figure 1. Differentially expressed bone marrow circulating miRNAs in newly diagnosed AML patients compared to healthy donors. (A) Hierarchical clustering and heatmap of microRNA (miRNA) expression profile in acute myeloid leukemia (AML) (n = 27) versus healthy donors (n = 11). The list of miRNAs was filtered with a p-value < 0.005 and fold change ≤ 0.5 or ≥ 2. A total of 39 miRNAs (21 up- and 18 downregulated) were significant differentially expressed. The color key indicates centred and standardized miRNA expression levels (Z-scores). Upper color bar labels samples by baseline diagnosis. (B) Principal Component Analysis (PCA) showing the grouping of AML patients (red) and HD (blue) based on the 39 DEmiRNAs profile.
Ijms 21 07065 g001
Figure 2. AML bone marrow circulating miRNA signature. The dot blot (A,B) and receiver operating characteristic (ROC) curves (C,D) reflecting the ability of the bone marrow circulating miRNA expression signature to differentiate the AML cases (n = 27) from the controls (n = 11). (A) Downregulated miRNAs: miR-448, 890, 504, 145, 365 and. (B) Upregulated miRNAs: miR-9; 21, 629 and 449b. (C) ROC curves reflecting the performance of the individual features of the 9 miRNA signature in discriminating AML patients from healthy donors. (D) Combined ROC showing the overall discriminatory power of the 9-miRNA signature. AUROC: Area Under ROC; CI: 95% Confidence Intervals; p-value: correspond to a Mann-Whitney U test addresses testing the null hypothesis of the AUROC is 0.5 (i.e., the classifier is random).
Figure 2. AML bone marrow circulating miRNA signature. The dot blot (A,B) and receiver operating characteristic (ROC) curves (C,D) reflecting the ability of the bone marrow circulating miRNA expression signature to differentiate the AML cases (n = 27) from the controls (n = 11). (A) Downregulated miRNAs: miR-448, 890, 504, 145, 365 and. (B) Upregulated miRNAs: miR-9; 21, 629 and 449b. (C) ROC curves reflecting the performance of the individual features of the 9 miRNA signature in discriminating AML patients from healthy donors. (D) Combined ROC showing the overall discriminatory power of the 9-miRNA signature. AUROC: Area Under ROC; CI: 95% Confidence Intervals; p-value: correspond to a Mann-Whitney U test addresses testing the null hypothesis of the AUROC is 0.5 (i.e., the classifier is random).
Ijms 21 07065 g002
Figure 3. Confirmation of differentially expressed miRNAs in a second cohort (n = 15).(A) The dot plots of Downregulated and Upregulated AML miRNA signature in a second cohort of AML patients at diagnosis compared to HD (B) ROC curves reflecting the performance of the individual features of the 9 miRNA signature in discriminating AML patients from healthy donors. (C) ROC plot showing the discriminatory value of the 9-miRNA signature in the second cohort. AUROC: Area Under ROC; CI: 95% Confidence Intervals; p-value: correspond to a Mann-Whitney U test addresses testing the null hypothesis of the AUROC is 0.5 (i.e., the classifier is random).
Figure 3. Confirmation of differentially expressed miRNAs in a second cohort (n = 15).(A) The dot plots of Downregulated and Upregulated AML miRNA signature in a second cohort of AML patients at diagnosis compared to HD (B) ROC curves reflecting the performance of the individual features of the 9 miRNA signature in discriminating AML patients from healthy donors. (C) ROC plot showing the discriminatory value of the 9-miRNA signature in the second cohort. AUROC: Area Under ROC; CI: 95% Confidence Intervals; p-value: correspond to a Mann-Whitney U test addresses testing the null hypothesis of the AUROC is 0.5 (i.e., the classifier is random).
Ijms 21 07065 g003
Figure 4. Prognosis value of AML bone marrow circulating miRNAs. The expression dot blot and receiver operating characteristic (ROC) curves reflecting the representative bone marrow circulating miRNAs (4 miRNAs) differentially expressed between the AML patients based on their overall survival at diagnosis (A) and validated in an independent second cohort of patients (B). (C) Receiver Operating Characteristic (ROC) plot validating the combined prognosis power of miR-671-3p and miR-328 in the first and second cohorts. AMLDCR: AML at diagnosis samples in which the patients have complete remission 4 years post diagnosis. AMLDR: AML at diagnosis samples in which the patients relapsed and died during the 4 years post diagnosis.
Figure 4. Prognosis value of AML bone marrow circulating miRNAs. The expression dot blot and receiver operating characteristic (ROC) curves reflecting the representative bone marrow circulating miRNAs (4 miRNAs) differentially expressed between the AML patients based on their overall survival at diagnosis (A) and validated in an independent second cohort of patients (B). (C) Receiver Operating Characteristic (ROC) plot validating the combined prognosis power of miR-671-3p and miR-328 in the first and second cohorts. AMLDCR: AML at diagnosis samples in which the patients have complete remission 4 years post diagnosis. AMLDR: AML at diagnosis samples in which the patients relapsed and died during the 4 years post diagnosis.
Ijms 21 07065 g004
Figure 5. Prognosis value of AML bone marrow circulating miRNAs related to remission after treatment. Correlated differentially expressed miRNAs between AMLD vs. AMLPT patients and AMLD patients vs. HD. The expression dot plot curves reflecting the correlated differentially expressed bone marrow circulating miRNAs in AML patients at diagnosis (n = 12) compared to both healthy donors (n = 11) and the same patients in relative remission (blast < 5%) post treatment (n = 12). AMLD: AML patients at diagnosis. HD: Healthy donors. AMLPT: AML first complete remission post treatment patients.
Figure 5. Prognosis value of AML bone marrow circulating miRNAs related to remission after treatment. Correlated differentially expressed miRNAs between AMLD vs. AMLPT patients and AMLD patients vs. HD. The expression dot plot curves reflecting the correlated differentially expressed bone marrow circulating miRNAs in AML patients at diagnosis (n = 12) compared to both healthy donors (n = 11) and the same patients in relative remission (blast < 5%) post treatment (n = 12). AMLD: AML patients at diagnosis. HD: Healthy donors. AMLPT: AML first complete remission post treatment patients.
Ijms 21 07065 g005
Table 1. Bone marrow circulating miRNAs differentially expressed in AML newly diagnosed compared to healthy donors.
Table 1. Bone marrow circulating miRNAs differentially expressed in AML newly diagnosed compared to healthy donors.
Up regulated miRNAs in AMLD vs. HD
MiR ConnotationpValueFC AMLD1 vs. HD
hsa-miR-520a-3p0.0000004103.59
hsa-miR-548b-5p0.000018715.35
hsa-miR-6510.000706613.01
hsa-miR-449b0.00299657.84
hsa-miR-520f0.00124037.19
hsa-miR-330-5p0.04768036.55
hsa-miR-34c-5p0.03004605.10
hsa-miR-90.003038815.45
hsa-miR-34a0.000839114.22
hsa-miR-548d-5p0.000039013.09
hsa-miR-6550.00619178.55
hsa-miR-548c-5p0.00029377.99
hsa-miR-135a0.02259666.95
hsa-miR-502-5p0.00283526.68
hsa-miR-576-3p0.00086815.99
hsa-miR-449a0.00033045.97
hsa-miR-518f0.00152375.88
hsa-miR-210.00001185.61
hsa-miR-6290.00000005.60
hsa-miR-1950.00000505.32
hsa-miR-517c0.00110335.02
hsa-miR-5970.00266634.94
hsa-miR-199b-5p0.04216354.85
hsa-miR-548d-3p0.02038884.76
hsa-miR-181a0.00393924.76
hsa-miR-5700.01804774.75
hsa-miR-6600.00001014.56
hsa-miR-2020.01006134.24
hsa-miR-240.02251844.24
hsa-miR-3750.00168714.18
hsa-miR-130b0.00001153.81
hsa-miR-361-5p0.00203773.65
hsa-miR-4510.00199803.58
hsa-miR-181c0.01319023.49
hsa-miR-5790.00024173.49
hsa-miR-362-3p0.01060663.22
hsa-miR-250.00057643.15
hsa-miR-29c0.00950143.00
hsa-miR-5110.01036692.90
hsa-miR-146b-3p0.00967302.88
hsa-miR-532-5p0.00142762.70
hsa-miR-106a0.00128182.62
hsa-miR-8740.00894342.61
hsa-miR-2120.00123512.50
hsa-miR-5980.00462312.48
hsa-miR-2210.01448402.47
hsa-miR-18a0.00521622.46
hsa-miR-27a0.01608792.45
hsa-miR-590-5p0.00157332.40
hsa-miR-20b0.04713032.36
hsa-miR-1010.01627022.23
hsa-miR-146a0.01742182.22
hsa-miR-142-3p0.02469462.21
hsa-miR-146b-5p0.03050512.14
hsa-miR-930.01756701.97
hsa-miR-140-5p0.03622661.96
hsa-miR-18b0.02245771.90
hsa-miR-423-5p0.00885641.87
hsa-miR-92a0.04434791.71
hsa-miR-2100.04294231.68
hsa-miR-483-5p0.04235411.62
Downregulated miRNAs in AMLD vs. HD
MiR Connotationp ValueFC AMLD1 vs. HD
hsa-miR-3260.006633260.103
hsa-miR-1980.002826230.078
hsa-miR-518d-3p0.000012850.018
hsa-miR-1070.000001300.012
hsa-miR-2150.000826730.009
hsa-miR-5040.000000040.006
hsa-miR-8900.000009950.001
hsa-miR-4480.000074980.001
hsa-miR-532-3p0.052453650.616
hsa-miR-342-3p0.048843260.602
hsa-miR-200c0.049946800.547
hsa-miR-491-5p0.011875700.543
hsa-miR-28-3p0.007185980.531
hsa-miR-99b0.006868130.444
hsa-miR-574-3p0.012322310.432
hsa-miR-7440.000824440.358
hsa-miR-1900.054209730.353
hsa-miR-1500.003172630.322
hsa-miR-1970.000058920.270
hsa-miR-125a-5p0.006147510.262
hsa-miR-2030.020148820.258
hsa-miR-891a0.026271270.248
hsa-miR-485-3p0.054814950.247
hsa-miR-4890.009387280.245
hsa-miR-2040.000271580.207
hsa-miR-193b0.004875720.203
hsa-miR-1840.016942390.203
hsa-miR-4840.000124740.173
hsa-miR-6180.034359690.152
hsa-miR-1340.013903110.145
hsa-miR-4330.046892470.135
hsa-miR-1450.000000420.120
hsa-miR-1490.027042960.119
hsa-miR-3280.000042610.105
hsa-miR-654-5p0.030867970.079
hsa-miR-3650.000006860.066
hsa-miR-518b0.001690710.063
Table 2. Confirmed differentially expressed microRNAs between AML patients and HD in a second cohort.
Table 2. Confirmed differentially expressed microRNAs between AML patients and HD in a second cohort.
MiRNAs
Connotation
p Value
AMLD1
FC
AMLD1
p Value
AMLD2
FC
AMLD2
p Value
AMLD Total
FC.
AMLD Total
hsa-miR-520a-3p4.08 × 10−7103.580.0163.7249.22 × 10−531.587
hsa-miR-90.00315.4513.20 × 10−541.6780.000222.022
hsa-miR-548b-5p1.87 × 10−515.3540.222.0030.00267.418
hsa-miR-34a0.000814.2210.00613.4436.19 × 10−513.938
hsa-miR-548d-5p3.89 × 10−513.0910.0215.2687.85 × 10−59.458
hsa-miR-6510.000713.0110.0047.1290.000610.496
hsa-miR-548c-5p0.00027.9930.1501.9610.00314.839
hsa-miR-449b0.00297.8430.0076.3010.00197.253
hsa-miR-520f0.00127.1900.0013.5620.00115.594
hsa-miR-502-5p0.00286.6760.1322.8980.01094.955
hsa-miR-576-3p0.00085.9892.11 × 10−58.3642.68 × 10−56.748
hsa-miR-449a0.00035.9720.0782.8140.00214.565
hsa-miR-211.17 × 10−55.6111.76 × 10−78.9138.07 × 10−86.619
hsa-miR-6292.98 × 10−85.6002.23 × 10−73.6933.41 × 10−94.826
hsa-miR-1955.006 × 10−65.3150.201.5460.0023.419
hsa-miR-517c0.00115.0190.351.2640.0203.068
hsa-miR-5970.00264.9410.00088.2150.00015.925
hsa-miR-181a0.00394.7600.01612.7550.00443.916
hsa-miR-6601.00 × 10−54.5580.05901.5360.00033.091
hsa-miR-3750.0014.1770.00099.5440.00015.611
hsa-miR-130b1.14 × 10−53.8150.17941.7660.00772.897
hsa-miR-361-5p0.0023.6510.01681.9200.00252.902
hsa-miR-4510.00193.5840.01282.6590.00163.221
hsa-miR-5790.00023.4870.02901.9270.00062.821
hsa-miR-250.00053.1545.15 × 10−54.0602.71 × 10−53.451
hsa-miR-106a0.00122.6160.071.5580.00502.174
hsa-miR-2120.00122.5010.491.0030.02511.805
hsa-miR-5980.00462.4820.241.3460.02801.994
hsa-miR-18a0.00522.4590.00083.1010.00112.671
hsa-miR-7440.00080.3583.03 × 10−50.2018.80 × 10−50.291
hsa-miR-1500.00310.3221.10 × 10−50.1160.00010.223
hsa-miR-1975.8910−50.2700.190.7340.00410.386
hsa-miR-2040.00020.2071.39 × 10−50.1138.73 × 10−60.167
hsa-miR-193b0.00480.2039.67 × 10−70.0520.00010.125
hsa-miR-4840.00010.1730.110.5520.00230.262
hsa-miR-1454.21 × 10−70.1204.47 × 10−60.1123.11 × 10−90.117
hsa-miR-3284.26 × 10−50.1050.421.1350.0140.245
hsa-miR-1980.00280.0780.220.4090.0130.141
hsa-miR-3656.86 × 10−60.0660.00050.1323.58 × 10−60.084
hsa-miR-518b0.0010.0634.28 × 10−50.0075.05 × 10−50.028
hsa-miR-518d-3p1.28 × 10−50.0180.370.6560.0050.066
hsa-miR-1071.30 × 10−60.0120.110.1740.00040.031
hsa-miR-2150.00080.0090.060.0830.00240.019
hsa-miR-5043.50 × 10−80.0065.70 × 10−60.0121.70 × 10−100.008
hsa-miR-8909.94 × 10−60.0010.00010.0019.74 × 10−80.001
hsa-miR-4487.49 × 10−50.0010.00070.0011.63 × 10−60.001
The 46 miRNAs identified in the first cohort were confirmed in a second cohort. The list of miRNAs from first cohort was filtered with a p-value < 0.005 and fold change ≤ 0.5 or ≥ 2.0. FC: fold change.
Table 3. Gene Ontology classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed circulating bone marrow miRNAs in AML. A) KEGG Pathway Enrichment Analysis of differentially expressed bone marrow circulating miRNAs in AML. A. GO annotation of differentially expressed bone marrow circulating miRNAs in AML.B) GO annotation of differentially expressed bone marrow circulating miRNAs in AML.
Table 3. Gene Ontology classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed circulating bone marrow miRNAs in AML. A) KEGG Pathway Enrichment Analysis of differentially expressed bone marrow circulating miRNAs in AML. A. GO annotation of differentially expressed bone marrow circulating miRNAs in AML.B) GO annotation of differentially expressed bone marrow circulating miRNAs in AML.
A
KEGG ID and TermCountp value
hsa05206: MicroRNAs in cancer606.05 × 10−11
hsa05200: Pathways in cancer621.51 × 10−10
hsa04014: Ras signaling pathway482.29 × 10−5
hsa05221:Acute myeloid leukemia453.98 × 10−5
hsa04068: FoxO signaling pathway520.000106
hsa04151: PI3K-Akt signaling pathway540.000182
hsa04012: ErbB signaling pathway460.00181
hsa05202: Transcriptional misregulation in cancer490.003887
hsa04066: HIF-1 signaling pathway470.003892
hsa05231: Choline metabolism in cancer420.00457
hsa04520: Adherens junction500.004866
hsa04210: Apoptosis350.005405
hsa04620: Toll-like receptor signaling pathway370.005751
hsa04010: MAPK signaling pathway520.009669
hsa04672: Intestinal immune network for IgA production160.010748
hsa04064: NF-kappa B signaling pathway370.012295
hsa04150: mTOR signaling pathway300.014025
hsa04650: Natural killer cell mediated cytotoxicity340.014803
hsa04062: Chemokine signaling pathway480.01481
hsa04550: Signaling pathways regulating pluripotency of stem cells450.016837
hsa04660: T cell receptor signaling pathway320.031392
hsa04115: p53 signaling pathway370.045474
hsa04330: Notch signaling pathway260.047169
hsa04110: Cell cycle440.049139
B
Gene Ontology: GO ID and TermCountp value
Biological Process
GO:0010604~positive regulation of macromolecule metabolic process702.52384 × 10−16
GO:0010628~positive regulation of gene expression681.05757 × 10−12
GO:0042127~regulation of cell proliferation691.97068 × 10−11
GO:0008219~cell death683.48089 × 10−10
GO:0006915~apoptotic process664.40894 × 10−10
GO:0002682~regulation of immune system process656.91088 × 10-10
GO:0045595~regulation of cell differentiation681.57355 × 10−9
GO:0060548~negative regulation of cell death621.0149 × 10−8
GO:0008284~positive regulation of cell proliferation621.04 × 10−8
GO:0097190~apoptotic signaling pathway541.20513 × 10−8
GO:0043410~positive regulation of MAPK cascade562.1753 × 10−6
GO:0050776~regulation of immune response574.45138 × 10−6
GO:0070489~T cell aggregation576.58436 × 10−6
GO:0051726~regulation of cell cycle588.80856 × 10−6
GO:0043408~regulation of MAPK cascade601.07104 × 10−5
GO:0045596~negative regulation of cell differentiation651.11453 × 10−5
GO:0097191~extrinsic apoptotic signaling pathway471.20729 × 10−5
GO:0002819~regulation of adaptive immune response339.06477 × 10−5
GO:0007219~Notch signaling pathway439.88879 × 10−5
GO:0046651~lymphocyte proliferation450.000115119
GO:0006955~immune response620.000124958
GO:0002683~negative regulation of immune system process450.000299385
GO:0001959~regulation of cytokine-mediated signaling pathway370.00094784
GO:0030099~myeloid cell differentiation540.001058782
GO:0097194~execution phase of apoptosis310.00363619
GO:0070227~lymphocyte apoptotic process320.008369403
GO:0097300~programmed necrotic cell death130.011048278
Molecular Function
GO:0019899~enzyme binding672.15784 × 10−7
GO:0000981~RNA polymerase II transcription factor activity. SSB651.2604 × 10−5
GO:0044212~transcription regulatory region DNA binding643.84654 × 10−5
GO:0000975~regulatory region DNA binding645.11725 × 10−5
GO:0044877~macromolecular complex binding625.44329 × 10−5
GO:0019900~kinase binding626.77415 × 10−5
GO:0008134~transcription factor binding640.000131465
GO:0003690~double-stranded DNA binding620.000142272
GO:0003682~chromatin binding520.00091488
GO:0016301~kinase activity610.001966828
GO:0001077~transcriptional activator activity. RNA polymerase II SSB490.002467546
GO:0050839~cell adhesion molecule binding450.007658872
GO:0001085~RNA polymerase II transcription factor binding400.01087498
GO:0005126~cytokine receptor binding490.010986866
GO:0071837~HMG box domain binding160.012673444
GO:0019838~growth factor binding420.021399477
GO:0042826~histone deacetylase binding400.02268476
GO:0008327~methyl-CpG binding150.039968845
GO:0005125~cytokine activity370.04028971
GO:0070851~growth factor receptor binding380.046274794
Cellular Component
GO:0000785~chromatin550.000142352
GO:0000790~nuclear chromatin510.000199439
GO:0005829~cytosol680.001232442
GO:0044454~nuclear chromosome part520.002342868
GO:0000792~heterochromatin250.002647328
GO:0000791~euchromatin200.003993973
GO:0005578~proteinaceous extracellular matrix420.004102668
GO:0044427~chromosomal part560.00606402
GO:0005794~Golgi apparatus500.006148438
GO:0005741~mitochondrial outer membrane320.010046195
GO:0005576~extracellular region610.010189067
GO:0005667~transcription factor complex470.017135475
GO:0016604~nuclear body420.017270393
GO:0019867~outer membrane320.018716537
GO:0098857~membrane microdomain510.020024305
GO:0045121~membrane raft400.024624821
GO:0005739~mitochondrion580.027721756
GO:0005912~adherens junction510.02781721
GO:0070161~anchoring junction360.028521889
Table 4. Differentially expressed miRNAs based on genetic risk AML Good prognosis (FAV) compared to Adverse risk (ADV).
Table 4. Differentially expressed miRNAs based on genetic risk AML Good prognosis (FAV) compared to Adverse risk (ADV).
MiRNAs Connotationpvalue FAV vs. ADV FC FAV vs. ADV
hsa-miR-886-3p0.0033620620.242595592
hsa-miR-671-3p0.0035507660.533445656
hsa-miR-1870.004011370.071572526
hsa-miR-886-5p0.0074224180.36174907
hsa-miR-99b0.0114231560.405734354
hsa-miR-337-5p0.0169092310.147489611
hsa-miR-501-5p0.0197007570.515368965
hsa-miR-125a-5p0.0197601740.252095185
hsa-miR-532-3p0.0230407540.49492166
hsa-miR-6360.0245382720.436038555
hsa-miR-196b0.028576870.210572395
hsa-miR-3630.0305404570.459860525
hsa-miR-1520.0371243430.44325062
hsa-miR-125a-3p0.0453928560.277844263
hsa-miR-6160.0469182540.557903991
hsa-miR-1840.046933330.148403457
hsa-miR-3280.0469901450.315492217
hsa-miR-490-3p0.0498228190.075649011
hsa-miR-2220.0499409173.023869496
The list of miRNAs was filtered with a p-value < 0.05 and fold change ≥ 1.5.
Table 5. Differentially expressed bone marrow circulating miRNAs between newly diagnosed (AMLD) and first complete remission post-treated (AMLPT) AML patients.
Table 5. Differentially expressed bone marrow circulating miRNAs between newly diagnosed (AMLD) and first complete remission post-treated (AMLPT) AML patients.
MIRNAs Connotationp Value FC AMLD vs. AMLPT
hsa-miR-10b0.0142197147.195
hsa-miR-220.0277767830.223
has-miR-1552.1736 × 10−615.886
hsa-miR-29c7.6596 × 10−711.930
hsa-miR-181a0.0001883611.307
hsa-miR-181c0.0006092810.557
hsa-miR-310.043464739.674
hsa-miR-193a-3p0.00177439.137
hsa-miR-542-5p0.002558459.027
hsa-miR-240.02025248.248
hsa-miR-517a0.001484947.944
hsa-miR-2050.033070497.550
hsa-miR-146b-5p2.0118 10−77.488
hsa-miR-320.011631377.056
hsa-miR-1500.000523946.297
hsa-miR-196b0.010015776.294
hsa-miR-29a0.004993116.213
hsa-miR-950.000127235.918
hsa-miR-140-3p2.4156 × 10−65.603
hsa-miR-5030.037011345.591
hsa-miR-6160.001532985.563
hsa-miR-3451.9647 × 10−55.303
hsa-miR-324-3p0.006326345.086
hsa-miR-6270.007967995.039
hsa-miR-5230.042556254.853
hsa-miR-574-3p0.050356574.795
hsa-miR-362-3p0.011897984.470
hsa-miR-339-3p0.002111714.439
hsa-miR-518f0.019149464.261
hsa-miR-548b-5p0.036728254.166
hsa-miR-6520.002219813.952
hsa-miR-4890.018687623.762
hsa-miR-7080.045421883.758
hsa-miR-362-5p0.000668853.745
hsa-miR-324-5p0.00045523.737
hsa-miR-146b-3p0.003503973.544
hsa-miR-2129.932810−53.541
hsa-miR-106b0.000223763.533
hsa-miR-628-5p0.007242923.350
hsa-miR-1380.005215573.343
hsa-miR-342-3p0.001341693.336
hsa-miR-146a0.01323473.315
hsa-miR-140-5p0.00134073.143
hsa-miR-374b0.001726762.978
hsa-miR-548c-5p0.040398872.967
hsa-miR-361-5p0.00937242.963
hsa-miR-200c0.002750032.961
hsa-miR-3400.035780442.952
hsa-miR-200b1.0495 × 10−52.810
hsa-miR-1260.001027532.638
hsa-miR-7440.011469152.633
hsa-miR-502-5p0.028893172.510
hsa-miR-590-5p0.000941892.509
hsa-miR-28-5p0.011057952.491
hsa-miR-142-5p0.009510692.371
hsa-let-7g0.008836282.368
hsa-miR-374a0.011895762.338
hsa-miR-199b-5p0.046575832.248
hsa-miR-532-5p0.018651582.236
hsa-miR-27a0.023442862.230
hsa-miR-1860.013908672.225
hsa-miR-15b0.016054292.115
hsa-miR-1950.022926562.105
hsa-miR-26a0.022807522.101
hsa-miR-106a0.009256262.099
hsa-miR-26b0.034715091.985
hsa-miR-30c0.031235221.869
hsa-miR-30b0.045255011.765
hsa-miR-491-5p0.03059361.652
hsa-miR-3750.036349040.393
hsa-miR-99b0.005494370.391
hsa-miR-3650.048376960.373
hsa-miR-92a0.009743460.371
hsa-miR-671-3p2.371 × 10−50.312
hsa-miR-450a0.046650150.232
hsa-miR-4840.004246980.187
hsa-miR-885-5p0.007589730.185
hsa-miR-1900.006989370.154
hsa-miR-654-3p0.019755620.123
hsa-miR-3280.013661780.121
hsa-miR-2110.024520680.099
hsa-miR-296-5p0.000350130.078
hsa-miR-486-5p4.0707 × 10−50.075
hsa-miR-490-3p0.020914990.055
hsa-miR-485-3p0.003782250.039
hsa-miR-23a0.019134520.015
hsa-miR-518d-3p0.000551430.008
hsa-miR-3268.7126 × 10−70.003
Differentially expressed bone marrow circulating miRNAs between AML patients at diagnosis (AMLD) and first complete remission post treatment (AMLPT), consisting of 69 upregulated and 19 downregulated miRNAs, were identified with a threshold of p value < 0.05 and 1,5-fold change.
Table 6. Summary of AML patient clinical details used for the analysis.
Table 6. Summary of AML patient clinical details used for the analysis.
Clinical Details of AML Patients (42)Number
Sex and age
Males20
Females 22
Age < 55y17
Age ≥ 55y25
Classification of AML Patients
Cytogenetic abnormalities
Normal Karyotype10
Chromosome 9 deletion 4
Inversion (16) MYH11-CBFB4
Translocation (t8;21)4
Translocation (t15;17)6
Translocation (X:21) (p11;q22)2
trisomy 81
complex Karyotype3
WHO 2016 System
AML with recurrent genetic abnormalities (t8;21)4
AML with recurrent genetic abnormalities (inv 16)4
AML with recurrent genetic abnormalities (t15;17)6
Acute monoblastic/monocytic leukemia 2
Pure erythroid leukemia2
AML not otherwise specified (NOS)20
AML with myelodysplasia-related changes6
ELN 2017 Genetic Risk Stratification
Favorable14
Intermediate7
Adverse16
unkown5
The classification of AML patients was performed according to WHO 2016. The genetic risk stratification of AML patients was performed according to the 2017 European LeukemiaNet genetic risk stratification [1]: Favorable = Good prognosis; IT = Intermediate prognosis; Adverse = adverse risk.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/ijms21197065

AMA Style

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. International Journal of Molecular Sciences. 2020; 21(19):7065. https://doi.org/10.3390/ijms21197065

Chicago/Turabian Style

Moussa Agha, Douâa, Redouane Rouas, Mehdi Najar, Fatima Bouhtit, Najib Naamane, Hussein Fayyad-Kazan, Dominique Bron, Nathalie Meuleman, Philippe Lewalle, and Makram Merimi. 2020. "Identification of Acute Myeloid Leukemia Bone Marrow Circulating MicroRNAs" International Journal of Molecular Sciences 21, no. 19: 7065. https://doi.org/10.3390/ijms21197065

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