Next Article in Journal
Efficacy of Lymph Node Location-Number Hybrid Staging System on the Prognosis of Gastric Cancer Patients
Next Article in Special Issue
miR-410 Is a Key Regulator of Epithelial-to-Mesenchymal Transition with Biphasic Role in Prostate Cancer
Previous Article in Journal
Preoperative Arterial Embolization of Musculoskeletal Tumors: A Tertiary Center Experience
Previous Article in Special Issue
A Network of MicroRNAs and mRNAs Involved in Melanosome Maturation and Trafficking Defines the Lower Response of Pigmentable Melanoma Cells to Targeted Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Significance of microRNAs in Hematologic Malignancies and Hematopoietic Stem Cell Transplantation

1
Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
2
Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
3
Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(9), 2658; https://doi.org/10.3390/cancers15092658
Submission received: 2 March 2023 / Revised: 14 April 2023 / Accepted: 2 May 2023 / Published: 8 May 2023
(This article belongs to the Collection miRNAs: New Insights in Tumor Biology)

Abstract

:

Simple Summary

Dysregulated microRNA (miRNA) expression has been described in the initiation and progression of a wide spectrum of hematologic malignancies. Moreover, aberrant miRNA expression patterns have been shown to be involved in severe post-transplant complications in patients receiving hematopoietic stem cell transplantations (HSCT), suggesting their use as promising diagnostic and prognostic biomarkers for acute graft-versus-host disease (aGvHD). Recently, activation or inhibition of deregulated miRNAs appears to be an attractive therapeutic option for increasing treatment efficacy. Further research concerning the associations between alterations in miRNA levels and malignant hematopoiesis and treatment response might bring new possibilities for specific miRNA-based approaches to improve the outcomes for hematologic cancer patients.

Abstract

Hematologic malignancies are a group of neoplastic conditions that can develop from any stage of the hematopoiesis cascade. Small non-coding microRNAs (miRNAs) play a crucial role in the post-transcriptional regulation of gene expression. Mounting evidence highlights the role of miRNAs in malignant hematopoiesis via the regulation of oncogenes and tumor suppressors involved in proliferation, differentiation, and cell death. In this review, we provide current knowledge about dysregulated miRNA expression in the pathogenesis of hematological malignancies. We summarize data about the clinical utility of aberrant miRNA expression profiles in hematologic cancer patients and their associations with diagnosis, prognosis, and the monitoring of treatment response. Moreover, we will discuss the emerging role of miRNAs in hematopoietic stem cell transplantation (HSCT), and severe post-HSCT complications, such as graft-versus-host disease (GvHD). The therapeutical potential of the miRNA-based approach in hemato-oncology will be outlined, including studies with specific antagomiRs, mimetics, and circular RNAs (circRNAs). Since hematologic malignancies represent a full spectrum of disorders with different treatment paradigms and prognoses, the potential use of miRNAs as novel diagnostic and prognostic biomarkers might lead to improvements, resulting in a more accurate diagnosis and better patient outcomes.

1. Introduction

Hematologic malignancies represent a set of malignant disorders of the blood and lymphatic systems characterized by rapid progression. Genetic and epigenetic changes result in the clonal proliferation of progenitor and stem cells. Changes in signaling pathways disrupt the self-renewal ability of hematopoietic cells, together with their proliferation and differentiation into other lineages. Due to the high heterogeneity of hematologic cancer subtypes, the evaluations of new diagnostic and prognostic biomarkers are greatly required to improve the outcomes of patients.
Small non-coding microRNAs (miRNAs), first described by Lee et al. [1], represent a group of short regulatory molecules that participate in practically every physiological process, playing a crucial role in the post-transcriptional regulation of gene expression [2,3]. MiRNAs are partially complementary to several mRNAs, and binding to mRNA leads to the degradation or downregulation of gene expression by several mechanisms, including translational repression, mRNA cleavage, and deadenylation [4]. Thus, miRNAs regulate numerous cellular processes, including differentiation, proliferation, migration, and apoptosis. Importantly, miRNAs are critically involved in tumorigenesis and metastatic spreading, and their dysregulation has been described in a wide spectrum of hematologic malignancies [5].
Malignant transformation of hematopoietic cells is associated with changes in the expression levels of oncogenes and tumor suppressor genes. The link between miRNAs and cancer was initially documented in the case of chronic lymphocytic leukemia (CLL) [6]. Altered miRNA biogenesis and post-transcriptional gene silencing have serious consequences for hematopoietic differentiation and proliferation, leading to malignant hematopoiesis. Recently, the results of preclinical and clinical studies have suggested the clinical utility of miRNAs as diagnostic and prognostic biomarkers for treatment response. Mounting evidence has found that miRNA-inducing gene expression changes and heterogenous genetic profiles might serve as potential tools for the personalized treatment of patients receiving hematopoietic stem cell transplantation (HSCT).
Herein, we aim to provide current knowledge about the involvement of miRNAs in the physiology and pathogenesis of hematologic malignancies. The emerging role of miRNAs in HSCT and patient outcomes will be discussed. Moreover, we outline the new research trends and the possibilities of miRNA-based approaches for clinical practice.

2. MicroRNA in Hematopoiesis and Hematologic Malignancies

Hematopoiesis is the process of blood cell formation [7,8] in which hematopoietic stem cells (HSCs) generate and form blood cells, including multi-potent progenitors (MPPs) [9]. This continuous process is dependent on the proper expression of transcription factors and consists of several phases [10]. HSCs differentiate into either common myeloid progenitors (CMPs) or common lymphoid progenitors (CLPs) [11]. CMPs later develop into megakaryocytic and myeloid lineages, while CLPs give rise to B- and T-lymphocytes and NK-cells [12,13]. Mounting evidence shows that hematopoietic lineage differentiation is regulated by miRNAs [14,15,16].

2.1. MiRNA Biology

MiRNAs represent a group of small non-coding RNA molecules consisting of approximately 22 nucleotide sequences with key regulatory functions. Their post-transcriptional repression of relevant mRNA targets plays a crucial role in the regulation of various biological processes, including cell cycle, differentiation, growth, and cell death [17,18]. Most miRNA sequences are localized in introns or exons of non-coding RNA molecules. However, some miRNAs originate from introns in pre-mRNA (mirtrons). Different miRNA localizations determine canonical or non-canonical pathways [19]. In most cases, miRNA genes are transcribed in the nucleus by RNA polymerase II to primary miRNA (pri-miRNA), followed by cleavage to the precursor (pre-miRNA) structures. Further exporting to the cytoplasm and cleavage by the Dicer complex to small dsRNAs enables their binding to Argonaute proteins (AGO) and the RISC complex, leading to mature single-stranded miRNAs [20]. The canonical mechanism of miRNA biogenesis is dependent on Drosha and DGCR8 proteins, while some non-canonical pathways (e.g., mirtrons) are Drosha- and Dicer-independent processes [21].
Mature miRNAs recognize target mRNAs and mediate mRNA silencing or can be transported from the cell cytoplasm into extracellular circulation. Studies show much higher stability of extracellular miRNAs in comparison to cellular miRNAs [22]. Recently, circulating or cell-free miRNAs analyzed in body fluids represented potential biomarkers of various pathological states, including cancer diagnosis, prognosis, and therapy [23]. Besides their associations with proteins, especially AGO2, extracellular miRNAs can be transported in vesicles, such as exosomes, apoptotic bodies, and microvesicles [24]. Exosomes may also contain mature or pre-miRNA forms since some cancer exosomes contain a protein processing complex (RISC Loading complex) and thus represent cell-independent miRNA maturation [25]. As documented, exosomes function as traffic vehicles to the surrounding microenvironment. They can be received by recipient cells, and subsequent miRNA releasing could mediate changes in gene expression of target genes [26]. Studies concerning the exosomal miRNAs reveal their involvement in cancer pathogenesis with an emerging clinical potential [26].

2.2. The Role of microRNAs in Normal Hematopoiesis

The mechanism of miRNA biosynthesis includes a lot of components. During hematopoiesis, loss of Drosha or DGCR in the Dicer-independent mechanism impacts the way miRNAs mature. The absence of AGO2 deactivates the process of miRNA biosynthesis and expression [2,9].
In normal hematopoiesis, miRNAs regulate the differentiation, state, function, and self-renewal ability of HSCs, apoptosis levels, and differentiation of myeloid and lymphoid progenitor cells [27]. Moreover, miRNAs regulate the erythropoiesis, megakaryopoiesis, granulopoiesis, and differentiation of B- and T-lymphocytes [10]. According to the findings, miR-125b regulates the hematopoietic differentiation of HSCs through the modulation of transcription factors for B- and T-cell differentiation via targeting BLIMP-1, IRF-4, IFNG, IL2RB, IL10RA, and PRDM1 [28]. The normal cycling and self-renewal of HSCs are regulated by miR-126 [29] and miR-33 [30], while miR-125b promotes the expansion of HSCs [31].
MiR-155 controls the development of myeloid lineage and differentiation of erythroid progenitor cells [32,33]. Fontana and colleagues found that the monocyte differentiation was regulated by miRNAs 17-5p, 20a, and 106a [34]. Furthermore, miR-223 controlled myeloid differentiation through the LMO, MEF2C, NFI-A, and E2F1 transcription factors [28]. MiR-223 also regulates the differentiation and maturation of granulocytes [35]. Elevated expression promotes the development of mature granulocytes from myeloid progenitors [36]. Basal granulopoiesis is regulated by miR-125a. In addition, miR-125a regulates the production of neutrophils via the G-CSF signaling pathway [37]. Pro-myelocytic cells differentiate into monocytes via cooperation between miR-424 and transcription factors of the myeloid lineage [36].
MiR-146a activates the innate immunity targets involved in megakaryopoiesis [38]. Suppressing the inflammatory cytokines from innate immune cells by miR-146a inhibits the development of megakaryocytes [39]. Overexpression of miR-22 promotes the differentiation of megakaryocyte cells [40]. Zini and colleagues demonstrated that the lineage of granulocytes and megakaryocytes was modulated by the expression of miR-382-5p via downregulation of MXD1. Overexpression of miR-382-5p in CD34(+) HSCs resulted in decreased levels of megakaryocyte precursors while granulocytes were elevated [41]. The study of miR-22 functions in hematopoiesis discovered that the miR-22/MECOM (EVI1) regulated macrophage differentiation, and that MECOM mRNA degradation was mediated by miR-22 [42].
Differentiation of primitive erythroid progenitors from multipotent hematopoietic stem cells drives erythropoiesis. Overexpression of miR-486 multiplies erythroid proliferation and differentiation [43]. The inhibition of miR-150 expression levels enhances terminal erythropoiesis [44]. Kotaki et al. found that in the mouse model, the differentiation of erythrocytes was inhibited by miR-669m overexpression. Using bioinformatic analyses, authors showed that miR-669m targeted the Akap7 and Xk and inhibited erythroid hematopoiesis [45]. The experimental study by Xu et al. demonstrated the impact of miR-451 on the development of erythrocytes. Erythropoiesis was inhibited by miR-144 in CD34+ cells [46]. The mechanisms of the miRNA biogenesis pathway affect erythropoiesis. MiR-451 with Ago2-dependent and Dicer-independent processes regulate the maturation of erythrocytes [47,48].
MiR-128-2 overexpression affects the development of CLPs and immature lymphocytes [14,15]. MiR-24 inhibits B-cell differentiation and induces lymphoid cell apoptosis [49]. Lymphopoiesis of the B-cell lineage is inhibited by the miR-23a cluster [50]. MiR-150 regulates the differentiation of T lymphocytes. During the maturation of the T-cells, miR-150 is upregulated, but during T-cell differentiation into Th1 and Th2 subtypes, miR-150 expression is downregulated [51].

2.3. MicroRNA Dysregulation in Malignant Hematopoiesis

The development of hematologic malignancies is linked to chromosomal translocations, mutations, and epigenetic regulation, leading to dysregulation of tumor suppressor or oncogenic pathways. Han et al. reported that alterations in mechanisms of miRNA expression, and thereby in post-transcriptional miRNA regulations, represent critical events in malignant hematopoiesis [52]. Most miRNAs play an oncogenic (oncomiRs) or tumor-suppressive role, but in some cases, miRNAs might have a dual function [5].
According to the findings, abnormal miRNA expression is caused by chromosomal aberration, epigenetic deregulation of miRNA, and alterations in miRNA biosynthesis [53,54]. In addition, DNA methylation of tumor suppressor miRNAs and histone modification represent epigenetic mechanisms, which also contribute to malignant hematopoiesis [55]. Aggire et al. found that miR-124a expression was associated with decreased levels of 3mK4H3 and elevated 2mK9H3 markers, which promote the closed chromatin structure [56].
An aberrant miR-126 transcription plays a role in the leukemogenesis of acute myeloid leukemia (AML). An increased miR-126 expression was documented in a subpopulation of leukemic stem-like cells, which correlated with poor survival and a higher chance of relapse in AML patients [57]. As shown, miR-3662 reduced the growth and survival of AML cells in vitro. Interaction of miR-3632 and HBS1L–MYB regions located at the 6q23.3 chromosome affected hematopoiesis, especially erythroid differentiation, and leukemogenesis [58]. Lu et al. documented a dramatic upregulation of miRNA-301b-3p related to increased proliferation and inhibited apoptosis in AML cells [59]. In chronic myelogenous leukemia (CML), leukemogenesis is promoted by miR-17-92 targeting the A20 gene via the NF-kB signaling pathway. Jia et al. described the mechanism of aberrant leukemogenesis by miR-17-92 in the mouse model of CML, showing that overexpressed A20 induced apoptosis and inhibited cell proliferation [60]. The study of the long non-coding RNA (lncRNA) MALAT1/miR-328 axis provided data that CML development was suppressed after miR-328-dependent MALAT1 knockdown inhibited proliferation and led to cell cycle arrest [61]. Furthermore, miR-146a displays antitumor activity in myeloid leukemia via regulation of the NF-κB pathway by targeting IRAK1 and TRAF6 transcripts. C-miR-146a, a miR-146a mimic oligonucleotide conjugated to a scavenger receptor/Toll-like receptor 9 agonist reduces the expression of miR-146a targets and blocks NF-κB activation [62]. The correlation between aberrant miRNA expression and reduced apoptosis was observed in several hematologic malignancies [5,63]. In the human megakaryoblastic leukemia cell line MEG-01, derived from a patient with CML, expression of miR-15a and miR-16-1 promoted apoptosis by repression of the anti-apoptotic BCL-2 gene [64]. The results from in vitro and in vivo analyses showed that miR-125b inhibited the apoptosis in hematopoietic cells via repressing the Trp53inp1 gene [65].
Several studies focused on the variable expression of miRNA in myeloproliferative neoplasms. A comparison of miRNA expression levels in patients and healthy controls showed upregulated levels of miR-125b-5p and miR-125a-5p in polycythemia vera (PV) as well as in essential thrombocythemia (ET) patients. These findings highlight the possible role of aberrant miR-125 expression in the phenotype of patients with myeloproliferative malignancies [66]. Bruchova et al. investigated the relationship between dysregulated miRNAs and PV pathophysiology. The results documented the downregulation of let-7a and upregulation of miR-182 in granulocytes of PV patients. Furthermore, upregulation of miR-143, miR-145, miR-223, and high miR-26b levels were found in PV mononuclear cells and platelets, respectively. On the other hand, reticulocytes from PV patients showed miR-30b, miR-30c, and miR-150 downregulation [67]. Gebauer et al. aimed to identify potential miRNA biomarkers for differentiation between the subtypes of myeloproliferative neoplasia, including PV, ET, and early primary myelofibrosis (PMF). According to the findings, alterations in the expression of miR-10a and miR-150 were demonstrated for ET and PMF and also for PV and PMF, respectively. Moreover, the authors observed a correlation between miR-150 with a JAK2 allele burden and peripheral blood counts [68].
Kuang et al. studied the impact of miR-378 overexpression on myelodysplastic syndrome (MDS), showing that elevated levels of miR-378 promoted apoptosis and inhibited the growth of leukemic cells by activating the intrinsic and extrinsic pathways [69].
MiR-21 and miR-221/222 are oncogenes allowing increased cell proliferation and growth in multiple myeloma (MM) [70,71], while miR-342-3p suppresses MM development [72]. Long et al. revealed that miR-140-3p acts as a tumor suppressor by decreasing the proliferation and inducing the apoptosis of MM cells [73].
In T-cell acute lymphoblastic leukemia (T-ALL), DNA methylation in the CpG promoter region leads to miR-204 downregulation, while miR-204 overexpression in vitro inhibits the proliferation and enhances the apoptosis of T-ALL cells [74]. In CLL, reduced expression of pro-apoptotic miR-15a/16-1 is critical for cell survival. Kasar et al. reported the relationship between miR-15a/16 overexpression, followed by increased apoptosis of malignant cells and inhibition of histone deacetylation and the knockdown of B-cell-specific activator protein [75]. Recently, decreased levels of miR-125a and miR-223, together with an increased expression of their targets BCL-2 and STAT3, were identified in CLL patients [76].
Tumor suppressor miR-34a was found methylated in 75% of lymphoma cell lines [77]. MiR-155 regulates histone deacetylase 4 and transcriptional machinery in B-cell lymphoma 6 (BCL6), leading to leukemogenesis [78]. In aggressive B-cell lymphomas, miR-29 suppresses tumorigenesis through the knockdown of histone acetylation [79]. Meyer et al. found the inhibition of TAL1 expression by miR-17-92 cluster in primary human CD34+ cells. Deregulation in hematopoietic stem cells, caused by the TAL1 transcriptional complex, impaired erythroid differentiation. At the erythroid lineage, differentiation and proliferation of HSCs are modulated by the miR-17-92/TAL1 complex and can promote lymphoma development [80]. Zhou et al. found that cell proliferation and apoptosis of mantle cell lymphoma were suppressed by increased expression of miR-223 [81].
More details about miRNAs and their targets participating in hematologic malignancies are summarized in Table 1.

2.4. Aberrant microRNA Expressions in Diagnosis, Prognosis, and Monitoring of Therapy

Due to the origin of hematologic malignancies in the bone marrow and lymph nodes, miRNA expression analyses were performed in peripheral blood, plasma and serum, and bone marrow aspirates. MiRNA passes into circulation directly via apoptotic or necrotic events, or they are actively secreted by both normal and cancer cells. MiRNAs become highly stable when they are encapsulated in microvesicles or exosomes. This allows better miRNA transport and mediates the cell–cell communication between cancer cells, as well as between cancer cells and surrounding non-malignant cells [88,89].
Similarly to other cancers, an increasing number of exosomes was found in hematologic malignancies compared to normal blood samples. They extensively influence numerous pathological processes, including tumor survival, progression, and therapy resistance [90,91]. Moreover, exosomal miRNAs could possess a smaller amount of unspecific background compared to free extracellular miRNAs [92]. Several studies concerning hematologic malignancies assessed the correlations between exosomal miRNAs and diagnosis, prognosis, and chemotherapy resistance [93,94,95,96,97,98]. The results of aberrant miRNA expressions in acute and chronic leukemias, lymphomas, and myelomas, whose associations were evaluated with regard to any clinical consequences, are summarized in Table 2.
The majority of presented miRNA studies were conducted in pediatric patients with a diagnosis of the most common pediatric malignancy, ALL, in BM aspirate samples [100,101,102,103,104,105,106,107,108]. The researchers identified specific miRNAs for discrimination between several genetic, and T-ALL and B-ALL subtypes [104,105]. Other up- and downregulated miRNAs are associated with the prediction of early relapse [107] and drug resistance [102,103,104]. In adults with acute leukemias, upregulated miR-128a and miR-128b together with downregulated let-7b and miR-223 discriminated ALL from the most common AML in adult patients [109]. Furthermore, miR-363 seems to be a prognostic factor for optimal therapeutic strategy in AML. In patients with higher miR-363 expression in PB samples, early allogeneic transplantation (allo-HSCT) was recommended as a more effective strategy compared to chemotherapy but improved OS was found in those with a low level of miR-363 [110]. Several adult CML studies identified a single or set of specific miRNAs, which could be useful for the prediction of imatinib response [120,121,123,125]. In MM patients, the aberrant expression of other miRNA was found mostly in association with shorter PFS and OS [95,127,129,130,131,133,134,135,137] and bortezomib response [94,127]. In patients with leukemias and lymphomas, the opposite tendency in the expression of specific miRNAs was detected after the chemotherapy compared to the state before treatment. These findings might be used for therapy management.
Among the summarized results, most were performed in malignant samples. Only 30% of studies investigated miRNA expressions in plasma or serum samples, where the extracellular miRNA molecules originating from both cancer and normal cells coexist. It is generally assumed that exosomes are secreted mainly by live cells, which influence many physiological and pathological processes. Thereby, exosomal miRNA from patients with hematologic malignancies could be less contaminated by the background from the apoptotic and necrotic cells [89]. Moreover, several panels of exosomal miRNA were identified for early detection, discrimination of different subtypes, and prediction of drug resistance [89,93,94].

3. MicroRNA in Hematopoietic Stem Cell Transplantation

3.1. Hematopoietic Stem Cell Transplantation

Hematopoietic stem cell transplantation (HSCT) is a routine procedure for treating malignant and non-malignant diseases [138]. The pre-transplant course, known as the conditioning regimen, consists of chemo- or radiotherapy, or their combination, aiming to eradicate cancer cells and prepare recipient bone marrow for new donor cells [139]. Transfer of healthy HSCs helps to augment the function of the recipient bone marrow to generate functional cells [140]. For autologous transplantation (auto-HSCT), stem cells are obtained from the recipient before conditioning, cryopreserved, and then reinfused. On the other hand, allo-HSCT uses stem cells obtained from related/unrelated donors. Donors of HSCs might be family members, volunteers, or banked umbilical cord blood cells [141]. Human leukocyte antigen (HLA) matching between donors and recipients plays a key role in transplantation outcomes [142]. Over the past twenty years, data has documented increasing transplant rates for auto/allo- HSCT [143].
In the case of cord blood as a source of HSC, HLA matching requirements are less stringent due to the naive immunity of newborns. The risk of a rejected graft is less in the case of mismatched cord blood cells than transplantation of adult peripheral blood cells [144,145]. One of the main concerns of allo-HSCT is a high incidence of graft-versus-host disease (GvHD). GvHD is associated with the immunological attack and rejection of host tissue by donor alloreactive T lymphocytes [146]. However, in the case of identical twins as donor and recipient, there is no risk of GvHD and no failure of graft in the recipient [140].
Acute and chronic GvHD represent major specific post-transplant complications affecting patient life. The differences between these two forms are the time of onset, clinical features, and immunopathological mechanisms [147]. An acute GvHD (aGvHD) develops within the first 100 days post-transplantation and primarily affects the gastrointestinal tract, liver, skin, eyes, and oral mucosa [148]. A chronic GvHD (cGVHD) represents a delayed complication associated with transplantation, significantly reducing the quality of life [149]. The onset of cGvHD is more than 100 days post-transplantation, and the incidence varies from 30 to 70% of patients [150,151]. An elevated occurrence of cGvHD is documented in pediatric patients [152]. Transplant-caused mortality and both aGvHD and cGvHD were reduced in the case of T-cell depletion using antibodies. Results showed that T-cell depletion might prevent GvHD only in patients with early-stage leukemia and low relapse risk [153]. Timely diagnosis of GvHD is very important for patient outcomes, and early biomarkers for the prevention of morbid complications associated with HSCT need to be uncovered [154]. Recently, immune activation markers, organ-specific markers, and miRNAs as systemic markers are widely studied in routine use for GvHD prognosis and diagnosis [155,156].

3.2. MicroRNA and Graft-Versus-Host Disease

Several preclinical and clinical studies in various periods pre- and post-HSCT showed dysregulated miRNA levels and their function in massive immune responses leading to the development of GvHD [157,158]. A differently expressed miRNA profile appears to be a promising noninvasive tool for specific diagnosis and prediction of aGvHD occurrence (Figure 1).
MiR-146a and miR-155 are among the most studied miRNAs in association with GvHD. MiR-155 plays an important role in T-cell proliferation, affecting the T and B lymphocyte function. Significantly upregulated miR-155 was observed in the serum of hematologic cancer patients with aGvHD compared to non-aGvHD patients. Moreover, a higher miR-155 expression correlated with higher grade (III-IV) aGvHD patients and elevated levels of soluble IL-17, IL-9, and IFN-gamma factors. These are specific for donor T-cell activation and tissue damage during aGvHD [159]. Importantly, upregulated miR-155 was found also in endothelial microparticles from peripheral blood. The frequent presence of microparticles is common for GvHD, and the increasing peak of miR-155 levels in these particles occurred even before miR-155 elevation in T-lymphocytes. In addition, miR-155 inhibition induced aberrant Th and Treg cell differentiation. Delivery of miR-155 by endothelial microparticles is involved in specific functions of T-lymphocytes and could mediate the onset of aGvHD [160]. MiR-146a is a key controller of immune T-cell response. In a mouse model, the overexpression of miR-146a was associated with reduced GvHD severity caused by the negative regulation of T-cells from donors by targeting TNF receptor-associated factor 6 (TRAF6), and subsequent TNF transcription inhibition [161]. Moreover, authors observed that SNP polymorphism rs2910164 in the precursor miR-146a of HSCT donors and recipients predisposes them to a higher risk for severe aGvHD [161,162]. According to the findings, the downregulation of miR-146a in the 28-day period after allo-HSCT correlated with a higher incidence of aGvHD. Furthermore, the statistical model concerning miR-146a and miR-155 expression in the peripheral blood showed a significant biological synergy [163].
For a complex view, Crossland et al. analyzed miR-146a and miR-155 levels in skin and gastrointestinal biopsies and various types of body fluids, including serum and urine. In all sample types, higher levels of both miRNAs were detected in aGvHD patients compared to non-GvHD patients. In addition, the upregulation of miR-146a and miR-155 in the 14-day period after HSCT was found in serum samples of patients who later developed aGvHD. Nevertheless, expression results of both miR-146a and miR-155 supported their potential to be biomarkers for GvHD development [164].
In relationship to aGvHD, many authors found the upregulation of miR-423, miR-199a, miR-93, and miR-377 and detected an association between their higher expression and diagnostic or prognostic ability and GvHD severity [165,166]. A broad-range miRNA profiling study analyzing 799 mature miRNAs in serum samples detected 61 miRNAs with different expressions in aGvHD. Among them, 10 miRNAs were re-analyzed in another patient cohort. Significant upregulation of miR-20a and miR-15a and downregulation of miR146a, miR-30b, miR-374-5p, and miR-181a were verified in aGvHD patients. However, receiver operating characteristic (ROC) analysis suggested diagnostic potential only for miR-30b, miR-374-5p, and miR-15a expression. On the other hand, prognostic significance demonstrated upregulation of miR-18, miR-19a, miR-19b, miR-20a, miR-146a, and miR-451 in a 14-day post-HSCT period before GvHD onset [167]. Xie et al. found contradictory results, showing higher miR-181a expression in aGvHD compared to non-aGvHD patients. Moreover, the results found a correlation between the severity of GvHD and a positive or negative correlation with serum IL-2, IL-17a, IL-22, and IL-13 cytokines. Further investigations are needed to elucidate the role of miR-181a as a promising GvHD biomarker [168].
Upregulation of miR-194, miR-518f, miR-29a, miR-586, miR-153, and miR-548a and downregulation of miR-455 and miR-5787 in plasma samples significantly mediated aGvHD development in post-HSCT patients [169,170,171,172,173]. Zhang et al. evaluated a diagnostic value of novel serum miRNAs in aGvHD, including lower miR-28 and higher miR-489 and miR-671 expression levels. A predictive model for increased GvHD risk identified two downregulated miR-26b and miR-374a. In addition, the results uncovered the role of miR-411 in aGvHD monitoring since the decreased expression of miR-411 was detected in the case of aGvHD development, compared to higher miR-411 levels when aGvHD was under control [174]. Exosomal miRNA expression represents a promising diagnostic biomarker for late-onset aGvHD. Using a low-density miRNA array, differences in expression profiles of 55 miRNAs were identified between late-onset aGvHD and non-aGvHD. Higher expression of miR-128 was validated for diagnosis of late-onset aGvHD. As known, miR-128 target genes play a role in inflammation and immune response pathways [175].
The changes in miRNA expression profiles were also detected in cutaneous aGvHD. Differentially expressed let-7c, miR-503, miR-365a, miR-34a-5p, and miR-34a-3p were shown in pre-HSCT, post-HSCT, and control skin biopsies [176].
Studies concerning diagnostic and prognostic biomarkers for cGvHD are still missing [177]. Recent results identified the distinction between miRNAs in cGvHD patients. The regression model and bioinformatic analysis showed that the upregulation of miR-365a, miR-148a, and miR-378a in plasma samples had diagnostic relevance for patients one year post-HSCT with cGvHD symptoms [178]. In plasma extracellular vesicles (EVs), Lacina et al. detected differentially expressed miRNA characteristics for cGvHD. Upregulation of miR-29c and downregulation of miR-630 and miR-374b were found in cGvHD compared to non-cGvHD patients [179].

3.3. MicroRNA and Post-HSCT Patient Outcomes

MiRNA expression profiling suggests that numerous miRNAs are associated with mortality risk in HSCT patients. The group of high-risk patients with a pre-transplant comorbidity index score ≥4 compared to low-risk patients with index score 0 showed significantly downregulated miR-374b and miR-454 but upregulated miR-142, miR-191, miR-424, miR-590, miR-15b, and miR-29c. These results outline the remarkable predictive potential of miRNA expression for patients’ outcomes [180]. Cheng et al. noticed that AML patients who underwent allo-HSCT and showed high miR-99a expression had poor outcomes. MiR-99a upregulation represented an unfavorable prognostic marker for EFS and OS in multivariate analysis [181]. In addition, the miRNA expression profile can stratify patients who profited from allo-HSCT or chemotherapy. A better OS and EFS were detected in the AML group receiving allo-HSCT and expressing low levels of miR-425 compared to patients undergoing chemotherapy [182]. Furthermore, miR-363 could serve as a prognostic marker for an appropriate therapeutic strategy with improved clinical outcomes in the allo-HSCT group, since higher miR-363 expression was associated with improved OS and EFS [110].
Survival analyses were included in several clinical studies concerning the relationship between miRNA expressions and GvHD. Four-miRNA panels consisting of miR-423, miR-199a-3p, miR-93, and miR-377 were analyzed in plasma samples and showed that high-risk patients with upregulated miRNAs had a poorer outcome than low-risk patients. Moreover, the elevated expression of miRNA panels appeared as an independent prognostic factor for shorter OS in aGvHD patients [165]. In contrast, higher expression of miR-19b, miR-20a, and miR-30b at the time of diagnosis was associated with improved OS [167].
The association between the miRNA profile and cutaneous aGvHD was investigated in skin samples, showing that underexpressions of miR-503 and miR-34a-3p were implicated in improved OS in post-HSCT patients. The results documented overexpression of miR-503, mir-34a-5p, and miR-34a-3p in the serum of aGvHD patients, suggesting their potential use as circulatory biomarkers for cutaneous aGvHD development [176].
Lim et al. investigated the association between miRNA expression from EVs and clinical complications of allo-HSCT patients. Several correlations with infectious complications were observed for five different EV miRNAs, miR-223-3p, miR-21-5p, miR-23a-3p, miR-375, miR-423-5p, for positive, and miR-425-5p, miR-342-3p, miR-320b, miR-454-3p, miR-151a-3p for negative correlations, respectively [183]. Allo-HSCT with myeloablative conditioning is frequently accompanied by post-transplantation complications, including gastrointestinal toxicity, mucositis, and overall inflammation. Upregulation of miR-155 and downregulation of miR-146a are significantly associated with markers of gastrointestinal toxicity, with IL-6 and CRP representing plasma inflammatory factors. According to the findings, these miRNAs could play a critical role in inflammation response [184].
MiRNA expression was shown to influence engraftment efficacy. Higher expression of miRNA-15a, miRNA-16, miRNA-126, and miRNA-146a before auto-HSCT and early post-HSCT were positively correlated with a longer period until engraftment in patients with MM and lymphoma [185]. Rafiee et al. observed a specific correlation between the upregulation of miR-155 and platelet and neutrophile engraftment, suggesting miR-155 as a potential predictor of auto-HSCT patient outcome [186]. Similarly, higher expression of miR-193a-5p in MM patients before auto-HSCT predicted a reduction in early relapse and better PFS [187].

4. Investigating the Therapeutical Potential of the miRNA-Based Approach in Hemato-Oncology

Higher expression of oncogenic miRNA might be regulated by several strategies [52]. AntagomiRs represent antisense oligonucleotides prepared by genetic engineering, which are capable of miRNA silencing [188]. Dorrance et al. suggested the potential therapeutic option of antisense miRNA in AML patients, showing that antagomiR-126 reduced the number of leukemia stem cells in vivo [189]. According to these findings, treatment with imatinib led to the upregulation of miR-21 in Philadelphia chromosome-positive ALL cells. The application of antagomiR-21 demonstrated positive results in sensitizing cells to imatinib, inducing cancer cell apoptosis, and inhibiting the phosphatidylinositol-3-OH kinase/AKT (PI3K/AKT) pathway [190]. CBFB-MYH11 (CM) gene fusion upregulated the levels of miR-126 in AML, which led to the survival of leukemia-initiating cells. As shown, the genetic deletion of miR-126 prolonged survival in a murine model with CM gene fusion. Treatment with therapeutic anti-miR-126 in combination with chemotherapy effectively inhibited AML cell progression [191]. The positive effect of antagomiR-196b and antagomiR-21 was documented in the MLL-AF9–initiated leukemia-murine model with no changes in murine behavior or vital organ functions [192]. The results from an in vitro study on diffuse large B-cell lymphoma (DLBCL) cell lines revealed an attenuated proliferation and increased apoptosis after the application of an oligonucleotide miR-155 inhibitor. In addition, the delivery of the miR-155 inhibitor into a DLBCL-bearing murine model led to a decrease in tumor volume [193].
Accumulating evidence suggests that circular RNAs (circRNAs) also play a key role in hematopoiesis and hematologic malignancy initiation [194] and could be exploited in therapy. CircRNAs represent non-coding RNAs that contain a covalently closed loop structure. In the vast majority of eukaryotes, the loop originates mainly from the reverse splicing of pre-mRNA transcripts [195]. CircRNAs have multiple binding sites for miRNAs and, among other functions, could act as endogenous miRNA sponges by sequestering and preventing them from interacting with their target mRNAs [196]. Recent studies have pointed out the possibilities of using circRNA in the inhibition of miRNAs, which are related to cell proliferation, apoptosis, invasion, and migration of cells in hematologic malignancies. Han et al. revealed the ability of circ-0001947 to suppress the proliferation of AML cells by inhibiting miR-329-5p [197]. The expression of circ-KEL, in turn, can inhibit apoptosis of AML via sponging miR-335-5p [198], and circ-0009910 was associated with inhibition of apoptosis by sponging miR-34a-5p and miR-5195-3p in CML and AML, respectively [199,200]. In addition, circ-0069767 was found to reduce the invasive and migratory capacities of MM cells by sponging miR-636 with an impact on the regulation of K-RAS expression [201].
On the other hand, numerous miRNAs were downregulated in hematologic malignancies, leading to worse treatment outcomes. A significantly lower level of miR-29b was detected in blood mononuclear cells from patients with AML or CML, which correlated with poor treatment-free survival [202,203]. Garzon et al. documented the tumor suppressor effect of synthetic miR-29b oligonucleotides as a potential therapy option for AML. Synthetic miR-29b directly injected into murine tumors reduced tumor growth. Results from in vitro experiments showed the association between transfection of synthetic miR-29b and downregulation of anti-apoptotic MCL-1, leading to reduced cell growth [83]. Furthermore, synthetic miR-29b oligonucleotides effectively downregulated the expression of DNMT1, DNMT3A, and DNMT3B in AML cells. Prepared hypomethylating compound miR-29b increased promoter hypomethylation and reactivated the expression of p15INK4b and ESR1 genes [204]. A previous study by Blum et al. showed that the reexpression of both genes was associated with clinical response to therapy in leukemic patients treated with decitabine [205].
A higher expression of the ST8SIA4 gene correlates with leukemia multidrug resistance via the elevated activity of the PI3K/AKT pathway. In vivo experiments confirmed that the upregulation of miR-181c in combination with adriamycin significantly decreased tumor growth in adriamycin-resistant mice. Decreased level of ST8SIA4 was observed in tumors injected with miR-181c. Based on this result, inhibition of the PI3K/AKT pathway helped to target sensitive cancer cells in treatment and suppressed drug resistance in a murine model [206].
Nevertheless, free synthetic miRNA might be degraded in biofluids, and its cellular uptake is limited. The nanoparticle delivery system represents a novel strategy for miRNA delivery. Transferrin-conjugated nanoparticle delivery for synthetic miR-29b (Tf-NP-miR-29b) led to a 200-fold increase in mature miR-29b. In a leukemic murine model, treatment with Tf-NP-miR-29b led to more prolonged survival than in mice treated with free miR-29b. Moreover, other miR-29b targets were decreased, including KIT, DNMT3A, DNMT3B, DNMT1, SP1, CDK6, and FLT3 [207]. In another study, Huang et al. used lipopolyplex nanoparticles for more efficient delivery of miR-181a into KG1a, MV4-11, and OCI-AML cells. In a leukemic murine model, the administration of Tf-NP-miR-181a caused an increased level of miR-181a, reduced tumor burden, and prolonged mice survival [208]. Since higher expression of miR-181a in patients with AML correlated with longer survival and full remission compared to patients with low miR-181a levels [209], increasing miR-181a might bring a potential clinical benefit.

5. Conclusions and Future Directions

Due to the aggressive nature of hematologic malignancies, early and accurate diagnosis is the key to improving prognosis, treatment results, and patient survival. The discovery of miRNAs triggered intensive research aiming to determine their role in normal and malignant hematopoiesis. Altered miRNA levels in tumor and healthy cells enable the potential use of circulating miRNAs as diagnostic biomarkers. Mounting evidence describes the role of miRNAs in post-transplant complications, including mainly GvHD, which is the primary cause of premature mortality in patients receiving HSCT. Early detection of aGvHD is critical for prolonging patients’ OS. Since diagnostic and prognostic biomarkers for GvHD have not been determined yet, this research area represents a crucial experimental and medical challenge. Circulating miRNA levels in plasma or EVs represent a promising tool not only for aGvHD but also for cGvHD diagnosis, prognosis, and prediction.
Recently, the identification of predictive exosomal miRNA panels for drug resistance opens up possibilities for potential intervention. In this context, activation or inhibition of deregulated miRNAs appears to be an attractive therapeutic option for improving treatment efficacy. Animal models and in vitro findings evaluated the clinical significance of reducing oncogenic miRNAs by synthetic antagomiRs and restoring critically reduced levels of tumor-suppressor miRNAs through mimetics. A proper understanding of existing correlations is needed prior to routine clinical application. The research is particularly complicated in the case of dual miRNAs, showing both oncogenic and tumor suppressor activity. Safety and an effective way of delivery represent the major concerns.
Although miRNA’s association with HSCT, post-transplant complications, and patient outcome is a relatively new research area, some results have suggested miRNAs as potentially relevant prognostic and predictive markers. However, further investigations of miRNA dysregulation in hematopoiesis may bring novel strategies in miRNA-based approaches to improve outcomes for hematologic cancer patients.

Author Contributions

Conceptualization, S.C.; writing—original draft preparation, A.S., S.C., I.F., N.N., L.K., R.O, M.M.; writing—review and editing, S.C., L.K., A.S., R.O.; visualization, A.S., S.C., I.F., N.N.; project administration, S.C.; funding acquisition, S.C. All authors have read and agreed to the published version of the manuscript.

Funding

The study was funded by the Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences (VEGA), contract No. 2/0069/22. The funding source had no influence on the writing of the manuscript.

Acknowledgments

We greatly thank Michal Mego for critically reading and reviewing Figure 1 which was created with Biorender.com. Moreover, we would like to thank Renee Vick for reading the manuscript carefully and helping with language editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef] [PubMed]
  2. Nikitina, E.G.; Urazova, L.N.; Stegny, V.N. MicroRNAs and human cancer. Exp. Oncol. 2012, 34, 2–8. [Google Scholar] [PubMed]
  3. Han, T.S.; Hur, K.; Cho, H.S.; Ban, H.S. Epigenetic Associations between lncRNA/circRNA and miRNA in Hepatocellular Carcinoma. Cancers 2020, 12, 2622. [Google Scholar] [CrossRef] [PubMed]
  4. Anelli, L.; Zagaria, A.; Specchia, G.; Musto, P.; Albano, F. Dysregulation of miRNA in Leukemia: Exploiting miRNA Expression Profiles as Biomarkers. Int. J. Mol. Sci. 2021, 22, 7156. [Google Scholar] [CrossRef] [PubMed]
  5. Gounaris-Shannon, S.; Chevassut, T. The Role of miRNA in Haematological Malignancy. Bone Marrow Res. 2013, 2013, 269107. [Google Scholar] [CrossRef] [PubMed]
  6. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed]
  7. O’Connell, R.M.; Baltimore, D. MicroRNAs and hematopoietic cell development. Curr. Top. Dev. Biol. 2012, 99, 145–174. [Google Scholar] [PubMed]
  8. Jagannathan-Bogdan, M.; Zon, L.I. Hematopoiesis. Development 2013, 140, 2463–2467. [Google Scholar] [CrossRef] [PubMed]
  9. Kotaki, R.; Koyama-Nasu, R.; Yamakawa, N.; Kotani, A. miRNAs in Normal and Malignant Hematopoiesis. Int. J. Mol. Sci. 2017, 18, 1495. [Google Scholar] [CrossRef] [PubMed]
  10. Lazare, S.S.; Wojtowicz, E.E.; Bystrykh, L.V.; de Haan, G. microRNAs in hematopoiesis. Exp. Cell Res. 2014, 329, 234–238. [Google Scholar] [CrossRef] [PubMed]
  11. Grinenko, T.; Eugster, A.; Thielecke, L.; Ramasz, B.; Kruger, A.; Dietz, S.; Glauche, I.; Gerbaulet, A.; von Bonin, M.; Basak, O.; et al. Hematopoietic stem cells can differentiate into restricted myeloid progenitors before cell division in mice. Nat. Commun. 2018, 9, 1898. [Google Scholar] [CrossRef] [PubMed]
  12. Akashi, K.; Traver, D.; Miyamoto, T.; Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 2000, 404, 193–197. [Google Scholar] [CrossRef] [PubMed]
  13. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, D.; Tanaka-Yano, M.; Meader, E.; Kinney, M.A.; Morris, V.; Lummertz da Rocha, E.; Liu, N.; Liu, T.; Zhu, Q.; Orkin, S.H.; et al. Developmental maturation of the hematopoietic system controlled by a Lin28b-let-7-Cbx2 axis. Cell Rep. 2022, 39, 110587. [Google Scholar] [CrossRef] [PubMed]
  15. Yang, Y.; Xu, J.; Chen, H.; Fei, X.; Tang, Y.; Yan, Y.; Zhang, H.; Zhang, J. MiR-128-2 inhibits common lymphoid progenitors from developing into progenitor B cells. Oncotarget 2016, 7, 17520–17531. [Google Scholar] [CrossRef]
  16. Roberts, L.B.; Jowett, G.M.; Read, E.; Zabinski, T.; Berkachy, R.; Selkirk, M.E.; Jackson, I.; Niazi, U.; Anandagoda, N.; Araki, M.; et al. MicroRNA-142 Critically Regulates Group 2 Innate Lymphoid Cell Homeostasis and Function. J. Immunol. 2021, 206, 2725–2739. [Google Scholar] [CrossRef]
  17. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  18. Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef]
  19. Saliminejad, K.; Khorram Khorshid, H.R.; Soleymani Fard, S.; Ghaffari, S.H. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J. Cell. Physiol. 2019, 234, 5451–5465. [Google Scholar] [CrossRef]
  20. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  21. Salim, U.; Kumar, A.; Kulshreshtha, R.; Vivekanandan, P. Biogenesis, characterization, and functions of mirtrons. Wiley Interdiscip. Rev. RNA 2022, 13, e1680. [Google Scholar] [CrossRef] [PubMed]
  22. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed]
  23. Chakraborty, C.; Das, S. Profiling cell-free and circulating miRNA: A clinical diagnostic tool for different cancers. Tumor Biol. 2016, 37, 5705–5714. [Google Scholar] [CrossRef]
  24. Iftikhar, H.; Carney, G.E. Evidence and potential in vivo functions for biofluid miRNAs: From expression profiling to functional testing: Potential roles of extracellular miRNAs as indicators of physiological change and as agents of intercellular information exchange. Bioessays 2016, 38, 367–378. [Google Scholar] [CrossRef]
  25. Melo, S.A.; Sugimoto, H.; O’Connell, J.T.; Kato, N.; Villanueva, A.; Vidal, A.; Qiu, L.; Vitkin, E.; Perelman, L.T.; Melo, C.A.; et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014, 26, 707–721. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, Z.; Shi, K.; Yang, S.; Liu, J.; Zhou, Q.; Wang, G.; Song, J.; Li, Z.; Zhang, Z.; Yuan, W. Effect of exosomal miRNA on cancer biology and clinical applications. Mol. Cancer 2018, 17, 147. [Google Scholar] [CrossRef]
  27. Lawrie, C.H. MicroRNAs in hematological malignancies. Blood Rev. 2013, 27, 143–154. [Google Scholar] [CrossRef] [PubMed]
  28. Rajasekhar, M.; Schmitz, U.; Flamant, S.; Wong, J.J.; Bailey, C.G.; Ritchie, W.; Holst, J.; Rasko, J.E.J. Identifying microRNA determinants of human myelopoiesis. Sci. Rep. 2018, 8, 7264. [Google Scholar] [CrossRef] [PubMed]
  29. Lechman, E.R.; Gentner, B.; Ng, S.W.; Schoof, E.M.; van Galen, P.; Kennedy, J.A.; Nucera, S.; Ciceri, F.; Kaufmann, K.B.; Takayama, N.; et al. miR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells. Cancer Cell 2016, 29, 214–228. [Google Scholar] [CrossRef]
  30. Herrera-Merchan, A.; Cerrato, C.; Luengo, G.; Dominguez, O.; Piris, M.A.; Serrano, M.; Gonzalez, S. miR-33-mediated downregulation of p53 controls hematopoietic stem cell self-renewal. Cell Cycle 2010, 9, 3277–3285. [Google Scholar] [CrossRef]
  31. Ooi, A.G.; Sahoo, D.; Adorno, M.; Wang, Y.; Weissman, I.L.; Park, C.Y. MicroRNA-125b expands hematopoietic stem cells and enriches for the lymphoid-balanced and lymphoid-biased subsets. Proc. Natl. Acad. Sci. USA 2010, 107, 21505–21510. [Google Scholar] [CrossRef] [PubMed]
  32. Ranganath, P. MicroRNA-155 and Its Role in Malignant Hematopoiesis. Biomark. Insights 2015, 10, 95–102. [Google Scholar] [CrossRef] [PubMed]
  33. Bissels, U.; Bosio, A.; Wagner, W. MicroRNAs are shaping the hematopoietic landscape. Haematologica 2012, 97, 160–167. [Google Scholar] [CrossRef] [PubMed]
  34. Fontana, L.; Pelosi, E.; Greco, P.; Racanicchi, S.; Testa, U.; Liuzzi, F.; Croce, C.M.; Brunetti, E.; Grignani, F.; Peschle, C. MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat. Cell Biol. 2007, 9, 775–787. [Google Scholar] [CrossRef] [PubMed]
  35. Hong, S.H.; Kim, K.S.; Oh, I.H. Concise review: Exploring miRNAs—Toward a better understanding of hematopoiesis. Stem Cells 2015, 33, 1–7. [Google Scholar] [CrossRef]
  36. El Gazzar, M. microRNAs as potential regulators of myeloid-derived suppressor cell expansion. Innate Immun. 2014, 20, 227–238. [Google Scholar] [CrossRef]
  37. Qin, Y.; Wu, L.; Ouyang, Y.; Zhou, P.; Zhou, H.; Wang, Y.; Ma, J.; Zhang, J.; Chen, Y.; Qian, J.; et al. MiR-125a Is a critical modulator for neutrophil development. PLoS Genet. 2017, 13, e1007027. [Google Scholar] [CrossRef]
  38. Undi, R.B.; Kandi, R.; Gutti, R.K. MicroRNAs as Haematopoiesis Regulators. Adv. Hematol. 2013, 2013, 695754. [Google Scholar] [CrossRef]
  39. Opalinska, J.B.; Bersenev, A.; Zhang, Z.; Schmaier, A.A.; Choi, J.; Yao, Y.; D’Souza, J.; Tong, W.; Weiss, M.J. MicroRNA expression in maturing murine megakaryocytes. Blood 2010, 116, e128–e138. [Google Scholar] [CrossRef]
  40. Weiss, C.N.; Ito, K. microRNA-22 promotes megakaryocyte differentiation through repression of its target, GFI1. Blood Adv. 2019, 3, 33–46. [Google Scholar] [CrossRef]
  41. Zini, R.; Rossi, C.; Norfo, R.; Pennucci, V.; Barbieri, G.; Ruberti, S.; Rontauroli, S.; Salati, S.; Bianchi, E.; Manfredini, R. miR-382-5p Controls Hematopoietic Stem Cell Differentiation through the Downregulation of MXD1. Stem Cells Dev. 2016, 25, 1433–1443. [Google Scholar] [CrossRef] [PubMed]
  42. Shen, C.; Chen, M.T.; Zhang, X.H.; Yin, X.L.; Ning, H.M.; Su, R.; Lin, H.S.; Song, L.; Wang, F.; Ma, Y.N.; et al. The PU.1-Modulated MicroRNA-22 Is a Regulator of Monocyte/Macrophage Differentiation and Acute Myeloid Leukemia. PLoS Genet. 2016, 12, e1006259. [Google Scholar] [CrossRef] [PubMed]
  43. Shi, X.F.; Wang, H.; Kong, F.X.; Xu, Q.Q.; Xiao, F.J.; Yang, Y.F.; Ge, R.L.; Wang, L.S. Exosomal miR-486 regulates hypoxia-induced erythroid differentiation of erythroleukemia cells through targeting Sirt1. Exp. Cell Res. 2017, 351, 74–81. [Google Scholar] [CrossRef]
  44. Sun, Z.; Wang, Y.; Han, X.; Zhao, X.; Peng, Y.; Li, Y.; Peng, M.; Song, J.; Wu, K.; Sun, S.; et al. miR-150 inhibits terminal erythroid proliferation and differentiation. Oncotarget 2015, 6, 43033–43047. [Google Scholar] [CrossRef]
  45. Kotaki, R.; Kawashima, M.; Yamaguchi, A.; Suzuki, N.; Koyama-Nasu, R.; Ogiya, D.; Okuyama, K.; Yamamoto, Y.; Takamatsu, M.; Kurosaki, N.; et al. Overexpression of miR-669m inhibits erythroblast differentiation. Sci. Rep. 2020, 10, 13554. [Google Scholar] [CrossRef] [PubMed]
  46. Xu, L.; Wu, F.; Yang, L.; Wang, F.; Zhang, T.; Deng, X.; Zhang, X.; Yuan, X.; Yan, Y.; Li, Y.; et al. miR-144/451 inhibits c-Myc to promote erythroid differentiation. FASEB J. 2020, 34, 13194–13210. [Google Scholar] [CrossRef] [PubMed]
  47. Kretov, D.A.; Walawalkar, I.A.; Mora-Martin, A.; Shafik, A.M.; Moxon, S.; Cifuentes, D. Ago2-Dependent Processing Allows miR-451 to Evade the Global MicroRNA Turnover Elicited during Erythropoiesis. Mol. Cell 2020, 78, 317–328.e6. [Google Scholar] [CrossRef] [PubMed]
  48. Yi, T.; Arthanari, H.; Akabayov, B.; Song, H.; Papadopoulos, E.; Qi, H.H.; Jedrychowski, M.; Guttler, T.; Guo, C.; Luna, R.E.; et al. eIF1A augments Ago2-mediated Dicer-independent miRNA biogenesis and RNA interference. Nat. Commun. 2015, 6, 7194. [Google Scholar] [CrossRef]
  49. Nguyen, T.; Rich, A.; Dahl, R. MiR-24 promotes the survival of hematopoietic cells. PLoS ONE 2013, 8, e55406. [Google Scholar] [CrossRef]
  50. Kong, K.Y.; Owens, K.S.; Rogers, J.H.; Mullenix, J.; Velu, C.S.; Grimes, H.L.; Dahl, R. MIR-23A microRNA cluster inhibits B-cell development. Exp. Hematol. 2010, 38, 629–640.e1. [Google Scholar] [CrossRef]
  51. Vasilatou, D.; Papageorgiou, S.; Pappa, V.; Papageorgiou, E.; Dervenoulas, J. The role of microRNAs in normal and malignant hematopoiesis. Eur. J. Haematol. 2010, 84, 1–16. [Google Scholar] [CrossRef] [PubMed]
  52. Han, Z.; Rosen, S.T.; Querfeld, C. Targeting microRNA in hematologic malignancies. Curr. Opin. Oncol. 2020, 32, 535–544. [Google Scholar] [CrossRef] [PubMed]
  53. Ali Syeda, Z.; Langden, S.S.S.; Munkhzul, C.; Lee, M.; Song, S.J. Regulatory Mechanism of MicroRNA Expression in Cancer. Int. J. Mol. Sci. 2020, 21, 1723. [Google Scholar] [CrossRef] [PubMed]
  54. Weiss, C.N.; Ito, K. A Macro View of MicroRNAs: The Discovery of MicroRNAs and Their Role in Hematopoiesis and Hematologic Disease. Int. Rev. Cell Mol. Biol. 2017, 334, 99–175. [Google Scholar] [PubMed]
  55. Raghuwanshi, S.; Dahariya, S.; Kandi, R.; Gutti, U.; Undi, R.B.; Sharma, D.S.; Sahu, I.; Kovuru, N.; Yarla, N.S.; Saladi, R.G.V.; et al. Epigenetic Mechanisms: Role in Hematopoietic Stem Cell Lineage Commitment and Differentiation. Curr. Drug Targets 2018, 19, 1683–1695. [Google Scholar] [CrossRef]
  56. Agirre, X.; Vilas-Zornoza, A.; Jimenez-Velasco, A.; Martin-Subero, J.I.; Cordeu, L.; Garate, L.; San Jose-Eneriz, E.; Abizanda, G.; Rodriguez-Otero, P.; Fortes, P.; et al. Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia. Cancer Res. 2009, 69, 4443–4453. [Google Scholar] [CrossRef]
  57. De Leeuw, D.C.; Denkers, F.; Olthof, M.C.; Rutten, A.P.; Pouwels, W.; Schuurhuis, G.J.; Ossenkoppele, G.J.; Smit, L. Attenuation of microRNA-126 expression that drives CD34+38- stem/progenitor cells in acute myeloid leukemia leads to tumor eradication. Cancer Res. 2014, 74, 2094–2105. [Google Scholar] [CrossRef]
  58. Maharry, S.E.; Walker, C.J.; Liyanarachchi, S.; Mehta, S.; Patel, M.; Bainazar, M.A.; Huang, X.; Lankenau, M.A.; Hoag, K.W.; Ranganathan, P.; et al. Dissection of the Major Hematopoietic Quantitative Trait Locus in Chromosome 6q23.3 Identifies miR-3662 as a Player in Hematopoiesis and Acute Myeloid Leukemia. Cancer Discov. 2016, 6, 1036–1051. [Google Scholar] [CrossRef]
  59. Lu, Y.; Zhong, L.; Luo, X.; Liu, C.; Dan, W.; Chu, X.; Wan, P.; Zhang, Z.; Wang, X.; Liu, Z.; et al. MiRNA-301b-3p induces proliferation and inhibits apoptosis in AML cells by targeting FOXF2 and regulating Wnt/beta-catenin axis. Mol. Cell Probes 2022, 63, 101805. [Google Scholar] [CrossRef]
  60. Jia, Q.; Sun, H.; Xiao, F.; Sai, Y.; Li, Q.; Zhang, X.; Yang, S.; Wang, H.; Wang, H.; Yang, Y.; et al. miR-17-92 promotes leukemogenesis in chronic myeloid leukemia via targeting A20 and activation of NF-kappaB signaling. Biochem. Biophys. Res. Commun. 2017, 487, 868–874. [Google Scholar] [CrossRef]
  61. Wen, F.; Cao, Y.X.; Luo, Z.Y.; Liao, P.; Lu, Z.W. LncRNA MALAT1 promotes cell proliferation and imatinib resistance by sponging miR-328 in chronic myelogenous leukemia. Biochem. Biophys. Res. Commun. 2018, 507, 1–8. [Google Scholar] [CrossRef] [PubMed]
  62. Su, Y.L.; Wang, X.; Mann, M.; Adamus, T.P.; Wang, D.; Moreira, D.F.; Zhang, Z.; Ouyang, C.; He, X.; Zhang, B.; et al. Myeloid cell-targeted miR-146a mimic inhibits NF-kappaB-driven inflammation and leukemia progression in vivo. Blood 2020, 135, 167–180. [Google Scholar] [CrossRef] [PubMed]
  63. Macfarlane, L.A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar] [CrossRef] [PubMed]
  64. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [PubMed]
  65. Enomoto, Y.; Kitaura, J.; Hatakeyama, K.; Watanuki, J.; Akasaka, T.; Kato, N.; Shimanuki, M.; Nishimura, K.; Takahashi, M.; Taniwaki, M.; et al. Emu/miR-125b transgenic mice develop lethal B-cell malignancies. Leukemia 2011, 25, 1849–1856. [Google Scholar] [CrossRef]
  66. Ferdowsi, S.; Atarodi, K.; Amirizadeh, N.; Toogeh, G.; Azarkeivan, A.; Shirkoohi, R.; Faranoush, M.; Vaezi, M.; Alimoghaddam, K.; Ghavamzadeh, A.; et al. Expression analysis of microRNA-125 in patients with polycythemia vera and essential thrombocythemia and correlation with JAK2 allele burden and laboratory findings. Int. J. Lab. Hematol. 2015, 37, 661–667. [Google Scholar] [CrossRef]
  67. Bruchova, H.; Merkerova, M.; Prchal, J.T. Aberrant expression of microRNA in polycythemia vera. Haematologica 2008, 93, 1009–1016. [Google Scholar] [CrossRef]
  68. Gebauer, N.; Bernard, V.; Gebauer, W.; Feller, A.C.; Merz, H. MicroRNA expression and JAK2 allele burden in bone marrow trephine biopsies of polycythemia vera, essential thrombocythemia and early primary myelofibrosis. Acta Haematol. 2013, 129, 251–256. [Google Scholar] [CrossRef]
  69. Kuang, X.; Wei, C.; Zhang, T.; Yang, Z.; Chi, J.; Wang, L. miR-378 inhibits cell growth and enhances apoptosis in human myelodysplastic syndromes. Int. J. Oncol. 2016, 49, 1921–1930. [Google Scholar] [CrossRef]
  70. Leone, E.; Morelli, E.; Di Martino, M.T.; Amodio, N.; Foresta, U.; Gulla, A.; Rossi, M.; Neri, A.; Giordano, A.; Munshi, N.C.; et al. Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growth. Clin. Cancer Res. 2013, 19, 2096–2106. [Google Scholar] [CrossRef]
  71. Di Martino, M.T.; Gulla, A.; Cantafio, M.E.; Lionetti, M.; Leone, E.; Amodio, N.; Guzzi, P.H.; Foresta, U.; Conforti, F.; Cannataro, M.; et al. In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget 2013, 4, 242–255. [Google Scholar] [CrossRef] [PubMed]
  72. Li, Z.; Wong, K.Y.; Chan, G.C.; Chng, W.J.; Chim, C.S. Epigenetic silencing of EVL/miR-342 in multiple myeloma. Transl. Res. 2018, 192, 46–53. [Google Scholar] [CrossRef]
  73. Long, X.; Li, J.; Wen, F.; Cao, Y.; Luo, Z.; Luo, C. miR-140-3p attenuated the tumorigenesis of multiple myeloma via attenuating BZW2. Hematology 2022, 27, 173–180. [Google Scholar] [CrossRef] [PubMed]
  74. Lin, C.; Chen, D.; Xiao, T.; Lin, D.; Lin, D.; Lin, L.; Zhu, H.; Xu, J.; Huang, W.; Yang, T. DNA methylation-mediated silencing of microRNA-204 enhances T cell acute lymphoblastic leukemia by up-regulating MMP-2 and MMP-9 via NF-kappaB. J. Cell. Mol. Med. 2021, 25, 2365–2376. [Google Scholar] [CrossRef] [PubMed]
  75. Kasar, S.; Underbayev, C.; Yuan, Y.; Hanlon, M.; Aly, S.; Khan, H.; Chang, V.; Batish, M.; Gavrilova, T.; Badiane, F.; et al. Therapeutic implications of activation of the host gene (Dleu2) promoter for miR-15a/16-1 in chronic lymphocytic leukemia. Oncogene 2014, 33, 3307–3315. [Google Scholar] [CrossRef] [PubMed]
  76. Davari, N.; Ahmadpour, F.; Kiani, A.A.; Azadpour, M.; Asadi, Z.T. Evaluation of microRNA-223 and microRNA-125a expression association with STAT3 and Bcl2 genes in blood leukocytes of CLL patients: A case-control study. BMC Res. Notes 2021, 14, 21. [Google Scholar] [CrossRef]
  77. Wong, K.Y.; Yu, L.; Chim, C.S. DNA methylation of tumor suppressor miRNA genes: A lesson from the miR-34 family. Epigenomics 2011, 3, 83–92. [Google Scholar] [CrossRef]
  78. Sandhu, S.K.; Volinia, S.; Costinean, S.; Galasso, M.; Neinast, R.; Santhanam, R.; Parthun, M.R.; Perrotti, D.; Marcucci, G.; Garzon, R.; et al. miR-155 targets histone deacetylase 4 (HDAC4) and impairs transcriptional activity of B-cell lymphoma 6 (BCL6) in the Emu-miR-155 transgenic mouse model. Proc. Natl. Acad. Sci. USA 2012, 109, 20047–20052. [Google Scholar] [CrossRef]
  79. Zhang, X.; Zhao, X.; Fiskus, W.; Lin, J.; Lwin, T.; Rao, R.; Zhang, Y.; Chan, J.C.; Fu, K.; Marquez, V.E.; et al. Coordinated silencing of MYC-mediated miR-29 by HDAC3 and EZH2 as a therapeutic target of histone modification in aggressive B-Cell lymphomas. Cancer Cell 2012, 22, 506–523. [Google Scholar] [CrossRef]
  80. Meyer, A.; Herkt, S.; Kunze-Schumacher, H.; Kohrs, N.; Ringleb, J.; Schneider, L.; Kuvardina, O.N.; Oellerich, T.; Haupl, B.; Krueger, A.; et al. The transcription factor TAL1 and miR-17-92 create a regulatory loop in hematopoiesis. Sci. Rep. 2020, 10, 21438. [Google Scholar] [CrossRef]
  81. Zhou, K.; Feng, X.; Wang, Y.; Liu, Y.; Tian, L.; Zuo, W.; Yi, S.; Wei, X.; Song, Y.; Qiu, L. miR-223 is repressed and correlates with inferior clinical features in mantle cell lymphoma through targeting SOX11. Exp. Hematol. 2018, 58, 27–34.e21. [Google Scholar] [CrossRef]
  82. Chen, P.; Price, C.; Li, Z.; Li, Y.; Cao, D.; Wiley, A.; He, C.; Gurbuxani, S.; Kunjamma, R.B.; Huang, H.; et al. miR-9 is an essential oncogenic microRNA specifically overexpressed in mixed lineage leukemia-rearranged leukemia. Proc. Natl. Acad. Sci. USA 2013, 110, 11511–11516. [Google Scholar] [CrossRef]
  83. Garzon, R.; Heaphy, C.E.; Havelange, V.; Fabbri, M.; Volinia, S.; Tsao, T.; Zanesi, N.; Kornblau, S.M.; Marcucci, G.; Calin, G.A.; et al. MicroRNA 29b functions in acute myeloid leukemia. Blood 2009, 114, 5331–5341. [Google Scholar] [CrossRef]
  84. Leotta, M.; Biamonte, L.; Raimondi, L.; Ronchetti, D.; Di Martino, M.T.; Botta, C.; Leone, E.; Pitari, M.R.; Neri, A.; Giordano, A.; et al. A p53-dependent tumor suppressor network is induced by selective miR-125a-5p inhibition in multiple myeloma cells. J. Cell. Physiol. 2014, 229, 2106–2116. [Google Scholar] [CrossRef]
  85. Rodriguez-Otero, P.; Roman-Gomez, J.; Vilas-Zornoza, A.; Jose-Eneriz, E.S.; Martin-Palanco, V.; Rifon, J.; Torres, A.; Calasanz, M.J.; Agirre, X.; Prosper, F. Deregulation of FGFR1 and CDK6 oncogenic pathways in acute lymphoblastic leukaemia harbouring epigenetic modifications of the MIR9 family. Br. J. Haematol. 2011, 155, 73–83. [Google Scholar] [CrossRef]
  86. Palacios, F.; Abreu, C.; Prieto, D.; Morande, P.; Ruiz, S.; Fernandez-Calero, T.; Naya, H.; Libisch, G.; Robello, C.; Landoni, A.I.; et al. Activation of the PI3K/AKT pathway by microRNA-22 results in CLL B-cell proliferation. Leukemia 2015, 29, 115–125. [Google Scholar] [CrossRef]
  87. Chim, C.S.; Wong, K.Y.; Qi, Y.; Loong, F.; Lam, W.L.; Wong, L.G.; Jin, D.Y.; Costello, J.F.; Liang, R. Epigenetic inactivation of the miR-34a in hematological malignancies. Carcinogenesis 2010, 31, 745–750. [Google Scholar] [CrossRef]
  88. Buhagiar, A.; Borg, J.; Ayers, D. Overview of current microRNA biomarker signatures as potential diagnostic tools for leukaemic conditions. Non-Coding RNA Res. 2020, 5, 22–26. [Google Scholar] [CrossRef]
  89. Moloudizargari, M.; Hekmatirad, S.; Mofarahe, Z.S.; Asghari, M.H. Exosomal microRNA panels as biomarkers for hematological malignancies. Curr. Probl. Cancer 2021, 45, 100726. [Google Scholar] [CrossRef]
  90. Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal miRNAs as Biomarkers for Prostate Cancer. Front. Genet. 2013, 4, 36. [Google Scholar] [CrossRef]
  91. Wang, H.; You, Y.; Zhu, X. The Role of Exosomes in the Progression and Therapeutic Resistance of Hematological Malignancies. Front. Oncol. 2022, 12, 887518. [Google Scholar] [CrossRef]
  92. Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
  93. Hornick, N.I.; Huan, J.; Doron, B.; Goloviznina, N.A.; Lapidus, J.; Chang, B.H.; Kurre, P. Serum Exosome MicroRNA as a Minimally-Invasive Early Biomarker of AML. Sci. Rep. 2015, 5, 11295. [Google Scholar] [CrossRef]
  94. Zhang, L.; Pan, L.; Xiang, B.; Zhu, H.; Wu, Y.; Chen, M.; Guan, P.; Zou, X.; Valencia, C.A.; Dong, B.; et al. Potential role of exosome-associated microRNA panels and in vivo environment to predict drug resistance for patients with multiple myeloma. Oncotarget 2016, 7, 30876–30891. [Google Scholar] [CrossRef]
  95. Manier, S.; Liu, C.J.; Avet-Loiseau, H.; Park, J.; Shi, J.; Campigotto, F.; Salem, K.Z.; Huynh, D.; Glavey, S.V.; Rivotto, B.; et al. Prognostic role of circulating exosomal miRNAs in multiple myeloma. Blood 2017, 129, 2429–2436. [Google Scholar] [CrossRef]
  96. Bouyssou, J.M.; Liu, C.J.; Bustoros, M.; Sklavenitis-Pistofidis, R.; Aljawai, Y.; Manier, S.; Yosef, A.; Sacco, A.; Kokubun, K.; Tsukamoto, S.; et al. Profiling of circulating exosomal miRNAs in patients with Waldenstrom Macroglobulinemia. PLoS ONE 2018, 13, e0204589. [Google Scholar] [CrossRef]
  97. Jiang, L.; Deng, T.; Wang, D.; Xiao, Y. Elevated Serum Exosomal miR-125b Level as a Potential Marker for Poor Prognosis in Intermediate-Risk Acute Myeloid Leukemia. Acta Haematol. 2018, 140, 183–192. [Google Scholar] [CrossRef]
  98. Feng, Y.; Zhong, M.; Zeng, S.; Wang, L.; Liu, P.; Xiao, X.; Liu, Y. Exosome-derived miRNAs as predictive biomarkers for diffuse large B-cell lymphoma chemotherapy resistance. Epigenomics 2019, 11, 35–51. [Google Scholar] [CrossRef]
  99. Shahid, S.; Shahid, W.; Shaheen, J.; Akhtar, M.W.; Sadaf, S. Circulating miR-146a expression as a non-invasive predictive biomarker for acute lymphoblastic leukemia. Sci. Rep. 2021, 11, 22783. [Google Scholar] [CrossRef]
  100. El-Khazragy, N.; Noshi, M.A.; Abdel-Malak, C.; Zahran, R.F.; Swellam, M. miRNA-155 and miRNA-181a as prognostic biomarkers for pediatric acute lymphoblastic leukemia. J. Cell. Biochem. 2019, 120, 6315–6321. [Google Scholar] [CrossRef]
  101. Liang, C.; Li, Y.; Wang, L.N.; Zhang, X.L.; Luo, J.S.; Peng, C.J.; Tang, W.Y.; Huang, L.B.; Tang, Y.L.; Luo, X.Q. Up-regulated miR-155 is associated with poor prognosis in childhood acute lymphoblastic leukemia and promotes cell proliferation targeting ZNF238. Hematology 2021, 26, 16–25. [Google Scholar] [CrossRef]
  102. Ghodousi, E.S.; Rahgozar, S. MicroRNA-326 and microRNA-200c: Two novel biomarkers for diagnosis and prognosis of pediatric acute lymphoblastic leukemia. J. Cell. Biochem. 2018, 119, 6024–6032. [Google Scholar] [CrossRef]
  103. Zamani, A.; Fattahi Dolatabadi, N.; Houshmand, M.; Nabavizadeh, N. miR-324-3p and miR-508-5p expression levels could serve as potential diagnostic and multidrug-resistant biomarkers in childhood acute lymphoblastic leukemia. Leuk. Res. 2021, 109, 106643. [Google Scholar] [CrossRef]
  104. Schotte, D.; De Menezes, R.X.; Akbari Moqadam, F.; Khankahdani, L.M.; Lange-Turenhout, E.; Chen, C.; Pieters, R.; Den Boer, M.L. MicroRNA characterize genetic diversity and drug resistance in pediatric acute lymphoblastic leukemia. Haematologica 2011, 96, 703–711. [Google Scholar] [CrossRef]
  105. Almeida, R.S.; Costa, E.S.M.; Coutinho, L.L.; Garcia Gomes, R.; Pedrosa, F.; Massaro, J.D.; Donadi, E.A.; Lucena-Silva, N. MicroRNA expression profiles discriminate childhood T- from B-acute lymphoblastic leukemia. Hematol. Oncol. 2019, 37, 103–112. [Google Scholar] [CrossRef]
  106. Labib, H.A.; Elantouny, N.G.; Ibrahim, N.F.; Alnagar, A.A. Upregulation of microRNA-21 is a poor prognostic marker in patients with childhood B cell acute lymphoblastic leukemia. Hematology 2017, 22, 392–397. [Google Scholar] [CrossRef]
  107. Amankwah, E.K.; Devidas, M.; Teachey, D.T.; Rabin, K.R.; Brown, P.A. Six Candidate miRNAs Associated with Early Relapse in Pediatric B-Cell Acute Lymphoblastic Leukemia. Anticancer Res. 2020, 40, 3147–3153. [Google Scholar] [CrossRef]
  108. Avigad, S.; Verly, I.R.; Lebel, A.; Kordi, O.; Shichrur, K.; Ohali, A.; Hameiri-Grossman, M.; Kaspers, G.J.; Cloos, J.; Fronkova, E.; et al. miR expression profiling at diagnosis predicts relapse in pediatric precursor B-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2016, 55, 328–339. [Google Scholar] [CrossRef]
  109. Mi, S.; Lu, J.; Sun, M.; Li, Z.; Zhang, H.; Neilly, M.B.; Wang, Y.; Qian, Z.; Jin, J.; Zhang, Y.; et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2007, 104, 19971–19976. [Google Scholar] [CrossRef]
  110. 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] [CrossRef]
  111. 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. Pharmacol. Sci. 2019, 23, 1203–1213. [Google Scholar]
  112. Koutova, L.; Sterbova, M.; Pazourkova, E.; Pospisilova, S.; Svobodova, I.; Horinek, A.; Lysak, D.; Korabecna, M. The impact of standard chemotherapy on miRNA signature in plasma in AML patients. Leuk. Res. 2015, 39, 1389–1395. [Google Scholar] [CrossRef]
  113. Wang, X.; Zuo, D.; Yuan, Y.; Yang, X.; Hong, Z.; Zhang, R. MicroRNA-183 promotes cell proliferation via regulating programmed cell death 6 in pediatric acute myeloid leukemia. J. Cancer Res. Clin. Oncol. 2017, 143, 169–180. [Google Scholar] [CrossRef]
  114. Qi, X.; Zhang, Y. MicroRNA-199a deficiency relates to higher bone marrow blasts, poor risk stratification and worse prognostication in pediatric acute myeloid leukemia patients. Pediatr. Hematol. Oncol. 2022, 39, 500–507. [Google Scholar] [CrossRef]
  115. Lin, X.; Wang, Z.; Wang, Y.; Feng, W. Serum MicroRNA-370 as a potential diagnostic and prognostic biomarker for pediatric acute myeloid leukemia. Int. J. Clin. Exp. Pathol. 2015, 8, 14658–14666. [Google Scholar]
  116. Zhu, R.; Lin, W.; Zhao, W.; Fan, F.; Tang, L.; Hu, Y. A 4-microRNA signature for survival prognosis in pediatric and adolescent acute myeloid leukemia. J. Cell. Biochem. 2019, 120, 3958–3968. [Google Scholar] [CrossRef]
  117. Rahimi, Z.; Ghorbani, Z.; Motamed, H.; Jalilian, N. Aberrant expression profile of miR-32, miR-98 and miR-374 in chronic lymphocytic leukemia. Leuk. Res. 2021, 111, 106691. [Google Scholar] [CrossRef]
  118. Bagheri, M.; Khansarinejad, B.; Mosayebi, G.; Moradabadi, A.; Mondanizadeh, M. Diagnostic Value of Plasma miR-145 and miR-185 as Targeting of the APRIL Oncogene in the B-cell Chronic Lymphocytic Leukemia. Asian Pac. J. Cancer Prev. 2021, 22, 111–117. [Google Scholar] [CrossRef]
  119. Klumper, T.; Bruckmueller, H.; Diewock, T.; Kaehler, M.; Haenisch, S.; Pott, C.; Bruhn, O.; Cascorbi, I. Expression differences of miR-142-5p between treatment-naive chronic myeloid leukemia patients responding and non-responding to imatinib therapy suggest a link to oncogenic ABL2, SRI, cKIT and MCL1 signaling pathways critical for development of therapy resistance. Exp. Hematol. Oncol. 2020, 9, 26. [Google Scholar]
  120. Habib, E.M.; Nosiar, N.A.; Eid, M.A.; Taha, A.M.; Sherief, D.E.; Hassan, A.E.; Abdel Ghafar, M.T. Circulating miR-146a expression predicts early treatment response to imatinib in adult chronic myeloid leukemia. J. Investig. Med. 2021, 69, 333–337. [Google Scholar] [CrossRef]
  121. Habib, E.M.; Nosiar, N.A.; Eid, M.A.; Taha, A.M.; Sherief, D.E.; Hassan, A.E.; Abdel Ghafar, M.T. MiR-150 Expression in Chronic Myeloid Leukemia: Relation to Imatinib Response. Lab. Med. 2022, 53, 58–64. [Google Scholar] [CrossRef] [PubMed]
  122. Machova Polakova, K.; Lopotova, T.; Klamova, H.; Burda, P.; Trneny, M.; Stopka, T.; Moravcova, J. Expression patterns of microRNAs associated with CML phases and their disease related targets. Mol. Cancer 2011, 10, 41. [Google Scholar] [CrossRef] [PubMed]
  123. Ninawe, A.; Guru, S.A.; Yadav, P.; Masroor, M.; Samadhiya, A.; Bhutani, N.; Gupta, N.; Gupta, R.; Saxena, A. miR-486-5p: A Prognostic Biomarker for Chronic Myeloid Leukemia. ACS Omega 2021, 6, 7711–7718. [Google Scholar] [PubMed]
  124. Keramati, F.; Jafarian, A.; Soltani, A.; Javandoost, E.; Mollaei, M.; Fallah, P. Circulating miRNAs can serve as potential diagnostic biomarkers in chronic myelogenous leukemia patients. Leuk. Res. Rep. 2021, 16, 100257. [Google Scholar] [CrossRef] [PubMed]
  125. Ali Beg, M.M.; Guru, S.A.; Abdullah, S.M.; Ahmad, I.; Rizvi, A.; Akhter, J.; Goyal, Y.; Verma, A.K. Regulation of miR-126 and miR-122 Expression and Response of Imatinib Treatment on Its Expression in Chronic Myeloid Leukemia Patients. Oncol. Res. Treat. 2021, 44, 530–537. [Google Scholar] [CrossRef]
  126. Troppan, K.; Wenzl, K.; Pichler, M.; Pursche, B.; Schwarzenbacher, D.; Feichtinger, J.; Thallinger, G.G.; Beham-Schmid, C.; Neumeister, P.; Deutsch, A. miR-199a and miR-497 Are Associated with Better Overall Survival due to Increased Chemosensitivity in Diffuse Large B-Cell Lymphoma Patients. Int. J. Mol. Sci. 2015, 16, 18077–18095. [Google Scholar] [CrossRef]
  127. Hao, M.; Zang, M.; Wendlandt, E.; Xu, Y.; An, G.; Gong, D.; Li, F.; Qi, F.; Zhang, Y.; Yang, Y.; et al. Low serum miR-19a expression as a novel poor prognostic indicator in multiple myeloma. Int. J. Cancer 2015, 136, 1835–1844. [Google Scholar] [CrossRef]
  128. Corthals, S.L.; Sun, S.M.; Kuiper, R.; de Knegt, Y.; Broyl, A.; van der Holt, B.; Beverloo, H.B.; Peeters, J.K.; el Jarari, L.; Lokhorst, H.M.; et al. MicroRNA signatures characterize multiple myeloma patients. Leukemia 2011, 25, 1784–1789. [Google Scholar] [CrossRef]
  129. Papanota, A.M.; Karousi, P.; Kontos, C.K.; Artemaki, P.I.; Liacos, C.I.; Papadimitriou, M.A.; Bagratuni, T.; Eleutherakis-Papaiakovou, E.; Malandrakis, P.; Ntanasis-Stathopoulos, I.; et al. A Cancer-Related microRNA Signature Shows Biomarker Utility in Multiple Myeloma. Int. J. Mol. Sci. 2021, 22, 13144. [Google Scholar] [CrossRef]
  130. Yang, N.; Chen, J.; Zhang, H.; Wang, X.; Yao, H.; Peng, Y.; Zhang, W. LncRNA OIP5-AS1 loss-induced microRNA-410 accumulation regulates cell proliferation and apoptosis by targeting KLF10 via activating PTEN/PI3K/AKT pathway in multiple myeloma. Cell Death Dis. 2017, 8, e2975. [Google Scholar] [CrossRef]
  131. Li, F.; Xu, Y.; Deng, S.; Li, Z.; Zou, D.; Yi, S.; Sui, W.; Hao, M.; Qiu, L. MicroRNA-15a/16-1 cluster located at chromosome 13q14 is down-regulated but displays different expression pattern and prognostic significance in multiple myeloma. Oncotarget 2015, 6, 38270–38282. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, P.; Agnelli, L.; Walker, B.A.; Todoerti, K.; Lionetti, M.; Johnson, D.C.; Kaiser, M.; Mirabella, F.; Wardell, C.; Gregory, W.M.; et al. Improved risk stratification in myeloma using a microRNA-based classifier. Br. J. Haematol. 2013, 162, 348–359. [Google Scholar] [CrossRef] [PubMed]
  133. Ren, Y.; Li, X.; Wang, W.; He, W.; Wang, J.; Wang, Y. Expression of Peripheral Blood miRNA-720 and miRNA-1246 Can Be Used as a Predictor for Outcome in Multiple Myeloma Patients. Clin. Lymphoma Myeloma Leuk. 2017, 17, 415–423. [Google Scholar] [CrossRef] [PubMed]
  134. Gao, X.; Zhang, R.; Qu, X.; Zhao, M.; Zhang, S.; Wu, H.; Jianyong, L.; Chen, L. MiR-15a, miR-16-1 and miR-17-92 cluster expression are linked to poor prognosis in multiple myeloma. Leuk. Res. 2012, 36, 1505–1509. [Google Scholar] [CrossRef]
  135. Chi, J.; Ballabio, E.; Chen, X.H.; Kusec, R.; Taylor, S.; Hay, D.; Tramonti, D.; Saunders, N.J.; Littlewood, T.; Pezzella, F.; et al. MicroRNA expression in multiple myeloma is associated with genetic subtype, isotype and survival. Biol. Direct 2011, 6, 23. [Google Scholar] [CrossRef]
  136. Xiang, Y.; Zhang, L.; Xiang, P.; Zhang, J. Circulating miRNAs as Auxiliary Diagnostic Biomarkers for Multiple Myeloma: A Systematic Review, Meta-Analysis, and Recommendations. Front. Oncol. 2021, 11, 698197. [Google Scholar] [CrossRef]
  137. Xu, P.; Xia, T.; Ling, Y.; Chen, B. MiRNAs with prognostic significance in multiple myeloma: A systemic review and meta-analysis. Medicine 2019, 98, e16711. [Google Scholar] [CrossRef]
  138. Balassa, K.; Danby, R.; Rocha, V. Haematopoietic stem cell transplants: Principles and indications. Br. J. Hosp. Med. 2019, 80, 33–39. [Google Scholar] [CrossRef]
  139. Bazinet, A.; Popradi, G. A general practitioner’s guide to hematopoietic stem-cell transplantation. Curr. Oncol. 2019, 26, 187–191. [Google Scholar] [CrossRef]
  140. Khaddour, K.; Hana, C.K.; Mewawalla, P. Hematopoietic Stem Cell Transplantation. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  141. Wingard, J.R.; Hsu, J.; Hiemenz, J.W. Hematopoietic stem cell transplantation: An overview of infection risks and epidemiology. Infect. Dis. Clin. N. Am. 2010, 24, 257–272. [Google Scholar] [CrossRef]
  142. Hosseini, E.; Schwarer, A.P.; Ghasemzadeh, M. Do human leukocyte antigen E polymorphisms influence graft-versus-leukemia after allogeneic hematopoietic stem cell transplantation? Exp. Hematol. 2015, 43, 149–157. [Google Scholar] [CrossRef] [PubMed]
  143. Passweg, J.R.; Baldomero, H.; Bader, P.; Bonini, C.; Cesaro, S.; Dreger, P.; Duarte, R.F.; Dufour, C.; Kuball, J.; Farge-Bancel, D.; et al. Hematopoietic stem cell transplantation in Europe 2014: More than 40,000 transplants annually. Bone Marrow Transplant. 2016, 51, 786–792. [Google Scholar] [CrossRef] [PubMed]
  144. Wagner, J.E.; Barker, J.N.; DeFor, T.E.; Baker, K.S.; Blazar, B.R.; Eide, C.; Goldman, A.; Kersey, J.; Krivit, W.; MacMillan, M.L.; et al. Transplantation of unrelated donor umbilical cord blood in 102 patients with malignant and nonmalignant diseases: Influence of CD34 cell dose and HLA disparity on treatment-related mortality and survival. Blood 2002, 100, 1611–1618. [Google Scholar] [CrossRef] [PubMed]
  145. Copelan, E.A. Hematopoietic stem-cell transplantation. N. Engl. J. Med. 2006, 354, 1813–1826. [Google Scholar] [CrossRef] [PubMed]
  146. Ferrara, J.L.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef]
  147. Toubai, T.; Sun, Y.; Reddy, P. GVHD pathophysiology: Is acute different from chronic? Best Pract. Res. Clin. Haematol. 2008, 21, 101–117. [Google Scholar] [CrossRef] [PubMed]
  148. Ball, L.M.; Egeler, R.M.; EBMT Paediatric Working Party. Acute GvHD: Pathogenesis and classification. Bone Marrow Transplant. 2008, 41 (Suppl. S2), S58–S64. [Google Scholar] [CrossRef]
  149. Jagasia, M.H.; Greinix, H.T.; Arora, M.; Williams, K.M.; Wolff, D.; Cowen, E.W.; Palmer, J.; Weisdorf, D.; Treister, N.S.; Cheng, G.S.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol. Blood Marrow Transplant. 2015, 21, 389–401.e1. [Google Scholar] [CrossRef]
  150. Zecca, M.; Prete, A.; Rondelli, R.; Lanino, E.; Balduzzi, A.; Messina, C.; Fagioli, F.; Porta, F.; Favre, C.; Pession, A.; et al. Chronic graft-versus-host disease in children: Incidence, risk factors, and impact on outcome. Blood 2002, 100, 1192–1200. [Google Scholar] [CrossRef]
  151. Lee, S.J.; Vogelsang, G.; Flowers, M.E. Chronic graft-versus-host disease. Biol. Blood Marrow Transplant. 2003, 9, 215–233. [Google Scholar] [CrossRef]
  152. Fall-Dickson, J.M.; Pavletic, S.Z.; Mays, J.W.; Schubert, M.M. Oral Complications of Chronic Graft-Versus-Host Disease. J. Natl. Cancer Inst. Monogr. 2019, 2019, lgz007. [Google Scholar] [CrossRef] [PubMed]
  153. Holler, E. Risk assessment in haematopoietic stem cell transplantation: GvHD prevention and treatment. Best Pract. Res. Clin. Haematol. 2007, 20, 281–294. [Google Scholar] [CrossRef] [PubMed]
  154. Wolff, D.; Radojcic, V.; Lafyatis, R.; Cinar, R.; Rosenstein, R.K.; Cowen, E.W.; Cheng, G.S.; Sheshadri, A.; Bergeron, A.; Williams, K.M.; et al. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: IV. The 2020 Highly morbid forms report. Transplant. Cell. Ther. 2021, 27, 817–835. [Google Scholar] [CrossRef] [PubMed]
  155. Aladag, E.; Kelkitli, E.; Goker, H. Acute Graft-Versus-Host Disease: A Brief Review. Turk. J. Haematol. 2020, 37, 1–4. [Google Scholar] [CrossRef]
  156. Ali, A.M.; DiPersio, J.F.; Schroeder, M.A. The Role of Biomarkers in the Diagnosis and Risk Stratification of Acute Graft-versus-Host Disease: A Systematic Review. Biol. Blood Marrow Transplant. 2016, 22, 1552–1564. [Google Scholar] [CrossRef]
  157. Newmarch, M.; Kostantin, E.; Tsongalis, G.J.; Guimond, M.; Roy, J.; De Guire, V.; Ahmad, I. MicroRNAs in graft-versus-host disease: A review of the latest data. Bone Marrow Transplant. 2020, 55, 1014–1020. [Google Scholar] [CrossRef]
  158. Vajari, M.K.; Moradinasab, S.; Yousefi, A.M.; Bashash, D. Noncoding RNAs in diagnosis and prognosis of graft-versus-host disease (GVHD). J. Cell. Physiol. 2022, 237, 3480–3495. [Google Scholar] [CrossRef]
  159. Xie, L.N.; Zhou, F.; Liu, X.M.; Fang, Y.; Yu, Z.; Song, N.X.; Kong, F.S. Serum microRNA155 is increased in patients with acute graft-versus-host disease. Clin. Transplant. 2014, 28, 314–323. [Google Scholar] [CrossRef]
  160. Zhang, R.; Wang, X.; Hong, M.; Luo, T.; Zhao, M.; Shen, H.; Fang, J.; Li, X.; Zang, S.; Chen, P.; et al. Endothelial microparticles delivering microRNA-155 into T lymphocytes are involved in the initiation of acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Oncotarget 2017, 8, 23360–23375. [Google Scholar] [CrossRef]
  161. Stickel, N.; Prinz, G.; Pfeifer, D.; Hasselblatt, P.; Schmitt-Graeff, A.; Follo, M.; Thimme, R.; Finke, J.; Duyster, J.; Salzer, U.; et al. MiR-146a regulates the TRAF6/TNF-axis in donor T cells during GVHD. Blood 2014, 124, 2586–2595. [Google Scholar] [CrossRef]
  162. Stickel, N.; Hanke, K.; Marschner, D.; Prinz, G.; Kohler, M.; Melchinger, W.; Pfeifer, D.; Schmitt-Graeff, A.; Brummer, T.; Heine, A.; et al. MicroRNA-146a reduces MHC-II expression via targeting JAK/STAT signaling in dendritic cells after stem cell transplantation. Leukemia 2017, 31, 2732–2741. [Google Scholar] [CrossRef] [PubMed]
  163. Atarod, S.; Ahmed, M.M.; Lendrem, C.; Pearce, K.F.; Cope, W.; Norden, J.; Wang, X.N.; Collin, M.; Dickinson, A.M. miR-146a and miR-155 Expression Levels in Acute Graft-Versus-Host Disease Incidence. Front. Immunol. 2016, 7, 56. [Google Scholar] [CrossRef] [PubMed]
  164. Crossland, R.E.; Norden, J.; Ghimire, S.; Juric, M.K.; Pearce, K.F.; Lendrem, C.; Collin, M.; Mischak-Weissinger, E.; Holler, E.; Greinix, H.T.; et al. Profiling Tissue and Biofluid miR-155-5p, miR-155*, and miR-146a-5p Expression in Graft vs. Host Disease. Front. Immunol. 2021, 12, 639171. [Google Scholar] [CrossRef] [PubMed]
  165. Xiao, B.; Wang, Y.; Li, W.; Baker, M.; Guo, J.; Corbet, K.; Tsalik, E.L.; Li, Q.J.; Palmer, S.M.; Woods, C.W.; et al. Plasma microRNA signature as a noninvasive biomarker for acute graft-versus-host disease. Blood 2013, 122, 3365–3375. [Google Scholar] [CrossRef] [PubMed]
  166. Crossland, R.E.; Norden, J.; Kralj Juric, M.; Pearce, K.F.; Lendrem, C.; Bibby, L.A.; Collin, M.; Greinix, H.T.; Dickinson, A.M. Serum and Extracellular Vesicle MicroRNAs miR-423, miR-199, and miR-93* As Biomarkers for Acute Graft-versus-Host Disease. Front. Immunol. 2017, 8, 1446. [Google Scholar] [CrossRef]
  167. Crossland, R.E.; Norden, J.; Juric, M.K.; Green, K.; Pearce, K.F.; Lendrem, C.; Greinix, H.T.; Dickinson, A.M. Expression of Serum microRNAs is Altered During Acute Graft-versus-Host Disease. Front. Immunol. 2017, 8, 308. [Google Scholar] [CrossRef]
  168. Xie, L.; Zhou, F.; Liu, X.; Fang, Y.; Yu, Z.; Song, N.; Kong, F. Serum microRNA181a: Correlates with the intracellular cytokine levels and a potential biomarker for acute graft-versus-host disease. Cytokine 2016, 85, 37–44. [Google Scholar] [CrossRef]
  169. Gimondi, S.; Dugo, M.; Vendramin, A.; Bermema, A.; Biancon, G.; Cavane, A.; Corradini, P.; Carniti, C. Circulating miRNA panel for prediction of acute graft-versus-host disease in lymphoma patients undergoing matched unrelated hematopoietic stem cell transplantation. Exp. Hematol. 2016, 44, 624–634.e1. [Google Scholar] [CrossRef]
  170. Ranganathan, P.; Ngankeu, A.; Zitzer, N.C.; Leoncini, P.; Yu, X.; Casadei, L.; Challagundla, K.; Reichenbach, D.K.; Garman, S.; Ruppert, A.S.; et al. Serum miR-29a Is Upregulated in Acute Graft-versus-Host Disease and Activates Dendritic Cells through TLR Binding. J. Immunol. 2017, 198, 2500–2512. [Google Scholar] [CrossRef]
  171. Wang, Y.; Zhao, X.; Ye, X.; Luo, H.; Zhao, T.; Diao, Y.; Zhang, H.; Lv, M.; Zhang, W.; Huang, X.; et al. Plasma microRNA-586 is a new biomarker for acute graft-versus-host disease. Ann. Hematol. 2015, 94, 1505–1514. [Google Scholar] [CrossRef]
  172. Zhao, X.S.; Wang, Y.N.; Lv, M.; Kong, Y.; Luo, H.X.; Ye, X.Y.; Wu, Q.; Zhao, T.F.; Hu, Y.H.; Zhang, H.Y.; et al. miR-153-3p, a new bio-target, is involved in the pathogenesis of acute graft-versus-host disease via inhibition of indoleamine- 2,3-dioxygenase. Oncotarget 2016, 7, 48321–48334. [Google Scholar] [CrossRef] [PubMed]
  173. Motaei, J.; Kerachian, M.A.; Mousavi, S.A.; Alimoghadam, K.; Ghavamzadeh, A.; Manoochehrabadi, S.; Ahmadvand, M.; Yaghmaie, M. Circulating miR-455-3p, miR-5787, and miR-548a-3p as potential noninvasive biomarkers in the diagnosis of acute graft-versus-host disease: A validation study. Ann. Hematol. 2021, 100, 2621–2631. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, C.; Bai, N.; Huang, W.; Zhang, P.; Luo, Y.; Men, S.; Wen, T.; Tong, H.; Wang, S.; Tian, Y.P. The predictive value of selected serum microRNAs for acute GVHD by TaqMan MicroRNA arrays. Ann. Hematol. 2016, 95, 1833–1843. [Google Scholar] [CrossRef] [PubMed]
  175. Yoshizawa, S.; Umezu, T.; Saitoh, Y.; Gotoh, M.; Akahane, D.; Kobayashi, C.; Ohyashiki, J.H.; Ohyashiki, K. Exosomal miRNA Signatures for Late-Onset Acute Graft-Versus-Host Disease in Allogenic Hematopoietic Stem Cell Transplantation. Int. J. Mol. Sci. 2018, 19, 2493. [Google Scholar] [CrossRef] [PubMed]
  176. Atarod, S.; Norden, J.; Bibby, L.A.; Janin, A.; Ratajczak, P.; Lendrem, C.; Pearce, K.F.; Wang, X.N.; O’Reilly, S.; Van Laar, J.M.; et al. Differential MicroRNA Expression Levels in Cutaneous Acute Graft-Versus-Host Disease. Front. Immunol. 2018, 9, 1485. [Google Scholar] [CrossRef]
  177. Juric, M.K.; Shevtsov, M.; Mozes, P.; Ogonek, J.; Crossland, R.E.; Dickinson, A.M.; Greinix, H.T.; Holler, E.; Weissinger, E.M.; Multhoff, G. B-Cell-Based and Soluble Biomarkers in Body Liquids for Predicting Acute/Chronic Graft-versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Front. Immunol. 2016, 7, 660. [Google Scholar] [CrossRef]
  178. Reikvam, H.; Vo, A.K.; Johansen, S.; Hemsing, A.L.; Solheim, M.H.; Mosevoll, K.A.; Tvedt, T.H.A.; Hatfield, K.J. MicroRNA serum profiles and chronic graft-versus-host disease. Blood Adv. 2022, 6, 5295–5306. [Google Scholar] [CrossRef]
  179. Lacina, P.; Crossland, R.E.; Wielinska, J.; Czyz, A.; Szeremet, A.; Ussowicz, M.; Wrobel, T.; Dickinson, A.M.; Bogunia-Kubik, K. Differential expression of miRNAs from extracellular vesicles in chronic graft-versus-host disease: A preliminary study. Adv. Clin. Exp. Med. 2022. [Google Scholar] [CrossRef]
  180. Sorror, M.L.; Gooley, T.A.; Maclean, K.H.; Hubbard, J.; Marcondes, M.A.; Torok-Storb, B.J.; Tewari, M. Pre-transplant expressions of microRNAs, comorbidities, and post-transplant mortality. Bone Marrow Transplant. 2019, 54, 973–979. [Google Scholar] [CrossRef]
  181. Cheng, Z.; Zhou, L.; Hu, K.; Dai, Y.; Pang, Y.; Zhao, H.; Wu, S.; Qin, T.; Han, Y.; Hu, N.; et al. Prognostic significance of microRNA-99a in acute myeloid leukemia patients undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2018, 53, 1089–1095. [Google Scholar] [CrossRef]
  182. Yang, C.; Shao, T.; Zhang, H.; Zhang, N.; Shi, X.; Liu, X.; Yao, Y.; Xu, L.; Zhu, S.; Cao, J.; et al. MiR-425 expression profiling in acute myeloid leukemia might guide the treatment choice between allogeneic transplantation and chemotherapy. J. Transl. Med. 2018, 16, 267. [Google Scholar] [CrossRef] [PubMed]
  183. Lim, H.C.; Soneji, S.; Palmason, R.; Lenhoff, S.; Laurell, T.; Scheding, S. Development of acoustically isolated extracellular plasma vesicles for biomarker discovery in allogeneic hematopoietic stem cell transplantation. Biomark. Res. 2021, 9, 6. [Google Scholar] [CrossRef] [PubMed]
  184. Pontoppidan, P.L.; Jordan, K.; Carlsen, A.L.; Uhlving, H.H.; Kielsen, K.; Christensen, M.; Ifversen, M.; Nielsen, C.H.; Sangild, P.; Heegaard, N.H.; et al. Associations between gastrointestinal toxicity, micro RNA and cytokine production in patients undergoing myeloablative allogeneic stem cell transplantation. Int. Immunopharmacol. 2015, 25, 180–188. [Google Scholar] [CrossRef] [PubMed]
  185. Nowicki, M.; Szemraj, J.; Wierzbowska, A.; Misiewicz, M.; Malachowski, R.; Pluta, A.; Grzybowska-Izydorczyk, O.; Robak, T.; Szmigielska-Kaplon, A. miRNA-15a, miRNA-16, miRNA-126, miRNA-146a, and miRNA-223 expressions in autologous hematopoietic stem cell transplantation and their impact on engraftment. Eur. J. Haematol. 2018, 100, 426–435. [Google Scholar] [CrossRef] [PubMed]
  186. Rafiee, M.; Farsani, M.A.; Amiri, V.; Hajifathali, A.; Gharehbaghian, A.; Mohammadi, M.H. Circulatory miR-155 correlation with platelet and neutrophil recovery after autologous hematopoietic stem cell transplantation, a multivariate analysis. Int. J. Hematol. 2021, 114, 235–245. [Google Scholar] [CrossRef] [PubMed]
  187. Park, S.S.; Lim, J.Y.; Kim, T.W.; Ko, Y.H.; Jeon, W.J.; Lee, S.Y.; Lee, J.H.; Min, C.K. Predictive impact of circulating microRNA-193a-5p on early relapse after autologous stem cell transplantation in patients with multiple myeloma. Br. J. Haematol. 2020, 189, 518–523. [Google Scholar] [CrossRef]
  188. Innao, V.; Allegra, A.; Pulvirenti, N.; Allegra, A.G.; Musolino, C. Therapeutic potential of antagomiRs in haematological and oncological neoplasms. Eur. J. Cancer Care 2020, 29, e13208. [Google Scholar] [CrossRef]
  189. Dorrance, A.M.; Neviani, P.; Ferenchak, G.J.; Huang, X.; Nicolet, D.; Maharry, K.S.; Ozer, H.G.; Hoellarbauer, P.; Khalife, J.; Hill, E.B.; et al. Targeting leukemia stem cells in vivo with antagomiR-126 nanoparticles in acute myeloid leukemia. Leukemia 2015, 29, 2143–2153. [Google Scholar] [CrossRef]
  190. Wang, W.Z.; Lin, X.H.; Pu, Q.H.; Liu, M.Y.; Li, L.; Wu, L.R.; Wu, Q.Q.; Mao, J.W.; Zhu, J.Y.; Jin, X.B. Targeting miR-21 sensitizes Ph+ ALL Sup-b15 cells to imatinib-induced apoptosis through upregulation of PTEN. Biochem. Biophys. Res. Commun. 2014, 454, 423–428. [Google Scholar] [CrossRef]
  191. Zhang, L.; Nguyen, L.X.T.; Chen, Y.C.; Wu, D.; Cook, G.J.; Hoang, D.H.; Brewer, C.J.; He, X.; Dong, H.; Li, S.; et al. Targeting miR-126 in inv(16) acute myeloid leukemia inhibits leukemia development and leukemia stem cell maintenance. Nat. Commun. 2021, 12, 6154. [Google Scholar] [CrossRef]
  192. Velu, C.S.; Chaubey, A.; Phelan, J.D.; Horman, S.R.; Wunderlich, M.; Guzman, M.L.; Jegga, A.G.; Zeleznik-Le, N.J.; Chen, J.; Mulloy, J.C.; et al. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J. Clin. Investig. 2014, 124, 222–236. [Google Scholar] [CrossRef] [PubMed]
  193. Anastasiadou, E.; Seto, A.G.; Beatty, X.; Hermreck, M.; Gilles, M.E.; Stroopinsky, D.; Pinter-Brown, L.C.; Pestano, L.; Marchese, C.; Avigan, D.; et al. Cobomarsen, an Oligonucleotide Inhibitor of miR-155, Slows DLBCL Tumor Cell Growth In Vitro and In Vivo. Clin. Cancer Res. 2021, 27, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  194. Deng, F.; Zhang, C.; Lu, T.; Liao, E.J.; Huang, H.; Wei, S. Roles of circRNAs in hematological malignancies. Biomark. Res. 2022, 10, 50. [Google Scholar] [CrossRef]
  195. Ashwal-Fluss, R.; Meyer, M.; Pamudurti, N.R.; Ivanov, A.; Bartok, O.; Hanan, M.; Evantal, N.; Memczak, S.; Rajewsky, N.; Kadener, S. circRNA biogenesis competes with pre-mRNA splicing. Mol. Cell 2014, 56, 55–66. [Google Scholar] [CrossRef] [PubMed]
  196. Barta, T.; Peskova, L.; Hampl, A. miRNAsong: A web-based tool for generation and testing of miRNA sponge constructs in silico. Sci. Rep. 2016, 6, 36625. [Google Scholar] [CrossRef]
  197. Han, F.; Zhong, C.; Li, W.; Wang, R.; Zhang, C.; Yang, X.; Ji, C.; Ma, D. hsa_circ_0001947 suppresses acute myeloid leukemia progression via targeting hsa-miR-329-5p/CREBRF axis. Epigenomics 2020, 12, 935–953. [Google Scholar] [CrossRef]
  198. Wu, Z.J.; Sun, Q.; Gu, D.L.; Wang, L.Q.; Li, J.Y.; Jin, H. [Expression of circ-KEL in acute myeloid leukemia and its regulatory mechanisms in leukemic cells]. Zhonghua Xue Ye Xue Za Zhi 2021, 42, 230–237. [Google Scholar]
  199. Cao, H.X.; Miao, C.F.; Sang, L.N.; Huang, Y.M.; Zhang, R.; Sun, L.; Jiang, Z.X. Circ_0009910 promotes imatinib resistance through ULK1-induced autophagy by sponging miR-34a-5p in chronic myeloid leukemia. Life Sci. 2020, 243, 117255. [Google Scholar] [CrossRef]
  200. Wang, D.; Ming, X.; Xu, J.; Xiao, Y. Circ_0009910 shuttled by exosomes regulates proliferation, cell cycle and apoptosis of acute myeloid leukemia cells by regulating miR-5195-3p/GRB10 axis. Hematol. Oncol. 2021, 39, 390–400. [Google Scholar] [CrossRef]
  201. Chen, F.; Wang, X.; Fu, S.; Wang, S.; Fu, Y.; Liu, Z.; Zhang, J. Effect of the Up-Regulation of Circular RNA Hsa_circ_0069767 Derived from C-KIT on the Biological Behavior of Multiple Myeloma Cells. Cancer Manag. Res. 2020, 12, 11321–11331. [Google Scholar] [CrossRef]
  202. Xu, L.; Xu, Y.; Jing, Z.; Wang, X.; Zha, X.; Zeng, C.; Chen, S.; Yang, L.; Luo, G.; Li, B.; et al. Altered expression pattern of miR-29a, miR-29b and the target genes in myeloid leukemia. Exp. Hematol. Oncol. 2014, 3, 17. [Google Scholar] [CrossRef] [PubMed]
  203. Li, Y.; Mao, M.; Liu, H.; Wang, X.; Kou, Z.; Nie, Y.; Wang, Y.; Wang, Z.; Huang, Q.; Lang, T.; et al. miR-34a and miR-29b as indicators for prognosis of treatment-free survival of chronic lymphocytic leukemia patients in Chinese Uygur and Han populations. Mol. Cell. Probes 2019, 47, 101436. [Google Scholar] [CrossRef] [PubMed]
  204. Garzon, R.; Liu, S.; Fabbri, M.; Liu, Z.; Heaphy, C.E.; Callegari, E.; Schwind, S.; Pang, J.; Yu, J.; Muthusamy, N.; et al. MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood 2009, 113, 6411–6418. [Google Scholar] [CrossRef] [PubMed]
  205. Blum, W.; Klisovic, R.B.; Hackanson, B.; Liu, Z.; Liu, S.; Devine, H.; Vukosavljevic, T.; Huynh, L.; Lozanski, G.; Kefauver, C.; et al. Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia. J. Clin. Oncol. 2007, 25, 3884–3891. [Google Scholar] [CrossRef] [PubMed]
  206. Zhao, L.; Li, Y.; Song, X.; Zhou, H.; Li, N.; Miao, Y.; Jia, L. Upregulation of miR-181c inhibits chemoresistance by targeting ST8SIA4 in chronic myelocytic leukemia. Oncotarget 2016, 7, 60074–60086. [Google Scholar] [CrossRef] [PubMed]
  207. Huang, X.; Schwind, S.; Yu, B.; Santhanam, R.; Wang, H.; Hoellerbauer, P.; Mims, A.; Klisovic, R.; Walker, A.R.; Chan, K.K.; et al. Targeted delivery of microRNA-29b by transferrin-conjugated anionic lipopolyplex nanoparticles: A novel therapeutic strategy in acute myeloid leukemia. Clin. Cancer Res. 2013, 19, 2355–2367. [Google Scholar] [CrossRef]
  208. Huang, X.; Schwind, S.; Santhanam, R.; Eisfeld, A.K.; Chiang, C.L.; Lankenau, M.; Yu, B.; Hoellerbauer, P.; Jin, Y.; Tarighat, S.S.; et al. Targeting the RAS/MAPK pathway with miR-181a in acute myeloid leukemia. Oncotarget 2016, 7, 59273–59286. [Google Scholar] [CrossRef]
  209. Marcucci, G.; Radmacher, M.D.; Maharry, K.; Mrozek, K.; Ruppert, A.S.; Paschka, P.; Vukosavljevic, T.; Whitman, S.P.; Baldus, C.D.; Langer, C.; et al. MicroRNA expression in cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 2008, 358, 1919–1928. [Google Scholar] [CrossRef]
Figure 1. MiRNAs as potential diagnostic and prognostic biomarkers for aGvHD in patients receiving HSCT. The onset of aGvHD limits the successful therapy of hematologic malignancies. Conditioning regimens damage the host tissue with the induction of dangerous signals, including damage-associated molecular patterns (DAMPs). DAMPs lead to the activation of antigen-presenting cells (APCs), which subsequently stimulate the proliferation of donor T cells into effector T cells. When entering the target organs, effector T cells cause aGvHD development. Aberrant miRNA expression is considered as being involved in the pathology and onset of aGvHD. Selected miRNAs might be used as noninvasive diagnostic biomarkers due to their differential expression in plasma samples of patients with aGvHD compared to patients without GvHD. The panels on the right side of the figure represent upregulated (green arrows) and downregulated (red arrow) miRNAs in HSCT patients who developed aGvHD. Abbreviation: aGvHD, acute graft-versus-host disease; APCs, antigen-presenting cells; DAMPs, damage-associated molecular patterns; HSCT, hematopoietic stem cell transplantation.
Figure 1. MiRNAs as potential diagnostic and prognostic biomarkers for aGvHD in patients receiving HSCT. The onset of aGvHD limits the successful therapy of hematologic malignancies. Conditioning regimens damage the host tissue with the induction of dangerous signals, including damage-associated molecular patterns (DAMPs). DAMPs lead to the activation of antigen-presenting cells (APCs), which subsequently stimulate the proliferation of donor T cells into effector T cells. When entering the target organs, effector T cells cause aGvHD development. Aberrant miRNA expression is considered as being involved in the pathology and onset of aGvHD. Selected miRNAs might be used as noninvasive diagnostic biomarkers due to their differential expression in plasma samples of patients with aGvHD compared to patients without GvHD. The panels on the right side of the figure represent upregulated (green arrows) and downregulated (red arrow) miRNAs in HSCT patients who developed aGvHD. Abbreviation: aGvHD, acute graft-versus-host disease; APCs, antigen-presenting cells; DAMPs, damage-associated molecular patterns; HSCT, hematopoietic stem cell transplantation.
Cancers 15 02658 g001
Table 1. MicroRNA expression associated with malignant hematopoiesis.
Table 1. MicroRNA expression associated with malignant hematopoiesis.
MalignancymiRNA miRNA Expression
Level
Target Genes Study
Type
miRNA Function/Clinical ConsequenceReference
AMLmiR-3662DOWNIKBKBAM,
PS (N = 20),
TC
Acceleration of the growth and colony formation
of HP cells, survival
of leukemic cells
[58]
miR-9UPRHOH
RYBP + 15 other potential targets
AM
CL
PS (N = 85)
Increasing cell survival and decreasing apoptosis[82]
miR-29bDOWNMCL-1, CXXC6, CDK6AM,
CL,
PS (N = 100)
Cell growth and anti-apoptotic activity[83]
miR-126UPSLC9A7, ABCG1, MEF2C, RBMPS, LYZ, CSTA, HALAM,
CL,
PS (N = 6)
Cell growth and anti-apoptotic activity[57]
miR-301bUPFOXF2CLCell proliferation and decreasing apoptosis[59]
CMLmiR-19b
(miR-17-92 cluster)
UPA20AM,
BM,
CL
Cell proliferation, cell cycle, and decreasing apoptosis[60]
miR-328DOWNPIM-1,
TCF-4
CLCell proliferation, survival [61]
MDSmiR-378DOWNCDC40CL,
PS (BM) (N = 20)
Inducing the apoptosis and blocking the cell cycle of MDS cells[69]
MMmiR-21UPPTEN, Rho-B, BTG2AM,
CL
Growth and anti-apoptotic activity[70]
miR-221/222UPp27Kip1, PUMA, PTEN, p57Kip2AM,
CL
Proliferation and cell survival[71]
miR-342-3pDOWNFOXQ1
RAP2B
CDC42
CL,
PS diagnostic and relapsed MM (N = 93)
Methylation-derived silencing of miR-342-3p
might be an early event in MM pathogenesis
[72]
miR-140-3pDOWNBZW2AM,
CL
Cell proliferation, decreasing the apoptosis in
MM cells
[73]
miR-125a-5pUPTP53CLCell proliferation, cell growth
decreasing apoptosis
of cancer cells
[84]
miR-9DOWNFGFR1
CDK6
CL
PS (N = 200)
MiR-9 hypermethylation lead to the activation of oncogenic pathways and represents a prognostic factor for survival[85]
ALLmiR-124aDOWNCDK6AM,
CL,
PS (N = 353)
Cell proliferation and growth of ALL cells[56]
T-ALLmiR-204DOWNIRAK1,
NF-kB
AM,
CL,
PS (N = 32)
T-ALL growth and metastasis by increased IRAK1 and activation of NF-kB signaling pathway and targets[74]
CLLmiR-125a,
miR-223
DOWNBCL2,
STAT3
PS (N = 30)Control of white blood cell production[76]
miR-22UPPTENPS (N = 22)B-CLL cell proliferation[86]
miR-15a,
miR-16-1
DOWNDLEU2AM,
PBMC
Decreasing apoptosis and cell cycle of malignant B-cells[75]
NHLmiR-34aDOWNTP53,
CDK6
CL,
PS (N = 32)
MiR-34a is preferentially hypermethylated in NHL, the role of miR-34a in lymphomagenesis [87]
miR-29DOWNIGF-1R, CDK6CLCell survival and growth regulation in MCLs[79]
DLBCLmiR-155UPHDAC4AM,
CL
Block the development of B cells at the immature stage and induce cell proliferation [78]
Abbreviations: ALL, Acute lymphocytic leukemia; AM, animal model; AML, acute myeloid leukemia; BM, bone marrow samples; CL, cell lines; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; DLBCL, diffuse large B-cell lymphoma; MCLs, mantle cell lymphomas; MDS, myelodysplastic syndrome; MM, multiple myeloma; N, number of patients; NHL, Non-Hodgkin’s lymphoma; PBMC, peripheral blood mononuclear cells; T-ALL, T-cell ALL; TC, tissue culture; PS, patient study; T-cell acute lymphoblastic leukemia; UP/DOWN, up-/downregulation of miRNA expression.
Table 2. Aberrant miRNA expression in patients with different types of hematologic malignancies with potential for clinical utility in the diagnosis, prognosis, and monitoring of therapy response.
Table 2. Aberrant miRNA expression in patients with different types of hematologic malignancies with potential for clinical utility in the diagnosis, prognosis, and monitoring of therapy response.
MalignancyMiRNA Sample
Type
Expression
Pattern
No. of
Patients
DiagnosisPrognosis Therapy ResponseRef.
ALLmiR-146aPlasmaUPN = 66Diagnostic marker pediatric and adult ALL DOWN
after CHT
[99]
ALL
pediatric
miR-155aBMUPN = 45 MRD, poor prognosisDOWN
after CHT
[100]
miR-155BMUPN = 42 Poor outcome [101]
miR-200c and miR-326BMDOWNN = 46 Drug resistance[102]
miR-324-3p and miR-508-5pBMDOWNN = 50 Drug resistance[103]
let-7b, miR-511, and miR-708
miR-196a, miR-383, and miR-542-5p
BM, PB DOWN
UP
N = 81Genetic subtype discrimination [104]
miR-99a, miR-100, and miR-125b UP Drug resistance
miR-10a, miR-33, miR-134, miR -214, miR -215, miR-369-5p, miR -484, miR-496, miR-518d, miR-572, miR-580, miR-599, miR-624, and miR-627UP or DOWN Clinical outcome
miR-151a-5p, miR-151b, miR-195-5p, miR-371b-5p, miR-425-5p, miR-455-5p, miR-497-5p, miR-574-5p, miR-708-5p, and miR-1266-5pBM, PBDOWN N = 16T-ALL and
B-ALL discrimination
[105]
miR-29c-5p, miR-424-5p, miR-450a-5p miR-450b-5p, miR-542-5p, and miR-629-5p UP
B-ALL
pediatric
miR-21BM, PBUPN = 75 Shorter DFS
and OS
[106]
miR-101-3p, miR-631, miR-922, miR-1324, miR-4699-5p, and miR-4774-5p BMUPN = 40 Prediction of early relapse [107]
Precursor
B-ALL
pediatric
miR-151-5p and mR-451BM
DOWNN = 189 Shorter RFS [108]
miR-1290UP
ALL and AMLmiR-128a and miR-128b BMUP in ALLN = 136ALL and AML discrimination [109]
let-7b and miR-223DOWN in ALL
AMLmiR-125bExosomesUPN = 154 Higher risk of relapse and overall death [97]
miR-363PBUPN = 162 Shorter EFS
and OS
Preference allo-HCST to CHT [110]
miR-504-3pSerumDOWNN = 134 Shorter OS [111]
miR-199b-5p, miR-301b, miR-326, miR-361-5p, miR-625, and miR-655PlasmaUP N = 8 DOWN
after CHT
[112]
AML
pediatric
miR-183BM, serumUPN = 106 Shorter PFS
and OS
[113]
miR-199aBMDOWNN = 71Higher BM blastsShorter EFSLower therapy response[114]
miR-370BM, serumDOWNN = 106 Shorter RFS
and OS
[115]
miR-146a, miR-509, miR-542, and miR-3667unknownUPN = 229 * Shorter OS [116]
CLLmiR-32-5p, miR-98-5p, and miR-374b-5pPBDOWNN = 32Early diagnosis [117]
B-CLLmiR-145-5p and miR-185-5pPlasmaDOWNN = 40B-CLL detection [118]
CMLmiR-142-5pBM, PBDOWNN = 45 Drug resistance[119]
miR-146aPlasmaDOWNN = 60 Prediction of imatinib response[120]
miR-150PlasmaDOWNN = 60 Prediction of imatinib response,
UP after CHT
[121]
miR-150PBDOWNN = 24 Potential marker for blast crisis and hematologic relapses [122]
miR-486-5pPBDOWNN = 36Early diagnosis Prediction of imatinib response,
UP after CHT
[123]
miR-20, miR-106, and miR-222,PlasmaUPN = 50 Potential markers for therapy response [124]
miR-122 and miR-126PBDOWNN = 100 Prediction
of imatinib response,
UP after CHT
[125]
DLBCLmiR-99a-5p and miR-125b-5pExosomesUP Shorter PFSCHT resistance[98]
miR-199a and miR-497 LN biopsiesUPN = 63 Longer OSDrug sensitivity
DOWN after CHT
[126]
MMmiR-19aSerumDOWNN = 108 Shorter PFS
and OS
Bortezomib
sensitivity
[127]
miR-194BMUPN = 44 Longer OS [128]
miR-223-3pBMDOWNN = 94 Shorter OS [129]
miR-410BMUPN = 97 Shorter PFS
and OS
[130]
miR-15aBMDOWNN = 117 Shorter PFS
and OS
[131]
miR-16-1MM detection
let-7b and miR-18aExosomesDOWNN = 156 Shorter PFS
and OS
[95]
miR-17 and miR-885-5pBMUPN = 163 Risk stratification [132]
miR-720 and miR-1246PBUPN = 60 Shorter PFS [133]
miR-15a-5p, miR-16-5p, miR-17-5p, and miR-20a-5pExosomesDOWNN = 330 Bortezomib
resistance
[94]
miR-15a, miR-16-1, miR-17, miR-20a, and miR-92-1PlasmaUPN = 85 Shorter PFS [134]
miR-153, miR-296, miR-490, miR-455, miR-500, and miR-642 BMDOWNN = 33 Shorter EFS [135]
miR-373, miR-548d, miR-554, and miR-888UP
miR-4254Serum, plasma **UPN = 627 Potential marker for MM [136]
miR-92a Mainly serum **UPN = 1214 ‡‡ Shorter PFS
and OS
[137]
let-7e, miR-15a, miR-16, miR-25, and miR-744DOWN
* Data from the Cancer Genome Atlas (TCGA); ** Systemic review and meta-analysis of 32 and 10 ‡‡ relevant studies. Abbreviations: ALL, acute lymphoblastic leukemia; allo-HCST, allogeneic hematopoietic stem cell transplantation; AML, acute myeloid leukemia; BM, bone marrow; CLL, chronic lymphocytic leukemia; DLBCL, diffuse large B-cell lymphoma; EFS, event-free survival; CHT, chemotherapy; LN, lymph node; MDS, myelodysplastic syndrome; MM, multiple myeloma; MRD, minimal residual diseases; OS, overall survival; PB, peripheral blood; PFS, progression-free survival; RFS, relapse-free survival; UP/DOWN, up-/downregulation of miRNA expression compared to non-malignant controls.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sevcikova, A.; Fridrichova, I.; Nikolaieva, N.; Kalinkova, L.; Omelka, R.; Martiniakova, M.; Ciernikova, S. Clinical Significance of microRNAs in Hematologic Malignancies and Hematopoietic Stem Cell Transplantation. Cancers 2023, 15, 2658. https://doi.org/10.3390/cancers15092658

AMA Style

Sevcikova A, Fridrichova I, Nikolaieva N, Kalinkova L, Omelka R, Martiniakova M, Ciernikova S. Clinical Significance of microRNAs in Hematologic Malignancies and Hematopoietic Stem Cell Transplantation. Cancers. 2023; 15(9):2658. https://doi.org/10.3390/cancers15092658

Chicago/Turabian Style

Sevcikova, Aneta, Ivana Fridrichova, Nataliia Nikolaieva, Lenka Kalinkova, Radoslav Omelka, Monika Martiniakova, and Sona Ciernikova. 2023. "Clinical Significance of microRNAs in Hematologic Malignancies and Hematopoietic Stem Cell Transplantation" Cancers 15, no. 9: 2658. https://doi.org/10.3390/cancers15092658

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