Transcriptional regulation of nuclear miRNAs in tumorigenesis (Review)

  • Authors:
    • Junjie Liu
    • Tianhao Yang
    • Zishen Huang
    • Huifang Chen
    • Yinshan Bai
  • View Affiliations

  • Published online on: May 19, 2022     https://doi.org/10.3892/ijmm.2022.5148
  • Article Number: 92
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs/miRs) are a type of endogenous non‑coding small RNA that regulates gene expression. miRNAs regulate gene expression at the post‑transcriptional level by targeting the 3'‑untranslated region (3'UTR) of cytoplasmic messenger RNAs (mRNAs). Recent research has confirmed the presence of mature miRNAs in the nucleus, which bind nascent RNA transcripts, gene promoter or enhancer regions, and regulate gene expression via epigenetic pathways. Some miRNAs have been shown to function as oncogenes or tumor suppressor genes by modulating molecular pathways involved in human cancers. Notably, a novel molecular mechanism underlying the dysregulation of miRNA expression in cancer has recently been discovered, indicating that miRNAs may be involved in tumorigenesis via a nuclear function that influences gene transcription and epigenetic states, elucidating their potential therapeutic implications. The present review article discusses the import of nuclear miRNAs, nucleus‑cytoplasm transport mechanisms and the nuclear functions of miRNAs in cancer. In addition, some software tools for predicting miRNA binding sites are also discussed. Nuclear miRNAs supplement miRNA regulatory networks in cancer as a non‑canonical aspect of miRNA action. Further research into this aspect may be critical for understanding the role of nuclear miRNAs in the development of human cancers.

Introduction

MicroRNAs (miRNAs/miRs) are 21–24 nucleotide single-stranded non-coding small RNAs that play a crucial role in the regulation of gene expression (1). miRNAs have been shown to play a key role in the cellular response to environmental stresses, such as starvation, hypoxia, oxidative stress and DNA damage. The biological importance of miRNAs has been highlighted in recent decades by the deregulation of miRNA expression in a variety of human diseases, including cancer (2,3). Several studies have found that the dysregulation of miRNA expression is closely linked to tumor initiation, growth and metastasis (46). Numerous miRNAs can function as oncogenes or tumor suppressors, causing cancer or suppressing tumor growth (7,8). Several miRNAs have been found to be involved in epigenetic, transcriptional and post-transcriptional processes (9,10). miRNA dysfunction disrupts the expression of oncogenic or tumor-suppressing target genes, which is implicated in cancer pathogenesis. The majority of mature miRNAs are known to interact with the 3′UTR of target mRNAs via complete or incomplete pairing to induce mRNA degradation or translational inhibition (11,12). miRNAs are loaded onto Argonaute (AGO) proteins in the cytoplasm to direct the RNA-induced silencing complex (RISC), which is composed of AGO, DICER, TAR-RNA-binding protein (TRBP) and trinucleotide repeat containing 6 (TNRC6), to bind to the 3′UTR (13). Post-transcriptional gene silencing in the cytoplasm is a well-known function mediated by miRNAs through the miRNA-induced silencing complex (miRISC). Furthermore, certain studies have suggested that it occurs in the 5′-UTR (14) and even in protein-coding sequences (15). However, interactions of miRNAs with other regions, such as gene promoters, have been reported (16,17). miRNAs are active in the nuclei, according to research, and several miRNAs that are prominently localized in the nucleus are associated with transcriptional regulation. To support this, the majority of miRNAs have recently been reported to be imported back into the nucleus following maturation, with abundant putative miRNA target sites in gene promoters in both the sense and antisense orientations, which regulate transcriptions in the nucleus by binding to complementary sequences in the promoters (1820) of target genes. miRNAs have been found to have two functions: Transcriptional gene activation (TGA) and transcriptional gene silencing (TGS) (21). It is now clear that the transcriptional regulation of nuclear miRNAs can promote the occurrence and progression of a variety of malignant tumors, including gastric, prostate cancer (Pca) and lung (2224). In recent years, there has been a growing body of evidence indicating an epigenetic interaction between DNA methylation modification and nuclear miRNA expression in cancer (2426). Several mature miRNAs have been presently identified to be enriched in the nuclei of various tissues and cell types using evolving technologies such as high-throughput sequencing (27). These findings shed new light onto the mechanisms and treatment strategies of miRNAs in the control of tumor gene expression (23). However, the types and numbers of miRNAs found in the nuclei of different cells, as well as their potential biological regulatory functions, remain unknown (28). The understanding of the time frame and associated mechanisms through which nuclear miRNAs regulate transcription in tumor cells is particularly limited. As a result, it is of utmost importance to investigate the regulatory networks of gene expression that not only cover nuclear miRNA expression, but also its immediate effects on different tumor cell types for cancer treatment by targeting or manipulating the expression profile of nuclear miRNAs.

The present review article discusses a number of processes involved in the biogenesis of miRNAs and their transport into the nucleus. The structure, function and roles of miRNA in the regulatory effects on AGO proteins are also discussed. In addition, some software tools for predicting miRNA binding sites in order to identify and predict the corresponding miRNAs are noted and summarized. It is demonstrated that nuclear miRNAs, through epigenetic modifications, mediate the regulatory function of TGA or TGS, as well as their importance in tumorigenesis and development. The aim of the present review was to discuss the endogenous function of nuclear miRNAs in cancer cell growth and to propose potential applications in disease treatment, such as the development of drug targets via miRNAs (29).

The process of miRNAs entering the nuclei

The majority of miRNAs have their own promoters that exist in the genome on their own (30). The promoter, in conjunction with RNA polymerase II (RNAP II), transcribes the genes encoding miRNAs into primary miRNA transcripts (pri-miRNAs) (31). A microprocessor complex (32) converts these pri-miRNAs into short 70-nucleotide stem-loop structures known as precursor miRNAs (pre-miRNAs) in the nucleus. The DROSHA RNase III enzyme and the Di George syndrome critical region 8 double-stranded-RNA binding protein comprise the microprocessor complex (DGCR8). DROSHA cleaves the primary miRNAs in order to remove a portion of the unpaired RNA sequences (33).

The nuclear export machinery, which consists of Exportin-5 complexed with GTPase (Ran GTP), transports the pre-miRNAs to the cytoplasm, where they are converted into mature miRNAs, a 22-nt miRNA duplex, by cleavage of the hairpins by the enzyme DICER (34). The asymmetry rule is a strand-selection mechanism in which only one strand is preferentially retained to form the functional miRISC, while the other strand is degraded. In most cases, the miRISC induces mRNA decay and translational suppression by interacting with complementary sequences in the 3′UTR of target gene mRNAs (Fig. 1). Several previous studies have found that miRISC is imported into the nucleus of mammalian cells, and that nuclear RISC (an ~158 kDa complex) is smaller than its cytoplasmic counterpart (a 20-fold larger complex of nearly 3 MDa). Moreover, several RISC components have been identified to function in the nucleus (21). Western blot analysis has revealed the presence of AGO protein, DICER, TRBP and GW182/TNRC6 in the nucleus, where they combine to form multi-protein complexes (35). Furthermore, miRISC plays a role in the transcriptional regulation of a variety of life activities, including cell proliferation, differentiation, development and apoptosis (11).

A large number of nuclear transport receptor proteins are directly involved in the process of miRNA import to the nucleus (36). Importin 8 and exportin 1, importin family members that can recognize nuclear localization sequences in protein cargoes and assist their active transport through the nuclear pore complex, have been found to be involved in the nuclear and cytoplasmic shuttling of miRNAs and RNA interference/RNA activation (RNAa) components (37,38). To function in the nucleus, miRNAs must first form an AGO-miRNA complex with the AGO protein, then bind to importin 8 and TNRC6A (also known as GW182) in the cytoplasmic-processing body (P body) for introduction into the nucleus (35,36). Unlike other members of the cytoplasmic RISC, AGO2 lacks a nuclear localization signal. TNRC6A also contains a nuclear localization signal (NLS) and a nuclear export signal (NES), which can be transported between the cytoplasm and the nucleus by importin 8 or exportin 1 (39,40). As a result, by combining with TNRC6A, which possesses NLS, AGO2 can function as a nuclear-cytoplasmic shuttling protein to re-localize the miRNA to the nucleus. TNRC6A functions as a scaffold in the nucleus, allowing other effector proteins to be recruited (41). TNRC6A directs AGO-miRNA to promoter region of target the gene, eventually cleaving the target RNA or retaining it in the nucleus. As a result, TNRC6A is yet another core protein involved in miRNA-mediated gene transcription (42,43). Furthermore, exportin 1 can transport AGO-TNRC6 complexes back to the cytoplasm. Consequently, inhibiting exportin 1 leads to an accumulation of TNRC6A and AGO2 in the nucleus (44). DICER and TRBP, on the other hand, are not attached to the RISC during their import into the nucleus. Kalantari et al (45) used semi-quantitative mass spectrometry to confirm that the association of AGO2 with GW182/TNRC6 and AGO3 was most well preserved and more stable in both the nuclei and cytoplasm. The interactions of AGO2 with DICER and TRBP, on the other hand, are restricted to the cytoplasm. Although cytoplasm-processed miRNAs can be introduced into the nucleus to regulate gene expression, the time frame and mechanisms through which miRNAs perform their functions in the nucleus remain to be fully elucidated.

Argonaute protein family and its structural characteristics

AGO proteins are members of the Argonaute family, which is highly specialized and conserved (46). In several organisms, they are the primary participants in the small non-coding RNA-mediated gene silencing pathway, and they are primarily responsible for regulating gene expression (47). The Argonaute family is divided into two groups: The ubiquitously expressed AGO subfamily proteins, also known as the AGO clade, and the gonad-expressed PIWI subfamily proteins, also known as the PIWI clade (48). AGO subfamily proteins are typically associated with miRNAs and short-interfering RNAs (siRNAs) and collaborate in the process of post-transcriptional gene silencing (49). The PIWI clade proteins are only found in another type of small RNA known as PIWI-interacting RNA (piRNA). Unlike the AGO clade, PIWI proteins are primarily expressed in germ cells and help to silence male germline transposons by interacting with piRNAs (50).

The majority of AGO proteins have 700-1,000 amino acid residues and have molecular weights of ~100 kDa, with 80% amino acid sequence identity (51). AGO protein amino acid sequences are highly conserved across species (Fig. 2A). The AGO proteins have an amino-terminal (N), a PIWI/Argonaute/Zwille (PAZ) domain, a middle (MID) domain and a PIWI domain (5254). It should be noted, however, that the N-terminal domain is required for small RNA loading and double-stranded small RNA unwinding (55). Mammalian cells contain four AGO proteins (AGO1-4), all of which are loaded with siRNA and miRNA duplexes (56). They have similar biochemical preferences for binding to duplex RNA (45,57), but only AGO2 cleaves completely complementary mRNA targets (58).

AGO proteins have been previously shown to play critical roles in germ cell maintenance and division, as well as in transcriptional and translational regulation (59). These proteins are also involved in the repair of double-strand breaks in DNA through homologous recombination, chromatin modifications and alternative splicing. The AGO proteins, on the other hand, are incapable of catalyzing any reaction on their own. They are the core component of the RISC, interacting directly or indirectly with various partners, such as DICER, TRBP and GW182 family proteins, which are responsible for binding to short RNA and the cleavage or inhibition of target mRNA translation (60). A growing body of evidence suggests that AGO not only functions as a critical regulator of gene expression in the cytoplasm, but also mediates the repression of miRNA transcription in the nucleus, with importin 8 acting as an essential co-factor and assisting in the localization of AGO2 in the nucleus. miRNAs have also been found to colocalize in the nucleus with the AGO-TNRC6A complex and to exhibit gene-silencing activity (39). AGO1 and AGO2 proteins, in particular, promote histone H3 lysine 9 (H3K9) methylation and play critical roles in miRNA-mediated gene regulation (61). Notably, while previous research provided knowledge of the nuclear functions of AGO clade proteins, the mechanisms underlying the transport of cytoplasmic AGO proteins into the nucleus remain largely unknown.

Furthermore, the association of AGO-bound miRISC with various cellular organelles or structures remains unknown. Data on the cellular localization of the numerous steps involved in the assembly, function and recycling of these complexes are limited. It is well understood that AGO-mediated epigenetic modification is a common phenomenon that plays a role in the transcriptional regulation of nuclear miRNAs. However, further and more in-depth knowledge of the types and quantities of modified proteins recruited by the AGO complex, such as epigenetic enzymes and transcription factors (TFs) is required.

Identification and prediction of nuclear miRNAs

A number of mature miRNAs have been found to be enriched in the nucleus using high-throughput profiling technologies, such as microarray and deep sequencing. The number and types of nuclear miRNAs found in different species, tissues and cells are also distinct, emphasizing the specificity and importance of nuclear miRNA regulation (Table I).

Table I.

Identification of nuclear miRNAs.

Table I.

Identification of nuclear miRNAs.

Tissue/cellMethodNumber of nuclear miRNAsHighly expressed miRNAs(Refs.)
Neural stemChIP/RT-qPCR21miR-16(67)
miR-30c
Mouse hemopoieticChIP/RT-qPCR4miR-709/706(68)
miR-690/467a*
HeLaMicroRNA sequencing346miR-30a-5p(35)
miR-191-5p
miR-148a-3p
HeLaRT-qPCR profiling750miR-484(69)
miR-191
Human megakaryocytePerl script21miR-19a-3p(70)
miR-197-5p
miR-30c
Human nasopharyngeal carcinomaDeep sequencing339miR-29b(71)
miR-32
miR-148a
miR-148b
Rat myoblastsMicroarray assay/RT-qPCR5miR-340-5p(72)
miR-351
miR-494
Mouse liversmiRNA microarray assay44miR-30e(73)
miR-709
miR-690
Pig granulosaDigital droplet RT-PCR12miR-122(74)
miR-142-5p
miR-192
miR-378

The database RNALocate (http://www.rna-society.org/rnalocate/) is the first database which comprehensively focuses on RNA subcellular localization. miRNA symbols from the miRBase database are also provided in RNALocate. This database also includes some miRNAs with nuclear localization (62). Other databases, such as microPIR2 (https://tools4mirs.org/software/mirna_databases/micropir2/) (63), Tools4miRs (https://tools4mirs.org/) (64) and miRWalk2.0 (http://zmf.umm.uni-heidelberg.de/mirwalk2) (65) provide predicted and validated information of miRNA-target interaction. Furthermore, the program mirSTP (miRNA transcription Start sites Tracking Program) was created to predict the transcription start sites (TSSs) of miRNAs. mirSTP can be found at http://bioinfo.vanderbilt.edu/mirSTP/ (66). The knowledge of nuclear-localized miRNAs is critical for locating the core promoters of miRNAs and integrating the control of miRNA transcription into complex regulatory networks.

In Table I, a list of various nuclear miRNAs identified in various studies (35,6774) is presented.

Transcriptional regulation by nuclear miRNA-mediated epigenetic modification

The mechanism of nuclear miRNA-mediated transcription regulation is similar to that of piRNA/PIWI-mediated transposon silencing (75). piRNAs assemble with PIWI members of AGO proteins to form piRNA-induced RNA silencing complexes (piRISCs) and modulate key signaling pathways at the transcriptional or post-transcriptional level in the piRNA/PIWI pathway (76). Studies have demonstrated that the nuclear PIWI-piRNA complexes at the cytoplasmic PIWI proteins silence their targets post-transcriptionally via piRNA-directed cleavage and the transcriptional ping-pong cycle. PIWI is translocated to the nucleus after piRISC assembly and represses transposons co-transcriptionally by inducing the formation of local heterochromatin at target transposon loci (77). CG9754 is a critical downstream pathway protein of PIWI/piRNA that works with PIWI to repress transposon transcription (78). Its effect on PIWI/piRNA-guided transcriptional silencing is realized after histone H3K9 is methylated as a result of its binding to the target DNA or RNA and the inhibition of genetic transposition by PIWI (79). The overexpression of the PIWI/piRNA pathway in tumor cells can lead to dysregulated methylation at H3K9, and some tumor suppressor genes may be inhibited (80). Similarly, the transcriptional activation or inhibition of genes by nuclear miRNAs is primarily accomplished through the mediation of epigenetic modifications such as H3K9 methylation and H3 lysine 4 (H3K4) acetylation. AGO proteins, in particular, as well as proteins from the epigenetic machinery, alter the structure of chromatin (Fig. 3). This process is critical for tumor cell growth, proliferation, invasion and migration, which has become a hot topic in tumor regulation research.

Following maturation, the majority of miRNAs are imported back into the nucleus, according to sequencing and bioinformatics analyses (81). miRNAs guide AGO proteins to promoter targets (82), such as the TATA-box motif or regions associated with transcription factors, owing to an abundance of putative miRNA target sites in gene promoters (83,84). The recruitment of AGO proteins (primarily AGO2) and the enrichment of RNAP II at regulated gene promoters then alter histone epigenetic modification and transcription factor binding. During the initiation of transcription, the DNA double helix unwinds. miRNAs typically mediate acetylation at H3K9 and histone H3 lysine 27 (H3K27), as well as methylation at H3K4, which stabilizes the promoter region and increases the recognition and binding efficiency of RNAP II, thereby activating gene transcription (8587). Some miRNAs, such as miR-26a-1, miRNA-558, miR-195-5p and miR-24-1, have been shown to induce target gene expression. Recent research, for example, has demonstrated that nucleus-enriched miR-195-5p may regulate Foxo3 expression in porcine ovarian granulosa cells, thereby regulating follicular development and atresia. The mechanism of action is considered to involve miR-195-5p binding to the Foxo3 promoter TATA box, transcription activation, histone acetylation and hypomethylation and transcriptional regulation. Of note, it was found that AGO2, rather than other AGO proteins, regulates the effect of miR-195-5p on Foxo3 transcription (88). However, PcG protein recruitment increases H3K27 trimethylation (H3K27 me3) and decreases H3K4 trimethylation (H3K4 me3), and DNA methylation is frequently observed at some miRNA-targeted promoter regions. The chromatin structure would then be altered, and the promoter region would be tightened to establish a non-permissive transcriptional status (89,90). Several studies have found that nuclear miRNAs exert an inhibitory effect on transcriptional regulation. The lysosome is the critical organelle of autophagy, according to a study on autophagy and lysosome biogenesis, and the TF EB (TFEB) is the primary regulator of lysosome biogenesis. Dephosphorylated TFEB enters the nucleus and binds to the palindromic sequence (GTCACGTGAC) at the promoter of autophagy and lysosomal biogenesis-related genes, i.e., the coordinated lysosomal expression and regulation (CLEAR) element, to activate transcription. The miR-30b-5 represses the transcription of TFEB-dependent downstream genes by targeting CLEAR, the TFEB binding motif, further inhibiting the flux of autophagy and lysosome biogenesis (91).

miRNAs are now known to be potent gene regulators. However, there is a limited understanding of the time frame and mechanisms through which miRNAs regulate transcription (92). Several in vitro studies have been conducted over the past few decades, which have involved the transfection of pre-miRNAs or mature mRNA mimics into immortalized and cancer cell lines. However, further research is required to determine how well these findings reflect endogenous miRNA functions in vivo. Emerging evidence regarding miRNAs has demonstrated the significance of the regulatory mechanisms of nuclear miRNA expression in cancer (93,94). The critical role of nuclear miRNAs in tumor initiation, growth and metastasis is discussed below.

Role of nuclear miRNAs in the transcriptional regulation of tumor cells

Previous research has suggested that changes in miRNA expression and function may contribute to cancer formation and progression, as well as to cellular responses to various types of stress (95,96). The Let-7 family, for example, influences tumor differentiation, migration, invasion and proliferation, and its members are dysregulated in various types of cancer. They can be activated or deactivated, and they can function as tumor suppressors or oncogenes (97). miR-124 has the potential to be a potent tumor suppressor in cancers with a high myc expression. A previous study discovered that miR-124 had an RNA-activating function, which resulted in the direct induction of p27 protein levels by binding to and inducing transcription on the p27 promoter region, causing G1 arrest. In breast and ovarian cancers, miR-124 can not only reverse the myc/p27/phospho-Rb protein signature to target cell proliferation, but it can also block integrin 1 and increase sensitivity to etoposide (98). Another family of onco-suppressor miRNAs, miR-34, may target cell cycle-related genes (including BCL2, MET, MYC, AXL and CDK4) and has tumor-suppressive properties that mediate apoptosis, cell cycle arrest and senescence. miR-34a, in particular, has been shown to be a direct transcriptional target of p53, which is frequently mutated in epithelial ovarian carcinomas (99,100). Furthermore, the miR-21 and miR-17-92 clusters are potent carcinogenic factors. miR-21 is a robust modulator of tumor behavior and malignant transformation. The expression of miR-21 in non-small cell lung cancer (NSCLC) tissues was regulated by the transcriptional factor nuclear factor (NF)-B binding to its promoter element. As a result, inhibiting NF-B via RNA silencing protects cells from cisplatin by decreasing miR-21 expression (101). Clusters of miR-17-92, on the other hand, are linked to cell proliferation, migration and angiogenesis. According to the findings of previous studies, miR-17-5p may inhibit the proliferation and induce the apoptosis of NSCLC H460 cells by targeting TGFR2 (102,103).

A number of factors, including matrix metalloproteinase 14 (MMP-14), circular RNAs (circRNAs_ and Cockayne syndrome protein B (CSB), play a role in the miRNA-mediated regulation of tumorigenesis (26,104,105). MMP-14, a membrane-anchored MMP that promotes tumorigenesis and aggressiveness, has been found to be highly expressed in neuroblastoma (NB) and gastric cancer. Xiang et al (26) identified one binding site of miRNA-584-5p (miR-584-5p) within the MMP-14 promoter through mining computational algorithm program and Argonaute-chromosome interaction dataset. Their results indicate that promoter-targeting miR-584-5p exerts tumor suppressive functions in NB through repressing the transcription of MMP-14. Zheng et al (106) identified adjacent binding sites of the myeloid zinc finger 1 (MZF1) and miRNA-337-3p (miR-337-3p) in the MMP-14 promoter by mining computational algorithm programs and chromatin immunoprecipitation datasets. According to the findings, miR-337-3p binds directly to the MMP-14 promoter to repress MZF1-mediated MMP-14 expression, thereby suppressing GC progression (106). circ-PLEKHM3 was identified as one of the most significantly downregulated circRNAs in ovarian cancer tissues by Zhang et al (107). circPLEKHM3 overexpression was found to inhibit cell growth, migration and epithelial-mesenchymal transition (EMT). Mechanistically, when circPLEKHM3 was downregulated in ovarian cancer, the competitive binding of miR-9 was attenuated, resulting in a decrease in miR-9-targeted gene expression, which promoted tumor cell proliferation (107). It has been have demonstrated that the binding sites of Let-7 and miR-29 in the proximal 3′UTR of CSB are highly conserved. Let-7 and miR-29 suppress CSB expression by directly targeting the 3′UTR of CSB in lung cancer cells, resulting in NSCLC cell apoptosis (104). The importance of the regulatory mechanisms of nuclear miRNA expression in cancer has been demonstrated by emerging evidence exhibiting genetic and epigenetic dysregulations of the machinery of miRNA biogenesis and the regulatory mechanisms of miRNAs.

Nuclear miRNAs activate transcriptional regulation in tumor cells

Under certain conditions, nuclear miRNAs have been shown to activate gene transcription and increase the expression of target genes. The human miR-373, which induces gene transcription by targeting the E-cadherin (CDH1) and cold-shock domain-containing protein 2 (CSDC2) promoters, was the first to be identified as an activator of gene transcription (25).

Nuclear miRNAs typically act at the promoter site to achieve molecular regulation. It has been reported, have found that RNAP II is enriched at the E-cadherin and CSDC2 promoters following miR-373 transfection (25). Furthermore, there is direct evidence that RISC interacts with miRNA targets in the nucleus, allowing for miRNA enrichment. Christofides et al (108) found that miR-548c-5p is highly enriched in human podocyte nuclei and can promote podocyte differentiation. The RISC complex, which is guided by miR-548c-5p, interacts with the promoter region of the gene to increase FOXC2 expression. There is evidence to suggest that miRNAs may function as nucleoplasmic gene regulators via a mechanism other than classic post-transcriptional repression. The nuclear miRNA-mediated regulation of gene transcriptional activation contradicts previously discovered miRNA functions involving only the degradation of genes and the inhibition of gene expression (25,109,110). Deep sequencing analysis has revealed that AGO2 is responsible for the majority of miRNAs entering the nucleus (Table II). AGO can recruit TFs and epigenetic enzymes to the promoter region of a gene, such as chromatin-modifying protein (CMP), histone demethylase and histone methyltransferase (111,112), to initiate histone modification and activate gene transcription (Fig. 3). For example, miR-589 has been shown to bind to the cyclooxygenase-2 (COX-2) promoter RNA and activate transcription. Specifically, the recognition of the promoter RNA by the miR-589-AGO2-GW182 complex may result in the recruitment of other factors, such as WDR5 and the activation of COX-2 and phospholipase A2 group IVA (PLA2G4A) gene expression (112). Turner et al (113) also provided the first in vivo evidence of RNAa by an endogenous miRNA in Caenorhabditis elegans. It has also been demonstrated that miRNA lin-4 can attract RNAP II to its promoter region to initiate transcription. This lin-4 miRNA overexpression is sufficient for autoactivation (113,114).

Table II.

Advances in the transcriptional activation of miRNAs.

Table II.

Advances in the transcriptional activation of miRNAs.

miRNAType of cellType of AGOTarget locationMechanism(Refs.)
miR-373Prostate cancerAGO2E-cadherin promoter/CSDC2 promoter/(25)
miR-24-1293TAGO2FBP1 and FANCC enhancerIncrease in H3K27ac/H3K4me1 modification and p300/CBP combination; decrease in H3K9me3 modification(86)
miR-589Lung adenocarcinomaAGO2COX-2 promoterTFs are recruited(112)
miRNA-558NeuroblastomaAGO1HPSE promoterIncrease in H3K4me3 modification and decrease in H3K9me2/H3K27me3 modification(115)
miR-6734Colorectal cancer/p21 promoterIncrease in H2B and H3 acetylation modification and decrease in H3K9me2 modification(126)
miR-H3CD4+ T-lymphocytes/HIV-15′ LTRPol II pre-initiation complexes (PICs) are assembled at TATA box(127)
miR-877-3pBladder cancer/p16 promoter/(128)
miR-195-5pPorcine granulosaAGO2Foxo3 promoterActivation of RNA polymerase II initiates transcription or epigenetic modification(88)

[i] The forward slash symbol ‘/’ indicates unknown information. AGO, Argonaute; H3K, histone H3; Pol II, polymerase II; FBP1, fructose-bisphosphatase 1; FANCC, FA complementation group C; COX-2, cyclooxygenase 2; HPSE, heparanase.

Nuclear miRNAs are currently being studied in cancer therapeutics and tumorigenesis research, in addition to regulating gene transcription activation as previously described (110112). As previously demonstrated, in NB, AGO1 interacts with RNAP II at active promoters throughout the genome. Through the binding site in the promoter, miR-558 induces the transcriptional activation of heparanase (HPSE), facilitating tumorigenesis and NB aggressiveness. miR-558, in an AGO1-dependent manner, induces the enrichment of the active epigenetic marker and RNA pol II on the HPSE promoter in NB cells, which is inhibited by repressing the miR-558-promoter interaction (115). Huang et al (111) discovered that several miRNAs (e.g., miR-370, miR-1180 and miR-1236) with predicted target sites in the Ccnb1 promoter activated the expression of cyclin B1 (CCNB1) in mouse cells and influenced tumor development or growth in vivo. AGO1 transports miR-744 to the CCNB1 promoter region, where it promotes RNAP II and H3K4 me3 trimethylation at the CCNB1 TSS. Ccnb1 overexpression has been shown to promote tumorigenesis and to function as a putative oncogene in a variety of cancers (111,116). Wang et al (116) discovered that miR-370, miR-1180 and miR-1236 bound to the P21 promoter and upregulated p21 transcription in human bladder cancer cells. p21 expression attenuated the cell cycle of bladder cancer cells, preventing invasion and metastasis (116).

Of note, some enhancer markers, such as H3K27ac, are found in miRNA genes. miRNAs, according to Zou et al (117), interact at genomic loci, where enhancer-derived RNA (eRNA) is transcribed, and increase the mRNA levels of adjacent genes by promoting a transcriptionally active chromatin state. The overexpression of miR-24-1, for example, in 293T cells can increase the expression of its neighboring genes, FBP1 and FANC. Increased levels of miR-24-1 will result in the enrichment of RNA polymerase II, p300/CBP and enhancer RNAs, as well as an increase in histone H3K27ac/H3K4 me1 modification and a decrease in H3K9 me3 modification (86). Notably, ChIP-qPCR has revealed that AGO2 is present at the enhancer locus (86). Furthermore, the protein-coding genes, ITGA9, CTDSPL, VILL and PLCD1, surround the miR26a-1 gene. However, when the seed region of the miRNAs is deleted or mutated, or when the enhancer locus is deleted, the activation is disrupted, suggesting that this function is dependent on the base-pairing of miRNA-enhancers.

Inhibitory effect of nuclear miRNAs on the transcriptional regulation of tumor cells

It is now clear that miRNAs regulate gene expression in mammalian somatic cells at both the transcriptional and post-transcriptional levels. Apart from post-transcriptional gene silencing via the RISC pathway in the cytoplasm, it is well established that the regulatory function of miRNAs in the nucleus is primarily achieved through transcriptional activation or inhibition of genes via epigenetic modifications such as H3K9 methylation and H3K4 acetylation (118). Following miRNA transport into the nuclei, AGO proteins recruit inhibitory complexes to the miRNA-targeted promoter region (Fig. 3), which are primarily composed of RISC (AGO and DICER1 proteins), PcG elements (YY1, EZH2 and SUZ12), CMPs and histone deacetylase. This interaction allows the protein inhibitor complex to move closer to the targeted promoter region, causing an increase in H3K27 me3 modifications and a decrease in H3K4 me3 modifications, altering the chromatin structure and establishing a non-permissive transcriptional status (119). Some examples of the regulatory mechanisms of nuclear miRNA-mediated transcriptional repression are presented in Table III.

Table III.

Advances in the transcriptional repression of miRNAs.

Table III.

Advances in the transcriptional repression of miRNAs.

miRNAType of cellType of AGOTarget locationMechanism(Refs.)
miR-10aBreast cancerAGO1/AGO3HOXD4 promoterIncrease in DNA methylation and H3K27me3 modification(125)
miR-320Cervical carcinomaAGO1POLR3D promoterEZH2 binding and increase in H3K27me3 modification(129)
miR-423-5pBreast cancerAGO2PR promoterIncrease in H3K9me2 modification(61)
miR-939Neuroblastoma/NF-κB/(130)
miR-584-5pNeuroblastomaAGO2MMP-14 promoterPromote enhancer enrichment of zeste homolog 2 and the modification of H3K9me2/H3K27me3(26)
miRNA-337-3pGastric cancerAGO2MMP-14 promoterIncrease in H3K9me2/H3K27me3 modification and decrease in H3K4me3 modification(106)
miR-223Myeloid progenitorsAGO1/AGO2NFI-A promoterIncreased levels of H3K4me3 activation marks and decreased H3K27me3 repressive marks(131)
miR-130aAstrocytes/AQP4 M1 promoter/(132)
miR-552Hepatoma/CYP2E1 promoterInhibited Pol II binding to the CYP2E1(133)

[i] The forward slash symbol ‘/’ indicates unknown information. AGO, Argonaute; H3K, histone H3; Pol II, polymerase II. HOXD4, homeobox D4; POLR3D, RNA polymerase III subunit D; MMP-14, matrix metalloproteinase 14; NFI-A, nuclear factor IA; AQP4, aquaporin-4; CYP2E1, cytochrome P450 family 2 subfamily E member 1.

Studies revealing the epigenetic dysregulations of nuclear miRNAs, biogenesis-based machinery and nuclear miRNA regulatory mechanisms have demonstrated the importance of miRNA expression regulation in cancer (120). This may lead to the discovery of novel predictive markers for the prognosis of cancer patients. For example, in gastric cancer, Guo et al (121) demonstrated for the first time, using a dual-luciferase report system, that YWHAZ was a target gene of miR-375 and played a role in miR-375 regulation in the malignant phenotype of gastric cancer. However, the precise mechanism through which YWHAZ regulates gastric cancer cell migration and invasion remains unknown. The overexpression of miR-375 was shown to inhibit gastric cancer cell migration invasion, EMT, and the activation of the Wnt/β-catenin signaling pathway, indicating that the miR-375/YWHAZ axis may be a novel therapeutic target for gastric cancer (121). Similarly, Liu et al (22) found that miR-675 aided gastric cancer cell proliferation and invasion by targeting the paired-like homeodomain transcription factor 1 (PITX1) and promoting EMT and the activation of the Wnt/β-catenin signaling pathway. Furthermore, the role of miR-652 as an oncogene in gastric cancer in targeting the RORA pathway can promote gastric cancer cell proliferation, migration and invasion and may be a novel predictive marker for the poor prognosis of patients with gastric cancer (122). Furthermore, the role of miR-652 in targeting the VEGF pathway can impede the formation of vascular networks and may be used to develop anti-angiogenetic agents. The molecular mechanisms of nuclear miRNAs regulating the occurrence of cancer have been explained using newly developed techniques and more detailed exploration.

Majid et al (23) used miRNA-205 to transfect three human PCa cell lines (LNCaP, PC3 and Du145), as well as a non-malignant epithelial PCa cell line (RWPE-1). Their findings revealed that miR-205 functioned as a tumor suppressor miRNA in Pca by directly targeting the tumor suppressor genes, IL24 and IL32, via the apoptotic and cell-survival pathways, and that its overexpression easily induced the expression of both genes. Further investigations revealed that miR-205 directly targeted tumor suppressor genes (IL-24 and IL-32) to suppress tumorigenesis via active histone modifications, such as 2H3K4, 3H3K4, the acetylation of Lys 9/14 of histone 3 and pol II (23). miR-584-5p has been shown to inhibit MMP-14 expression in NB cells. AGO2 has been shown to transport miR-584-5p to bind to the MMP-14 promoter, promoting EZH2 enrichment and the methylation of H3K27 and H3K9. It has also been shown to prevent MMP-14 transcription from suppressing NB cell growth, migration, invasion and angiogenesis (26). ABCG2 repression has been linked to aberrant promoter methylation in a certain type of cancer, including renal carcinoma and multiple myeloma. In colorectal cancer (CRC), the de novo DNA methyltransferase (DNMT3B) is a direct target of miR-203. Notably, miR-203 expression in CRC can be downregulated by reducing DNMT3B repression, resulting in a lower miR-203 expression in CRC and causing methylation of the ABCG2 promoter, which significantly inhibits ABCG2 expression and promotes cancer development (123). Notably, miR-215-5p has been found to target both the PCDH9 gene promoter and the mRNA 3′UTR, inhibiting PCDH9 expression, while promoting glioma cell growth and proliferation (124). Furthermore, while several studies have demonstrated the oncogenic or tumor-suppressing roles of nuclear miRNAs, there are some unresolved issues regarding the possible regulatory mechanisms modulating nuclear miRNA expression in cancers.

Conclusions and future perspectives

miRNAs have been shown to be extensively deregulated in human cancers over the past few decades, highlighting their critical role in tumor initiation, growth and metastasis. Similarly, the regulatory mechanisms that control miRNA expression are strongly linked to cancer diagnosis, prognosis and treatment, as well as to cancer pathogenesis. miRNAs typically suppress target gene expression by partially binding to complementary sequences in the 3′-UTR of target mRNAs, inhibiting translation and degrading the target mRNAs. However, additional evidence suggests that miRNA-mediated gene expression regulation also has other functions, and the 5′-UTR coding sequence or promoter of the genes could be the target region. Several small RNAs within miRNA response elements, such as circRNAs and long non-coding RNAs, have been shown to interact with miRNAs to regulate target gene expression. Current research on the regulation of miRNAs, however, is still focused on the traditional cytoplasmic pathways and how miRNAs specifically control specific types of cancer. The present review article summarized the series of processes involved in the entry of miRNAs into the nucleus, particularly new mechanisms of nuclear miRNA expression regulation in tumors. Mature miRNAs that are carried into the nucleus and the target gene promoter region by AGO proteins to activate epigenetic modification and regulate transcription perform the majority of the regulatory functions of miRNAs in the nucleus. A better understanding of nuclear miRNA regulation and the potential underlying mechanisms may aid in the understanding of the association between miRNAs and tumorigenesis. These new signs of progress will lead to the development of more precise therapeutic targets and the identification of novel biological markers for the diagnosis of tumors and cancers, as well as new avenues for future research on a variety of malignant diseases.

Acknowledgements

Not applicable.

Funding

The present study was supported by a Special Project on Key-Area R&D Program of scientific research institutions in Guangdong, China (grant no. 2021B0707010007).

Availability of data and materials

Not applicable.

Authors' contributions

JL was mainly responsible for collecting all relevant information and completing the review article. TY was were mainly responsible for performing the literature search. ZH and HC were mainly responsible for revising the manuscript. YB was responsible for the conception of the review and the assignment of tasks. There was no additional assistance with manuscript preparation. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Huang V and Li LC: miRNA goes nuclear. RNA Biol. 9:269–273. 2012. View Article : Google Scholar

2 

Syeda ZA, Langden SS, Munkhzul C, Lee M and Song SJ: Regulatory mechanism of microRNA expression in cancer. Int J Mol Sci. 21:17232020. View Article : Google Scholar

3 

Bhat IP, Rather TB, Bhat GA, Maqbool I, Akhtar K, Rashid G, Parray FQ, Besina S and Mudassar S: TEAD4 nuclear localization and regulation by miR-4269 and miR-1343-3p in colorectal carcinoma. Pathol Res Pract. 231:1537912022. View Article : Google Scholar

4 

Zheng T, Zhou Y, Xu X, Qi X, Liu J, Pu Y, Zhang S, Gao X, Luo X, Li M, et al: MiR-30c-5p loss-induced PELI1 accumulation regulates cell proliferation and migration via activating PI3K/AKT pathway in papillary thyroid carcinoma. J Transl Med. 20:202022. View Article : Google Scholar : PubMed/NCBI

5 

Li L, Wei D, Zhang J, Deng R, Tang J and Su D: MiR-641 inhibited cell proliferation and induced apoptosis by targeting NUCKS1/PI3K/AKT signaling pathway in breast cancer. Comput Math Methods Med. 2022:52038392022.PubMed/NCBI

6 

Mirzaei S, Zarrabi A, Asnaf SE, Hashemi F, Zabolian A, Hushmandi K, Raei M, Goharrizi MASB, Makvandi P, Samarghandian S, et al: The role of microRNA-338-3p in cancer: Growth, invasion, chemoresistance, and mediators. Life Sci. 268:1190052021. View Article : Google Scholar

7 

El Fatimy R, Zhang Y, Deforzh E, Ramadas M, Saravanan H, Wei Z, Rabinovsky R, Teplyuk NM, Uhlmann EJ and Krichevsky AM: A nuclear function for an oncogenic microRNA as a modulator of snRNA and splicing. Mol Cancer. 21:172022. View Article : Google Scholar : PubMed/NCBI

8 

Luo X, Dong J, He X, Shen L, Long C, Liu F, Liu X, Lin T, He D and Wei G: MiR-155-5p exerts tumor-suppressing functions in Wilms tumor by targeting IGF2 via the PI3K signaling pathway. Biomed Pharmacother. 125:1098802020. View Article : Google Scholar : PubMed/NCBI

9 

Gong R and Jiang Y: Non-coding RNAs in pancreatic ductal adenocarcinoma. Front Oncol. 10:3092020. View Article : Google Scholar

10 

Gregorova J, Vychytilova-Faltejskova P and Sevcikova S: Epigenetic regulation of MicroRNA clusters and families during tumor development. Cancers (Basel). 13:13332021. View Article : Google Scholar : PubMed/NCBI

11 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 9:4022018. View Article : Google Scholar

12 

Yang YL, Chang YH, Li CJ, Huang YH, Tsai MC, Chu PY and Lin HY: New insights into the role of miR-29a in hepatocellular carcinoma: Implications in mechanisms and theragnostics. J Pers Med. 11:2192021. View Article : Google Scholar : PubMed/NCBI

13 

Kobayashi H and Tomari Y: RISC assembly: Coordination between small RNAs and Argonaute proteins. Biochim Biophys Acta. 1859:71–81. 2016. View Article : Google Scholar

14 

Zhang J, Zhou W, Liu Y, Liu T, Li C and Wang L: Oncogenic role of microRNA-532-5p in human colorectal cancer via targeting of the 5′UTR of RUNX3. Oncol Lett. 15:7215–7220. 2018.

15 

Liu M, Roth A, Yu M, Morris R, Bersani F, Rivera MN, Lu J, Shioda T, Vasudevan S, Ramaswamy S, et al: The IGF2 intronic miR-483 selectively enhances transcription from IGF2 fetal promoters and enhances tumorigenesis. Genes Dev. 27:2543–2548. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Xu W, San LA, Wang Z and Liu Y: Identifying microRNA targets in different gene regions. BMC Bioinformatics. 15 (Suppl 7):S42014. View Article : Google Scholar : PubMed/NCBI

17 

Li Z, Lan X, Han R, Wang J, Huang Y, Sun J, Guo W and Chen H: MiR-2478 inhibits TGFβ1 expression by targeting the transcriptional activation region downstream of the TGFβ1 promoter in dairy goats. Sci Rep. 7:426272017. View Article : Google Scholar : PubMed/NCBI

18 

Guo D, Barry L, Lin SSH, Huang V and Li LC: RNAa in action: From the exception to the norm. RNA Biol. 11:1221–1225. 2014. View Article : Google Scholar

19 

Stavast CJ and Erkeland SJ: The non-canonical aspects of microRNAs: Many roads to gene regulation. Cells Basel. 8:14652019. View Article : Google Scholar

20 

Fan L, Lai R, Ma N, Dong Y, Li Y, Wu Q, Qiao J, Lu H, Gong L, Tao Z, et al: MiR-552-3p modulates transcriptional activities of FXR and LXR to ameliorate hepatic glycolipid metabolism disorder. J Hepatol. 74:8–19. 2021. View Article : Google Scholar

21 

Liu H, Lei C, He Q, Pan Z, Xiao D and Tao Y: Nuclear functions of mammalian microRNAs in gene regulation, immunity and cancer. Mol Cancer. 17:642018. View Article : Google Scholar : PubMed/NCBI

22 

Liu L, Tian YC, Mao G, Zhang YG and Han L: MiR-675 is frequently overexpressed in gastric cancer and enhances cell proliferation and invasion via targeting a potent anti-tumor gene PITX1. Cell Signal. 62:1093522019. View Article : Google Scholar

23 

Majid S, Dar AA, Saini S, Yamamura S, Hirata H, Tanaka Y, Deng G and Dahiya R: MicroRNA-205-directed transcriptional activation of tumor suppressor genes in prostate cancer. Cancer. 116:5637–5649. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Kumar R and Xi Y: MicroRNA, epigenetic machinery and lung cancer. Thorac Cancer. 2:35–44. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Place RF, Li LC, Pookot D, Noonan EJ and Dahiya R: MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci USA. 105:1608–1613. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Xiang X, Mei H, Qu H, Zhao X, Li D, Song H, Jiao W, Pu J, Huang K, Zheng L and Tong Q: MiRNA-584-5p exerts tumor suppressive functions in human neuroblastoma through repressing transcription of matrix metalloproteinase 14. Biochim Biophys Acta. 1852:1743–1754. 2015. View Article : Google Scholar

27 

Bai B, Liu H and Laiho M: Small RNA expression and deep sequencing analyses of the nucleolus reveal the presence of nucleolus-associated microRNAs. FEBS Open Bio. 4:441–449. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Catalanotto C, Cogoni C and Zardo G: MicroRNA in control of gene expression: An overview of nuclear functions. Int J Mol Sci. 17:17122016. View Article : Google Scholar

29 

Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA and Jones PA: Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 9:435–443. 2006. View Article : Google Scholar

30 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar

31 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar

32 

Lai .Eric C: Micro RNAs are complementary to 3′UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 30:363–364. 2002. View Article : Google Scholar

33 

Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S and Kim VN: The nuclear RNase III Drosha initiates microRNA processing. Nature. 425:415–419. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Yi R, Qin Y, Macara IG and Cullen BR: Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Gene Dev. 17:3011–3016. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Gagnon KT, Li L, Chu Y, Janowski BA and Corey DR: RNAi factors are present and active in human cell nuclei. Cell Rep. 6:211–221. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Weinmann L, Höck J, Ivacevic T, Ohrt T, Mutze J, Schwille P, Kremmer E, Benes V, Urlaub H and Meister G: Importin 8 is a gene silencing factor that targets argonaute proteins to distinct mRNAs. Cell. 136:496–507. 2009. View Article : Google Scholar

37 

Wei Y, Li L, Wang D, Zhang CY and Zen K: Importin 8 regulates the transport of mature microRNAs into the cell nucleus. J Biol Chem. 289:10270–10275. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Azmi AS, Uddin MH and Mohammad RM: The nuclear export protein XPO1-from biology to targeted therapy. Nat Rev Clin Oncol. 18:152–169. 2021. View Article : Google Scholar

39 

Nishi K, Nishi A, Nagasawa T and Ui-Tei K: Human TNRC6A is an argonaute-navigator protein for microRNA-mediated gene silencing in the nucleus. RNA. 19:17–35. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Daniel S, Schindler SG, Johannes D, Elisabeth K, Janina P, Stefan H, Reinhard D and Gunter M: Importin-β facilitates nuclear import of human GW proteins and balances cytoplasmic gene silencing protein levels. Nucleic Acids Res. 43:7447–7461. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Behm-Ansmant I, Rehwinkel J, Doerks T, Stark A, Bork P and Izaurralde E: MRNA degradation by miRNAs and GW182 requires both CCR4:NOT deadenylase and DCP1:DCP2 decapping complexes. Genes Dev. 20:1885–1898. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Nishi K, Takahashi T, Suzawa M, Miyakawa T, Nagasawa T, Ming Y, Tanokura M and Ui-Tei K: Control of the localization and function of a miRNA silencing component TNRC6A by argonaute protein. Nucleic Acids Res. 43:9856–9873. 2015.PubMed/NCBI

43 

Hicks JA, Li L, Matsui M, Chu Y, Volkov O, Johnson KC and Corey DR: Human GW182 paralogs are the central organizers for RNA-Mediated control of transcription. Cell Rep. 20:1543–1552. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Castanotto D, Lingeman R, Riggs AD and Rossi JJ: CRM1 mediates nuclear-cytoplasmic shuttling of mature microRNAs. Proc Natl Acad Sci USA. 106:21655–21659. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Kalantari R, Hicks JA, Li L, Gagnon KT, Sridhara V, Lemoff A, Mirzaei H and Corey DR: Stable association of RNAi machinery is conserved between the cytoplasm and nucleus of human cells. RNA. 22:1085–1098. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Kuhn CD and Joshua-Tor L: Eukaryotic argonautes come into focus. Trends Biochem Sci. 38:263–271. 2013. View Article : Google Scholar

47 

Ryazansky S, Kulbachinskiy A and Aravin AA: The expanded universe of prokaryotic argonaute proteins. mBio. 9:e01935–18. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Peters L and Meister G: Argonaute proteins: Mediators of RNA silencing. Mol Cell. 26:611–623. 2007. View Article : Google Scholar

49 

Hutvagner G and Simard MJ: Argonaute proteins: Key players in RNA silencing. Nat Rev Mol Cell Biol. 9:22–32. 2008. View Article : Google Scholar

50 

Siomi MC, Sato K, Pezic D and Aravin AA: PIWI-interacting small RNAs: The vanguard of genome defence. Nat Rev Mol Cell Biol. 12:246–258. 2011. View Article : Google Scholar

51 

Sasaki T, Shiohama A, Minoshima S and Shimizu N: Identification of eight members of the argonaute family in the human genome. Genomics. 82:323–330. 2003. View Article : Google Scholar : PubMed/NCBI

52 

Faehnle CR, Elkayam E, Haase AD, Hannon GJ and Joshua-Tor L: The making of a slicer: Activation of human argonaute-1. Cell Rep. 3:1901–1909. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Schirle NT, Sheu-Gruttadauria J, Chandradoss SD, Joo C and MacRae IJ: Water-mediated recognition of t1-adenosine anchors argonaute2 to microRNA targets. Elife. 4:e076462015. View Article : Google Scholar

54 

Park MS, Phan HD, Busch F, Hinckley SH, Brackbill JA, Wysocki VH and Nakanishi K: Human argonaute3 has slicer activity. Nucleic Acids Res. 45:11867–11877. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Kwak PB and Tomari Y: The N domain of argonaute drives duplex unwinding during RISC assembly. Nat Struct Mol Biol. 19:145–151. 2012. View Article : Google Scholar

56 

Czech B and Hannon GJ: Small RNA sorting: Matchmaking for argonautes. Nat Rev Genet. 12:19–31. 2011. View Article : Google Scholar

57 

Yoda M, Kawamata T, Paroo Z, Ye X, Iwasaki S, Liu Q and Tomari Y: ATP-dependent human RISC assembly pathways. Nat Struct Mol Biol. 17:17–23. 2010. View Article : Google Scholar

58 

Liu J, Carmell MA, Rivas FV, Marsden CG, Thomson JM, Song JJ, Hammond SM, Joshua-Tor L and Hannon GJ: Argonaute2 is the catalytic engine of mammalian RNAi. Science. 305:1437–1441. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Huang V and Li LC: Demystifying the nuclear function of argonaute proteins. RNA Biol. 11:18–24. 2014. View Article : Google Scholar

60 

Huntzinger E and Izaurralde E: Gene silencing by microRNAs: Contributions of translational repression and mRNA decay. Nat Rev Genet. 12:99–110. 2011. View Article : Google Scholar

61 

Younger ST and Corey DR: Transcriptional gene silencing in mammalian cells by miRNA mimics that target gene promoters. Nucleic Acids Res. 39:5682–5691. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Zhang T, Tan P, Wang L, Jin N, Li Y, Zhang L, Yang H, Hu Z, Zhang L, Hu C, et al: RNALocate: A resource for RNA subcellular localizations. Nucleic Acids Res. 45:D135–D138. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Piriyapongsa J, Bootchai C, Ngamphiw C and Tongsima S: MicroPIR2: A comprehensive database for human-mouse comparative study of microRNA-promoter interactions. Database (Oxford). 2014:bau1152014. View Article : Google Scholar : PubMed/NCBI

64 

Lukasik A, Wójcikowski M and Zielenkiewicz P: Tools4miRs-one place to gather all the tools for miRNA analysis. Bioinformatics. 32:2722–2724. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Parveen A, Gretz N and Dweep H: Obtaining miRNA-target interaction information from miRWalk2.0. Curr Protoc Bioinformatics. 55:12.15.1–12.15.27. 2016. View Article : Google Scholar

66 

Liu Q, Wang J, Zhao Y, Li CI, Stengel KR, Acharya P, Johnston G, Hiebert SW and Shyr Y: Identification of active miRNA promoters from nuclear run-on RNA sequencing. Nucleic Acids Res. 45:e1212017. View Article : Google Scholar : PubMed/NCBI

67 

Jeffries CD, Fried HM and Perkins DO: Nuclear and cytoplasmic localization of neural stem cell microRNAs. RNA. 17:675–86. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Wong JJ, Ritchie W, Gao D, Lau KA, Gonzalez M, Choudhary A, Taft RJ, Rasko JE and Holst J: Identification of nuclear-enriched miRNAs during mouse granulopoiesis. J Hematol Oncol. 7:422014. View Article : Google Scholar

69 

Li ZF, Liang YM, Lau PN, Shen W, Wang DK, Cheung WT, Xue CJ, Poon LM and Lam YW: Dynamic localisation of mature microRNAs in human nucleoli is influenced by exogenous genetic materials. PLoS One. 8:e708692013. View Article : Google Scholar : PubMed/NCBI

70 

Sahu I, Hebalkar R, Kar S, Sreevidya TS, Gutti U and Gutti RK: Systems biology approach to study the role of miRNA in promoter targeting during megakaryopoiesis. Exp Cell Res. 366:192–198. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Liao JY, Ma LM, Guo YH, Zhang YC, Zhou H, Shao P, Chen YQ and Qu LH: Deep sequencing of human nuclear and cytoplasmic small RNAs reveals an unexpectedly complex subcellular distribution of miRNAs and tRNA 3′trailers. PLoS One. 5:e105632010. View Article : Google Scholar : PubMed/NCBI

72 

Politz JCR, Hogan EM and Pederson T: MicroRNAs with a nucleolar location. RNA. 15:1705–1715. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Tang R, Li L, Zhu D, Hou D, Cao T, Gu H, Zhang J, Chen J, Zhang CY and Zen K: Mouse miRNA-709 directly regulates miRNA-15a/16-1 biogenesis at the posttranscriptional level in the nucleus: Evidence for a microRNA hierarchy system. Cell Res. 22:504–515. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Toms D, Pan B, Bai Y and Li J: Small RNA sequencing reveals distinct nuclear microRNAs in pig granulosa cells during ovarian follicle growth. J Ovarian Res. 14:542021. View Article : Google Scholar : PubMed/NCBI

75 

Sato K and Siomi MC: The piRNA pathway in Drosophila ovarian germ and somatic cells. Proc Jpn Acad Ser B Phys Biol Sci. 96:32–42. 2020. View Article : Google Scholar

76 

Gunawardane LS, Saito K, Nishida KM, Miyoshi K, Kawamura Y, Nagami T, Siomi H and Siomi MC: A slicer-mediated mechanism for repeat-associated siRNA 59 end formation in drosophila. Science. 315:1587–1590. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Brennecke J, Aravin AA, Stark A, Dus M, Kellis M, Sachidanandam R and Hannon GJ: Discrete small RNA-generating loci as master regulators of transposon activity in drosophila. Cell. 128:1089–1103. 2007. View Article : Google Scholar

78 

Yu Y, Gu J, Jin Y, Luo Y, Preall JB, Ma J, Czech B and Hannon GJ: Panoramix enforces piRNA-dependent cotranscriptional silencing. Science. 350:339–342. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Sienski G, Donertas D and Brennecke J: Transcriptional silencing of transposons by Piwi and maelstrom and its impact on chromatin state and gene expression. Cell. 151:964–980. 2012. View Article : Google Scholar

80 

Watanabe T, Tomizawa S, Mitsuya K, Totoki Y, Yamamoto Y, Kuramochi-Miyagawa S, Iida N, Hoki Y, Murphy PJ, Toyoda A, et al: Role for piRNAs and noncoding RNA in de novo DNA methylation of the imprinted mouse Rasgrf1 locus. Science. 332:848–852. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Zhang N, Hu G, Myers TG and Williamson PR: Protocols for the analysis of microRNA expression, biogenesis, and function in immune cells. Curr Protoc Immunol. 126:e782019. View Article : Google Scholar

82 

Fu Y, Zhang L, Zhang R, Xu S, Wang H, Jin Y and Wu Z: Enterovirus 71 suppresses miR-17-92 cluster through up-regulating methylation of the miRNA promoter. Front Microbiol. 10:6252019. View Article : Google Scholar

83 

Younger ST, Pertsemlidis A and Corey DR: Predicting potential miRNA target sites within gene promoters. Bioorg Med Chem Lett. 19:3791–3794. 2009. View Article : Google Scholar

84 

Chellini L, Frezza V and Paronetto MP: Dissecting the transcriptional regulatory networks of promoter-associated noncoding RNAs in development and cancer. J Exp Clin Cancer Res. 39:512020. View Article : Google Scholar : PubMed/NCBI

85 

Zhang Y and Zhang H: RNAa induced by TATA box-targeting microRNAs. Adv Exp Med Biol. 983:91–111. 2017. View Article : Google Scholar

86 

Xiao M, Li J, Li W, Wang Y, Wu F, Xi Y, Zhang L, Ding C, Luo H, Li Y, et al: MicroRNAs activate gene transcription epigenetically as an enhancer trigger. RNA Biol. 14:1326–1334. 2017. View Article : Google Scholar

87 

Zhang Y, Liu W, Chen Y, Liu J, Wu K, Su L, Zhang W, Jiang Y, Zhang X, Zhang Y, et al: A cellular microRNA facilitates regulatory t lymphocyte development by targeting the FOXP3 promoter TATA-box motif. J Immunol. 200:1053–1063. 2017. View Article : Google Scholar

88 

Bai Y, Pan B, Zhan X, Silver H and Li J: MicroRNA 195-5p targets foxo3 promoter region to regulate its expression in granulosa cells. Int J Mol Sci. 22:67212021. View Article : Google Scholar

89 

Cao R and Zhang Y: The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3. Curr Opin Genet Dev. 14:155–164. 2004. View Article : Google Scholar : PubMed/NCBI

90 

Mellor J, Dudek P and Clynes D: A glimpse into the epigenetic landscape of gene regulation. Curr Opin Genet Dev. 18:116–122. 2008. View Article : Google Scholar : PubMed/NCBI

91 

Guo H, Pu M, Tai Y, Chen Y and Ren J: Nuclear miR-30b-5p suppresses TFEB-mediated lysosomal biogenesis and autophagy. Cell Death Differ. 28:320–336. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Li LC: Chromatin remodeling by the small RNA machinery in mammalian cells. Epigenetics. 9:45–52. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Barlak N, Capik O, Kilic A, Sanli F, Aytatli A, Yazici A, Karatas EA, Ortucu S and Karatas OF: MicroRNA-145 transcriptionally regulates semaphorin 3A expression in prostate cancer cells. Cell Biol Int. 45:1082–1090. 2021. View Article : Google Scholar

94 

Song M, Wang Y, Zhou P, Wang J, Xu H and Zheng J: MicroRNA-361-5p aggravates acute pancreatitis by promoting interleukin-17A secretion via impairment of nuclear factor IA-dependent hes1 downregulation. J Med Chem. 64:16541–16552. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Zhang K, Wang YY, Xu Y, Zhang L, Zhu J, Si PC, Wang YW and Ma R: A two-miRNA signature of upregulated miR-185-5p and miR-362-5p as a blood biomarker for breast cancer. Pathol Res Pract. 222:1534582021. View Article : Google Scholar

96 

Van Roosbroeck K and Calin GA: Cancer hallmarks and MicroRNAs: The therapeutic connection. Adv Cancer Res. 135:119–149. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Kolenda T, Przybyla W, Teresiak A, Mackiewicz A and Lamperska KM: The mystery of let-7d-a small RNA with great power. Contemp Oncol (Pozn). 18:293–301. 2014.PubMed/NCBI

98 

Seviour EG, Sehgal V, Lu Y, Luo Z, Moss T, Zhang F, Hill SM, Liu W, Maiti SN, Cooper L, et al: Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene. 35:691–701. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Schmid G, Notaro S, Reimer D, Abdel-Azim S, Duggan-Peer M, Holly J, Fiegl H, Rossler J, Wiedemair A, Concin N, et al: Expression and promotor hypermethylation of miR-34a in the various histological subtypes of ovarian cancer. BMC Cancer. 16:1022016. View Article : Google Scholar : PubMed/NCBI

100 

Wong KY, Yu L and Chim CS: DNA methylation of tumor suppressor miRNA genes: A lesson from the miR-34 family. Epigenomics. 3:83–92. 2011. View Article : Google Scholar : PubMed/NCBI

101 

Yang Z, Fang S, Di Y, Ying W, Tan Y and Gu W: Modulation of NF-κB/miR-21/PTEN pathway sensitizes non-small cell lung cancer to cisplatin. PLoS One. 10:e01215472015. View Article : Google Scholar : PubMed/NCBI

102 

Xu X, Zhu S, Tao Z and Ye S: High circulating miR-18a, miR-20a, and miR-92a expression correlates with poor prognosis in patients with non-small cell lung cancer. Cancer Med. 7:21–31. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Li H, Zhou H, Luo J and Huang J: MicroRNA-17-5p inhibits proliferation and triggers apoptosis in non-small cell lung cancer by targeting transforming growth factor β receptor 2. Exp Ther Med. 13:2715–2722. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Yang Z, Liu C, Wu H, Xie Y and Zhang X: CSB affected on the sensitivity of lung cancer cells to platinum-based drugs through the global decrease of let-7 and miR-29. BMC Cancer. 19:9482019. View Article : Google Scholar : PubMed/NCBI

105 

Kristensen LS, Andersen MS, Stagsted L, Ebbesen KK, Hansen TB and Kjems J: The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 20:675–691. 2019. View Article : Google Scholar

106 

Zheng L, Jiao W, Mei H, Song H, Li D, Xiang X, Chen Y, Yang F, Li H, Huang K and Tong Q: MiRNA-337-3p inhibits gastric cancer progression through repressing myeloid zinc finger 1-facilitated expression of matrix metalloproteinase 14. Oncotarget. 7:40314–40328. 2016. View Article : Google Scholar

107 

Zhang L, Zhou Q, Qiu Q, Hou L and Lu Y: CircPLEKHM3 acts as a tumor suppressor through regulation of the miR-9/BRCA1/DNAJB6/KLF4/AKT1 axis in ovarian cancer. Mol Cancer. 18:1442019. View Article : Google Scholar : PubMed/NCBI

108 

Christofides A, Papagregoriou G, Dweep H, Makrides N, Gretz N, Felekkis K and Deltas C: Evidence for miR-548c-5p regulation of FOXC2 transcription through a distal genomic target site in human podocytes. Cell Mol Life Sci. 77:2441–2459. 2020. View Article : Google Scholar

109 

Dharap A, Pokrzywa C, Murali S, Pandi G and Vemuganti R: MicroRNA miR-324-3p induces promoter-mediated expression of RelA gene. PLoS One. 8:e794672013. View Article : Google Scholar : PubMed/NCBI

110 

Huang V: Endogenous miRNAa: MiRNA-mediated gene upregulation. Adv Exp Med Biol. 983:65–79. 2017. View Article : Google Scholar

111 

Huang V, Place RF, Portnoy V, Wang J, Qi Z, Jia Z, Yu A, Shuman M, Yu J and Li LC: Upregulation of Cyclin B1 by miRNA and its implications in cancer. Nucleic Acids Res. 40:1695–1707. 2012. View Article : Google Scholar : PubMed/NCBI

112 

Matsui M, Chu Y, Zhang H, Gagnon KT, Shaikh S, Kuchimanchi S, Manoharan M, Corey DR and Janowski BA: Promoter RNA links transcriptional regulation of inflammatory pathway genes. Nucleic Acids Res. 41:10086–10109. 2013. View Article : Google Scholar : PubMed/NCBI

113 

Turner M, Jiao A and Slack FJ: Autoregulation of lin-4 microRNA transcription by RNA activation (RNAa) in C. Elegans. Cell Cycle. 13:772–781. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Vera H, Yi Q, Ji W, Xiaoling W, Place RF, Guiting L, Lue TF, Long-Cheng L and Dong-Yan J: RNAa is conserved in mammalian cells. PLoS One. 5:e88482010. View Article : Google Scholar : PubMed/NCBI

115 

Qu H, Zheng L, Pu J, Mei H, Xiang X, Zhao X, Li D, Li S, Mao L, Huang K and Tong Q: MiRNA-558 promotes tumorigenesis and aggressiveness of neuroblastoma cells through activating the transcription of heparanase. Hum Mol Genet. 24:2539–2551. 2015. View Article : Google Scholar

116 

Wang C, Chen Z, Ge Q, Hu J, Li F, Hu J, Xu H, Ye Z and Li LC: Up-regulation of p21(WAF1/CIP1) by miRNAs and its implications in bladder cancer cells. FEBS Lett. 588:4654–4664. 2014. View Article : Google Scholar

117 

Zou Q, Liang Y, Luo H and Yu W: MiRNA-mediated RNAa by targeting enhancers. Adv Exp Med Biol. 983:113–125. 2017. View Article : Google Scholar

118 

Huang YP, Qiu LZ and Zhou GP: MicroRNA-939 down-regulates CD2-associated protein by targeting promoter in HEK-293T cells. Renal Failure. 38:508–513. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Mao H, Zhu C, Zong D, Weng C, Yang X, Huang H, Liu D, Feng X and Guang S: The nrde pathway mediates small-RNA-directed histone H3 lysine 27 Trimethylation in Caenorhabditis elegans. Curr Biol. 25:2398–2403. 2015. View Article : Google Scholar

120 

Liu X, Fan Z, Li Y, Li Z, Zhou Z, Yu X, Wan J, Min Z, Yang L and Li D: MicroRNA-196a-5p inhibits testicular germ cell tumor progression via NR6A1/E-cadherin axis. Cancer Med. 9:9107–9122. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Guo F, Gao Y, Sui G, Jiao D, Sun L, Fu Q and Jin C: MiR-375-3p/YWHAZ/β-catenin axis regulates migration, invasion, EMT in gastric cancer cells. Clin Exp Pharmacol Physiol. 46:144–152. 2019. View Article : Google Scholar

122 

Li J and Zou X: MiR-652 serves as a prognostic biomarker in gastric cancer and promotes tumor proliferation, migration, and invasion via targeting RORA. Cancer Biomark. 26:323–331. 2019. View Article : Google Scholar : PubMed/NCBI

123 

To KK, Leung WW and Ng SS: A novel miR-203-DNMT3b-ABCG2 regulatory pathway predisposing colorectal cancer development. Mol Carcinog. 56:4642016. View Article : Google Scholar : PubMed/NCBI

124 

Wang C, Chen Q, Li S, Li S and Zhao Z: Dual inhibition of PCDH9 expression by miR-215-5p up-regulation in gliomas. Oncotarget. 8:10287–10297. 2016. View Article : Google Scholar

125 

Tan Y, Zhang B, Wu T, Skogerbø G, Zhu X, Guo X, He S and Chen R: Transcriptional inhibiton of Hoxd4 expression by miRNA-10a in human breast cancer cells. BMC Mol Biol. 10:122009. View Article : Google Scholar

126 

Kang MR, Park KH, Yang JO, Lee CW and Kang JS: MiR-6734 up-regulates p21 gene expression and induces cell cycle arrest and apoptosis in colon cancer cells. PLoS One. 11:e1609612016. View Article : Google Scholar

127 

Zhang Y, Fan M, Geng G, Liu B, Huang Z, Luo H, Zhou J, Guo X, Cai W and Zhang H: A novel HIV-1-encoded microRNA enhances its viral replication by targeting the TATA box region. Retrovirology. 11:232014. View Article : Google Scholar : PubMed/NCBI

128 

Li S, Zhu Y, Liang Z, Wang X and Xie L: Up-regulation of p16 by miR-877-3p inhibits proliferation of bladder cancer. Oncotarget. 7:51773–51783. 2016. View Article : Google Scholar

129 

Kim DH, Saetrom P, Snove O Jr and Rossi JJ: MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci USA. 105:16230–16235. 2008. View Article : Google Scholar : PubMed/NCBI

130 

Cui C, Yu J, Huang S, Zhu H and Huang Z: Transcriptional regulation of gene expression by microRNAs as endogenous decoys of transcription factors. Cell Physiol Biochem. 33:1698–1714. 2014. View Article : Google Scholar : PubMed/NCBI

131 

Zardo G, Ciolfi A, Vian L, Starnes LM, Billi M, Racanicchi S, Maresca C, Fazi F, Travaglini L, Noguera N, et al: Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood. 119:4034–4046. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Sepramaniam S, Ying LK, Armugam A, Wintour EM and Jeyaseelan K: MicroRNA-130a represses transcriptional activity of aquaporin 4 M1 promoter. J Biol Chem. 287:12006–12015. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Miao L, Yao H, Li C, Pu M, Yao X, Yang H, Qi X, Ren J and Wang Y: A dual inhibition: MicroRNA-552 suppresses both transcription and translation of cytochrome P450 2E1. Biochim Biophys Acta. 1859:650–662. 2016. View Article : Google Scholar

Related Articles

Journal Cover

July-2022
Volume 50 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu J, Yang T, Huang Z, Chen H and Bai Y: Transcriptional regulation of nuclear miRNAs in tumorigenesis (Review). Int J Mol Med 50: 92, 2022
APA
Liu, J., Yang, T., Huang, Z., Chen, H., & Bai, Y. (2022). Transcriptional regulation of nuclear miRNAs in tumorigenesis (Review). International Journal of Molecular Medicine, 50, 92. https://doi.org/10.3892/ijmm.2022.5148
MLA
Liu, J., Yang, T., Huang, Z., Chen, H., Bai, Y."Transcriptional regulation of nuclear miRNAs in tumorigenesis (Review)". International Journal of Molecular Medicine 50.1 (2022): 92.
Chicago
Liu, J., Yang, T., Huang, Z., Chen, H., Bai, Y."Transcriptional regulation of nuclear miRNAs in tumorigenesis (Review)". International Journal of Molecular Medicine 50, no. 1 (2022): 92. https://doi.org/10.3892/ijmm.2022.5148