- Review
- Open access
- Published:
Nuclear-activating miRNAs: unveiling the intricacies of subcellular miRNA function and regulation in cancer and immunity disease
Cancer Cell International volume 25, Article number: 147 (2025)
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that traditionally recognized as negative regulators of gene expression through post-transcriptional regulation in the cytoplasm. However, recent discoveries have unveiled some novel miRNA functions in the cell nucleus, where a subset of miRNAs, termed nuclear-activating miRNAs (NamiRNAs), play pivotal roles in gene activation and transcriptional regulation for cancer and immunity disease. The discovery of NamiRNAs demonstrated a complementary regulatory function of miRNA, showing their differential activities in the nucleus and cytoplasm. This review aims to explore the biogenesis, mechanisms, and regulatory functions of NamiRNAs, deciphering their involvement in NamiRNA-gene network for gene expression modulation, and emerging significance as drug targets against cancer.
Introduction
The first discovery of microRNAs (miRNAs) can be dated back to the early 1990s, where lin-4 was identified by Victor Ambros and colleagues in the nematode Caenorhabditis elegans [1]. In 2000, Ruvkun lab discovered and reported a second miRNA, let-7, in C. elegans [2]. However, the conservation of let-7 across species underscored the importance of miRNAs in evolution and development of biology. Subsequent research in the early 2000s revealed that miRNAs are widespread in nature, and not restricted in peculiarities of C. elegans. Researchers gradually find out that miRNAs act as a fundamental component of gene regulation in many organisms, including humans [3]. The canonical pathway of miRNA biogenesis involves transcription and production of primary miRNA (pri-miRNA) with further cleavage into a precursor miRNA (pre-miRNA). The pre-miRNA is exported to the cytoplasm to process into a mature miRNA. The mature miRNA is incorporated into the RNA-induced silencing complex (RISC) and interact with target mRNA to repress gene expression [4]. miRNAs play crucial roles in various biological processes, including cell proliferation, differentiation, apoptosis, and metabolic regulation [4].
The functional divergence of miRNAs into cytoplasmic and nuclear roles underscores the importance of RISC assembly and nuclear import mechanisms in determining their functional outcomes [5, 6]. These distinctions highlight a complex regulatory interplay that is both conserved and diversified across species, reflecting the evolutionary adaptation of RNA-based regulatory pathways. For example, RNA interference (RNAi) and RNA activation (RNAa) serve as essential regulatory mechanisms in eukaryotic systems, though their processes exhibit significant differences between mammals and plants. In plants, RNAi primarily involves small interfering RNAs (siRNAs), which guide the RISC complex to degrade complementary mRNAs, thus ensuring robust antiviral defense and precise gene regulation. In mammals, RNAi is typically mediated by miRNAs in the cytoplasm, where they mainly repress translation or promote mRNA degradation.
Since Rovkun elucidated the theoretical mechanism of miRNA-mediated negative regulation, the notion of miRNAs functioning as cytoplasmic negative regulators has become widely accepted within the scientific community [2]. However, numerous studies have revealed that miRNAs can also positively regulate target genes under specific circumstances. These positive effects are often mediated through transcription factors or other regulatory elements, enabling miRNAs to activate target genes [7,8,9,10,11,12,13,14,15]. Emerging evidence further suggests that miRNAs are not confined to the cytoplasm; they are also found in the nucleus and other organelles [16], where they play significant roles, such as influencing tumor drug resistance. In 2017, Yu and colleagues systematically analyzed the ENCODE database and identified over 300 miRNA precursors, out of 1,594 examined across seven cell lines, that exhibited significant overlap with genomic enhancers. Notably, enhancers are marked by classic epigenetic signatures, including H3K27ac and H3K4me1. These nuclear-localized miRNAs, termed nuclear-activating miRNAs (NamiRNAs), activate nearby target genes [7, 15]. NamiRNAs interact with key regulatory elements of genes, such as promoters, enhancers, or even gene bodies, modulating gene expression at the transcriptional level. Unlike the traditional role of miRNAs in gene silencing, NamiRNAs engage in gene activation by recruiting transcription factors or altering the chromatin’s epigenetic state to promote transcription [17,18,19,20].
Biogenesis of canonical MiRNA and NamiRNA
Biogenesis of canonical MiRNA
The biogenesis of canonical miRNAs is a complex, multi-step process that occurs sequentially in the nucleus and cytoplasm of eukaryotic cells (Fig. 1a) [5, 6]. Initially, miRNA genes are transcribed by RNA polymerase II (RNA pol II) to produce pri-miRNAs. These pri-miRNAs are then processed by the DiGeorge syndrome critical region 8 (DGCR8) protein and the Drosha microprocessor complex, which ensure precise cleavage into pre-miRNAs [4]. Pri-miRNAs are characterized by an unstructured hairpin loop and flanking sequences at their 5′ and 3′ ends. In contrast, pre-miRNAs possess distinct structural features, including a hydroxyl group (OH) at the 3′ end, two nucleotides forming a 3′ overhang, and a phosphate (P) group at the 5′ end. The pre-miRNA is then transported from the nucleus to the cytoplasm by the nuclear export receptor Exportin-5 in a process that depends on the small GTP-binding protein Ran-GTP [21].
Schematic illustration of the biogenesis of canonical miRNA (a) and the network for NamiRNAs, enhancers, and gene activation (b) [15]
In the cytoplasm, Dicer processes the pre-miRNAs by recognizing the 5′ phosphate, 3′ overhang, and loop structure [5, 6]. It then cleaves the pre-miRNAs to produce ~ 22 bp miRNA duplexes, each with a 5′ phosphate and a 3′ two-nucleotide overhang on both strands. This catalytic process is modulated by interactions between Dicer and accessory proteins, including TAR RNA-binding protein 2 (TARBP2) and protein activator of the interferon-induced protein kinase (PACT). The resulting miRNA duplex is subsequently loaded onto Argonaute (AGO) proteins with the assistance of heat shock cognate protein 70 (HSC70) and heat shock protein 90 (HSP90), which maintain AGO in an open conformation to ensure successful loading [22]. After the duplex is loaded, the passenger strand (or miRNA) is expelled, leaving the guide strand retained within AGO. This guide strand directs the assembly of the RNA-induced silencing complex (RISC), enabling interaction with target mRNAs to suppress gene expression. miRNAs were originally characterized as post-transcriptional regulators of gene expression, mediating silencing by binding to complementary sequences on target mRNAs.
Biogenesis of NamiRNA
NamiRNAs, like other miRNAs, undergo initial transcription and processing within the nucleus [7, 14, 15]. Functionally, they play a role in gene activation by recruiting transcription factors or modifying the chromatin epigenetic state to facilitate gene transcription. Recent studies have revealed that NamiRNAs can be transcribed from miRNA genes by RNA polymerase II into primary transcripts. These transcripts are subsequently processed by RNA processing factors, such as cleavage and polyadenylation specificity factor 1 (CPSF1) and cleavage and polyadenylation specificity factor 3 (CPSF3). These factors remove intronic sequences and generate precursor miRNA (pre-miRNA) species in the nucleus [23]. In the nucleus, pre-miRNA is processed by Dicer and other RNA processing enzymes to generate double stranded miRNA. The double stranded miRNA unwinds and releases single strand miRNA to create RISC like structure. This structure guides the NamiRNA based pairing on enhancers and promoters of targeted gene to realize activation.
Certain NamiRNAs, initially synthesized through a pathway similar to canonical miRNAs, can shuttle back to the nucleus to mediate gene activation. Following their maturation in the cytoplasm, these NamiRNAs associate with a similar RISC-like complex, enabling their functional activation. This process is facilitated by Fragile X mental retardation syndrome-related protein 1 (FXR1), which plays a crucial role in their nuclear re-import. Once inside the nucleus, NamiRNAs engage with target genes and enhancers, promoting transcriptional activation [7, 14].
Mechanisms of NamiRNAs function
Mechanisms of NamiRNA-enhancer interaction
NamiRNAs and enhancer share several unique characteristics, including specific histone modification (e.g., H3K27ac and H3K4me1), binding of the P300/cAMP response element-binding protein (CREB) coactivator, DNase I hypersensitivity, and enhancer RNA (eRNA) production [15, 24,25,26] (Fig. 1b). Among 1,594 human miRNA precursor loci, 303 show partial overlap with enhancer regions enriched for H3K27ac [7]. For example, miR-24-1 has been shown to unconventionally activate gene transcription by targeting enhancers. This activation was completely abolished when the enhancer sequence was deleted using TALEN technology [7]. NamiRNA-enhancer interactions can modulate enhancer properties, such as influencing transcription and chromatin looping via nuclear AGO2 and RNA polymerase II (Pol II) and recruiting p300 to catalyze H3K27ac at activated enhancer sites. However, gene activation requires the interaction of active and intact enhancers with NamiRNAs. This suggests that inactive enhancers are unable to interact effectively with NamiRNAs.
Suzuki et al. reported that super-enhancers play a critical role in the biogenesis of master miRNAs essential for maintaining cell identity by enhancing both transcription and Drosha/DGCR8-mediated processing of primary miRNAs (pri-miRNAs). Together with broad H3K4me3 domains, super-enhancers establish a tissue-specific and evolutionarily conserved miRNA expression and functional landscape [8].
Mechanisms of NamiRNA-promoter interaction
The interaction of NamiRNAs with promoters is a complicated process that involves gene regulations and cellular responses. In recent years, researchers have been endowing their efforts in digging in the precise molecular mechanisms underlying NamiRNAs-promoter interactions and their roles in various diseases, contributing to targeted therapy and diagnostic development [9, 10]. NamiRNAs bind to specific sequences of targeted promoter genes via complementary base pair between NamiRNA and the targeted DNA [15]. This process can be facilitated by RNA-binding proteins (RBPs) as it can assist the recognition and alignment of NamiRNA and DNA. Typically, the binding of NamiRNAs to promoter region can prevent the recruitment of transcription factors and RNA polymerase to targeted genes, thus limiting gene transcription and expression [7]. Furthermore, the interactions between NamiRNAs and promoter regulate the process and stability of target mRNAs, affecting their translation and overall expression levels. In some cases, NamiRNA-promoter communication induces chromatin remodeling, resulting in varied gene accessibility and increased transcription factor recruitment [11]. This further promote the expression of key genes responsible for cell growth, differentiation and survival, and affecting signaling pathways regulation and cellular homeostasis. It may affect immune response, disease progression, cancer and drug resistance by regulating oncogene expression and drug targets.
The binding of miRNA to promoter is verified to be closely related with cancer development via the regulation of oncogene and targets. Several studies have provided examples of nuclear miRNA-promoter interaction in various cell lines. For instance, miRNA-122 can bind to the alpha-fetoprotein (AFP) promoter region, resulting in reduced AFP expression and inhibiting liver cancer cell growth [27]. miRNA-21 can interact with the Programmed Cell Death 4 (PDCD4) promoter region in breast cancer cells, decreasing PDCD4 expression and increasing cell proliferation [28]. Further research will provide insights into the precise molecular mechanisms underlying NamiRNA-promoter interaction and the role in various diseases, contributing to the development of targeted therapeutics and diagnostic markers.
Enhancement of transcription by NamiRNA association with RNA pol II
NamiRNA have been shown to enhance transcription by associating with RNA pol II. This interaction occurs in the initiation phase of transcription, where NamiRNAs help recruit RNA pol II to specific gene promoter sites [7, 14, 15]. By doing so, NamiRNAs regulate gene expression by promoting targeted gene transcription or inhibiting non-targeted gene transcription initiation.
NamiRNA can enhance transcription in several ways. One strategy is to recruit RNA Pol II to the promoters of specific genes to enhance transcription. NamiRNAs can recruit or alter the activity of epigenetic modulators such as histone acetyltransferases (HATs) or histone methyltransferases to make the chromatin more conducive for transcription, shaping a more open chromatin structure to increase gene expression [29]. Some miRNAs can interact with transcriptional co-activators and enhance the assembly of transcriptional machinery at gene promoters. For example, miRNA-200c has been shown to recruit the transcriptional activator CREB1 to promote target genes expression in cell proliferation and differentiation [12]. Moreover, NamiRNAs can influence the release of paused RNA Pol II into productive elongation, a key regulatory step in gene expression.
An example of the transcription-enhancing role of miRNAs is the action of miR-132 [30]. miR-132 can regulate neuronal plasticity by affecting the transcription of certain neuronal genes. To be specific, miR-132 interacts with transcriptional co-activator p300 and is associated with acetylation of histones at the promoters of miR-132 target genes. By promoting histone acetylation via p300, miR-132 facilitates the recruitment of RNA Pol II to the promoter regions and enhance gene transcription. miR-26a can target the enhancer of zester homolog 2 (EZH2), an essential component of the Polycom repressive complex 2 (PRC2) that is involved in H3K27me3 [30]. By downregulating EZH2, miR-26a reduces H3K27me3 marks at the promoters of certain genes, counteracting repression and promoting transcriptional levels. These examples illustrate the complex interplay between NamiRNAs and transcription machinery. NamiRNAs can serve as fine-tuners of gene expression by inhibiting mRNA as well as enhancing gene transcription with multiple mechanisms.
Therapeutic potentials of NamiRNA in cancer
NamiRNAs represent a promising frontier in therapeutic interventions for a variety of cancers due to their fundamental roles in regulating oncogene expression. The ability of NamiRNAs to modulate key processes such as cell differentiation, proliferation, metabolism, and stress responses makes them attractive targets for drug development [15]. NamiRNAs can interact with multiple components of the gene expression machinery, including transcription factors and tumour suppressor genes (TSGs). This allows NamiRNAs to regulate the expression of numerous genes involved in diverse can cell processes.
In breast and ovarian cancer, the transcription factor myc oncogene is widely expressed and regulates biological processes such as cell cycle, proliferation and tumor progression [13, 31, 32]. Typically, the high myc, low p27 and high phospho-Rb level is associated with poor prognosis of breast and ovarian cancer [33]. Combining the screening data of miRNAs library of cell lines and cancer patients, researchers have discovered five miRNA candidates, miR-124, miR-365, miR-34b*, miR-18a and miR-506, can suppress tumor progression and reverse p27/myc/phospho-Rb expression. Among these miRNAs, miR-124 can bind to p27 promoter region to increase p27 protein expression and induce subsequent G1 arrest, which leads to phospho-Rb loss and myc protein decrease. In vivo studies demonstrated that nanoparticle-mediated delivery of miR-124 could reduce tumor growth and sensitize cells to etoposide, suggesting clinical applications of miRNAs as therapeutics to target myc functions on breast and ovarian tumor growth.
Another common feature of carcinogenesis is the dysfunction of TSGs [35, 36]. TSGs participate in cell cycle regulation, DNA damage repair and cell senescence, protecting normal cells from transforming into cancer cells [37,38,39]. The function loss and inactivation of TSGs have been recognized as common factors of carcinogenesis. It is generally believed that promoters DNA methylation and abnormal histone modifications could down regulate TSGs in non-small cell lung cancer (NSCLC). Researchers found that DNA methylation differences are rare in promoter, whereas miR-26A1 remarkably reactivates VILL promotor in lung cancer cells and this activation could be suppressed by enhancer inhibitor JQ1 [40]. miR-26A1 functions as a tumor suppressor and inhibits lung cancer cell proliferation and metastasis. The overexpression of miR-26A1 significantly enriches H3K27ac at the enhancer regions in A549 cells, suggesting that miR-26A1 can act as key regulator and potential therapeutic target for NSCLC. Recent research revealed that TSGs dysfunction is positively related with breast cancer development [41,42,43,44]. Liang et al. reported that miR-339 reactivates tumor suppressor genes (TSGs) by enhancing enhancer activity, offering a potential therapeutic approach for breast cancer treatment [34]. In normal breast cells, miR-339 binds to the enhancer region co-localized with Argonaute 2 (AGO2), activating the TSG GPER1, which prevents malignant transformation. However, reduced miR-339 expression diminishes enhancer activity, silencing GPER1 and promoting tumorigenesis (Fig. 2). Analysis of 157 breast cancer samples revealed suppressed expression of miR-339 and GPER1 in luminal A/B and triple-negative subtypes. miR-339 upregulates GPER1 via enhancer activation, but this process is abolished with AGO2 knockdown or enhancer deletion via CRISPR/Cas9. In vivo, miR-339 inhibits tumor proliferation, while GPER1 knockdown reverses this effect. Additionally, Ki67-positive cells decrease with miR-339 upregulation and increase with GPER1 loss. Together, miR-339 and GPER1 suppress breast cancer progression, with their disruption driving tumorigenesis.
The model illustrated that how tumor suppressor gene GPER1 is silenced in breast cancer [34]
Pancreatic cancer is one of lethal malignancies in the world with the 5-year survival rate lower than 6.9% [45, 46]. Patients are usually diagnosed at higher stages of pancreatic cancer for the lack of specific and effective biomarkers [47, 48]. miR-492 was found to be upregulated in a myriad of tumor tissue and may serve as a potential therapeutic biomarker [49,50,51,52,53]. The activation of miR-492 genes can significantly improve transforming growth factor-β (TGF-β)/Smad3 signaling pathway in tumor cells to promote epithelial mesenchymal transition of pancreatic cancer [54]. The enhancer marker H3K27ac is enriched in the NamiRNA miR-492 region, and the expression of its neighboring genes Nuclear Receptor Subfamily 2 Group C Member 1 (NR2C1), NADH: Ubiquinone Oxidoreductase Subunit A12 (NDUFA12) and Transmembrane And Coiled-Coil Domain Family 3 (TMCC3) are upregulated in pancreatic cancer. Further studies showed that miR-492, trigger of NamiRNA-enhancer network, can accelerate pancreatic cancer progression by activating neighboring genes (NR2C1/NDUFA12/TMCC3) and NR2C1-TGF-β/Smad3 pathway. In vivo studies showed that utilizing miR-492 inhibitor, antagomir-492, can inhibit tumor growth when compared with the control group. Hematoxylin and eosin (H&E) and immunohistochemistry (IHC) staining results are consistent with the quantitative analysis of NR2C1/NDUFA12/TMCC3 expression after antagomir-492 treatment, suggesting that miR-492 is as a promising NamiRNA target for pancreatic cancer diagnose and treatment.
NamiRNA is efficient in suppressing tumor growth via interrupting the interaction of cancer-associated fibroblasts (CAFs) and tumor cells. Tumor initiation, invasion and metastasis are affected by the cross-talking of CAFs and tumor cells than cancer cells alone [55, 56]. The abnormality of long noncoding RNAs (lncRNAs)-NamiRNA system can accelerate glucose metabolism of CAFs in tumor microenvironment and thus expediting cancer progression [57,58,59,60,61]. After applying bioinformatic and epigenomic RNA sequencing technology, lncRNA-NamiRNA profiles of normal fibroblasts (NFs) and CAFs are well studied in patients with oral squamous cell carcinoma (OSCC) [62]. To be specific, lncRNA H19 was analyzed to be the key lncRNA that upregulated in oral CAFs and OSCC cell lines synchronously. Knocking down of lncRNA H19 would affect oral CAF proliferation, migration and glycolysis. Moreover, lncRNA H19/miR-675-5p/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) axis was involved and can promote glycolysis pathway of oral CAFs with the application of luciferase reporter assay and miR-675-5p inhibitor. This study presents how lncRNA H19/miR-675-5p network affects glucose metabolism in oral CAFs, highlighting the potential value in developing a novel biomarker for diagnosis and promising remedy for OSCC.
NamiRNA can suppress tumor growth via intervening the Warburg effect and energy releasing of cancer cells [63]. In the Warburg effect of solid tumor, the energy releasing of cancer cells is predominantly regulated by aerobic glycolysis that consists of glucose uptake and glycolysis, followed by lactic acid fermentation [63,64,65]. During this biological process, folate-binding protein (FBP) acts as a suppressive enzyme to function in the gluconeogenesis of renal cell carcinoma (RCC) and is related to poor prognosis of RCC [66, 67]. The NamiRNA, miR-24-1, with DNA locus overlapping with enhancer region, is capable to reactivate FBP1 [68]. Researcher validated that miR-24-1 can upregulate FBP1 gene surprisingly and reactivate FBP1 expression to suppress Warburg effect, thus inhibiting the proliferation and metastasis of RCC cells. This reactivation requires the integrity of enhancer. Subsequent suppressive effects were further validated in 786-O and ACHN cell lines. miR-24-1 expression can suppress RCC tumor growth significantly in a xenograft model within 30 days. It is found that Ki-67 expression is decreased in miR-24-1 expressing group, FBP1 expression and Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) positive cells are increased with the overexpression of miR-24-1. This study highlights miR-24-1 overlapped with enhancer can activate TUNEL 1 and hinder Warburg effect of RCC progression, providing an alternative therapeutic strategy for RCC.
Conclusion and future prospect
NamiRNAs represent a fascinating and novel future of gene regulation for translational therapeutic potential. NamiRNAs have the capacity to upregulate gene expression and transcription, supplementing our understanding of inhibitory RNA interference pathway. Using NamiRNAs to target disease could provide a therapeutic means to modulate disease-associated genes and signal pathways directly from transcriptional level, offer a precision approach to cancer and immunity disease treatment.
Applying NamiRNAs technology in therapeutical areas may face some challenges in the future. The first challenge is how to ensure accurate delivery with high specificity. Typically, it is quite challenging to design effective delivery systems to transport NamiRNAs into cellular nucleus. NamiRNA carrier is supposed to traverse cell membrane and nuclear membrane with high specificity and avoid nonspecific genetic regulation. Meanwhile, introducing synthetic NamiRNAs in vivo may trigger unexpected immune response, inflammation and unwanted biological reactions. It is highly recommended to evaluate risks with patients before introducing NamiRNAs based therapies. Moreover, promoting NamiRNAs levels could bring in genes and pathways abnormal variations due to NamiRNA-gene system regulation. Finally, we have to take ethical concerns into consideration if long-term applications and potential abuse of NamiRNAs applied. Regulatory frameworks for NamiRNAs treatments are still in demand to address a wide variety of safety and ethical issues.
Future research will undoubtedly continue to illuminate the roles and functions of NamiRNAs in various biological processes and diseases. As our understanding of NamiRNAs goes deeper, NamiRNAs based therapies will be able to address specificity, delivery efficiency, and safety issues in clinic. The technology advancements will pave the way for a new generation of RNA-based therapeutics with the potential to change the landscape of disease treatment.
Data availability
No datasets were generated or analysed during the current study.
References
Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54.
Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G. The 21-nucleotide let-7 RNA regulates developmental timing in caenorhabditis elegans. Nature. 2000;403(6772):901–6.
Carrington JC, Ambros V. Role of MicroRNAs in plant and animal development. Science. 2003;301(5631):336–8.
Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.
Ha M, Kim VN. Regulation of MicroRNA biogenesis. Nat Rev Mol Cell Bio. 2014;15(8):509–24.
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. 2017;14(10):1326–34.
Suzuki HI, Young RA, Sharp PA. Super-Enhancer-Mediated RNA processing revealed by integrative MicroRNA network analysis. Cell. 2017;168(6):1000–e10141015.
Liu H, Lei C, He Q, Pan Z, Xiao D, Tao Y. Nuclear functions of mammalian MicroRNAs in gene regulation, immunity and cancer. Mol Cancer. 2018;17(1):64.
Li LC. Small RNA-Guided transcriptional gene activation (RNAa) in mammalian cells. Adv Exp Med Biol. 2017;983:1–20.
Tao BB, Liu XQ, Zhang W, Li S, Dong D, Xiao M, Zhong J. Evidence for the association of chromatin and MicroRNA regulation in the human genome. Oncotarget. 2017;8(41):70958–66.
Chen P, Guo X, Zhang L, Zhang W, Zhou Q, Tian Z, Zheng Y, Liao Q, Wang H, Li G, et al. MiR-200c is a cMyc-activated MiRNA that promotes nasopharyngeal carcinoma by downregulating PTEN. Oncotarget. 2017;8(3):5206–18.
Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458(7239):762–5.
Odame E, Chen Y, Zheng S, Dai D, Kyei B, Zhan S, Cao J, Guo J, Zhong T, Wang L, et al. Enhancer RNAs: transcriptional regulators and workmates of NamiRNAs in myogenesis. Cell Mol Biol Lett. 2021;26(1):4.
Liang Y, Zou Q, Yu W. Steering against wind: A new network of NamiRNAs and enhancers. Genomics Proteom Bioinf. 2017;15(5):331–7.
Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs MicroRNA nuclear import. Science. 2007;315(5808):97–100.
Hu X, Yin G, Zhang Y, Zhu L, Huang H, Lv K. Recent advances in the functional explorations of nuclear MicroRNAs. Front Immunol. 2023;14:1097491.
Luo J, Ji Y, Chen N, Song G, Zhou S, Niu X, Yu D. Nuclear miR-150 enhances hepatic lipid accumulation by targeting RNA transcripts overlapping the PLIN2 promoter. iScience. 2023;26(10):107837.
El Fatimy R, Zhang Y, Deforzh E, Ramadas M, Saravanan H, Wei Z, Rabinovsky R, Teplyuk NM, Uhlmann EJ, Krichevsky AM. A nuclear function for an oncogenic MicroRNA as a modulator of SnRNA and splicing. Mol Cancer. 2022;21(1):17.
Wong JJ, Ritchie W, Gao D, Lau KA, Gonzalez M, Choudhary A, Taft RJ, Rasko JE, Holst J. Identification of nuclear-enriched MiRNAs during mouse granulopoiesis. J Hematol Oncol. 2014;7:42.
Santovito D, Weber C. Non-canonical features of MicroRNAs: paradigms emerging from cardiovascular disease. Nat Rev Cardiol. 2022;19(9):620–38.
Iwasaki S, Kobayashi M, Yoda M, Sakaguchi Y, Katsuma S, Suzuki T, Tomari Y. Hsc70/Hsp90 chaperone machinery mediates ATP-dependent RISC loading of small RNA duplexes. Mol Cell. 2010;39(2):292–9.
Mao Z, Zhao H, Qin Y, Wei J, Sun J, Zhang W, Kang Y. Post-Transcriptional dysregulation of MicroRNA and alternative polyadenylation in colorectal cancer. Front Genet. 2020;11:64.
Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine EJ, Hanna J, Lodato MA, Frampton GM, Sharp PA, et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci USA. 2010;107(50):21931–6.
Jin Q, Yu LR, Wang L, Zhang Z, Kasper LH, Lee JE, Wang C, Brindle PK, Dent SY, Ge K. Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation. EMBO J. 2011;30(2):249–62.
Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, Haugen E, Sheffield NC, Stergachis AB, Wang H, Vernot B, et al. The accessible chromatin landscape of the human genome. Nature. 2012;489(7414):75–82.
Dai M, Li L, Qin X. Clinical value of miRNA-122 in the diagnosis and prognosis of various types of cancer. Oncol Lett. 2019;17(4):3919–29.
Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo Y-Y. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 2008;18(3):350–9.
Mortazavi D, Sohrabi B, Mosallaei M, Nariman-Saleh-Fam Z, Bastami M, Mansoori Y, Daraei A, Zununi Vahed S, Navid S, Saadatian Z et al. Epi-miRNAs: Regulators of the Histone Modification Machinery in Human Cancer. Journal of oncology 2022, 2022:4889807.
Xu K, Chen C, Wu Y, Wu M, Lin L. Advances in miR-132-Based biomarker and therapeutic potential in the cardiovascular system. Front Pharmacol. 2021;12:751487.
Evan G. Cancer. Taking a back door to target Myc. Science. 2012;335(6066):293–4.
Miller DM, Thomas SD, Islam A, Muench D, Sedoris K. c-Myc and cancer metabolism. Clin cancer Research: Official J Am Association Cancer Res. 2012;18(20):5546–53.
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. 2016;35(6):691–701.
Liang Y, Lu Q, Li W, Zhang D, Zhang F, Zou Q, Chen L, Tong Y, Liu M, Wang S, et al. Reactivation of tumour suppressor in breast cancer by enhancer switching through NamiRNA network. Nucleic Acids Res. 2021;49(15):8556–72.
Zhao M, Sun J, Zhao Z. TSGene: a web resource for tumor suppressor genes. Nucleic Acids Res. 2013;41(Database issue):D970–976.
Zhao M, Kim P, Mitra R, Zhao J, Zhao Z. TSGene 2.0: an updated literature-based knowledgebase for tumor suppressor genes. Nucleic Acids Res. 2016;44(D1):D1023–1031.
Aubrey BJ, Strasser A, Kelly GL. Tumor-Suppressor functions of the TP53 pathway. Cold Spring Harbor Perspect Med 2016, 6(5).
Hatzistergos KE, Williams AR, Dykxhoorn D, Bellio MA, Yu W, Hare JM. Tumor suppressors RB1 and CDKN2a cooperatively regulate Cell-Cycle progression and differentiation during cardiomyocyte development and repair. Circul Res. 2019;124(8):1184–97.
Zhao W, Wiese C, Kwon Y, Hromas R, Sung P. The BRCA tumor suppressor network in chromosome damage repair by homologous recombination. Annu Rev Biochem. 2019;88:221–45.
Li H, Da D, Yu W, Chen L, Yang S, Zhang B, Wang Y, Li L, Dang C. Tumor suppressor genes are reactivated by miR-26A1 via enhancer reprogramming in NSCLC. Hum Mol Genet. 2022;32(1):79–92.
Ignatov T, Weißenborn C, Poehlmann A, Lemke A, Semczuk A, Roessner A, Costa SD, Kalinski T, Ignatov A. GPER-1 expression decreases during breast cancer tumorigenesis. Cancer Invest. 2013;31(5):309–15.
Poola I, Abraham J, Liu A, Marshalleck JJ, Dewitty RL. The cell surface Estrogen receptor, G Protein- coupled receptor 30 (GPR30), is markedly down regulated during breast tumorigenesis. Breast Cancer: Basic Clin Res. 2008;1:65–78.
Weißenborn C, Ignatov T, Poehlmann A, Wege AK, Costa SD, Zenclussen AC, Ignatov A. GPER functions as a tumor suppressor in MCF-7 and SK-BR-3 breast cancer cells. J Cancer Res Clin Oncol. 2014;140(4):663–71.
Ariazi EA, Brailoiu E, Yerrum S, Shupp HA, Slifker MJ, Cunliffe HE, Black MA, Donato AL, Arterburn JB, Oprea TI, et al. The G protein-coupled receptor GPR30 inhibits proliferation of Estrogen receptor-positive breast cancer cells. Cancer Res. 2010;70(3):1184–94.
Sun H, Ma H, Hong G, Sun H, Wang J. Survival improvement in patients with pancreatic cancer by decade: a period analysis of the SEER database, 1981–2010. Sci Rep. 2014;4:6747.
Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R, Jemal A. Cancer treatment and survivorship statistics, 2016. Cancer J Clin. 2016;66(4):271–89.
Hartwig W, Werner J, Jäger D, Debus J, Büchler MW. Improvement of surgical results for pancreatic cancer. Lancet Oncol. 2013;14(11):e476–85.
Renz BW, Boeck S, Roeder F, Trumm C, Heinemann V, Werner J. Oligometastatic disease in pancreatic Cancer - How to proceed?? Visc Med. 2017;33(1):36–41.
Liang Y, Xu P, Zou Q, Luo H, Yu W. An epigenetic perspective on tumorigenesis: loss of cell identity, enhancer switching, and NamiRNA network. Sem Cancer Biol. 2022;83:596–604.
Shi LP, Liang M, Li FF, Li T, Lai DH, Xie QL, Yin YF, Liu YF. MiR-492 exerts tumor-promoting function in prostate cancer through repressing SOCS2 expression. Eur Rev Med Pharmacol Sci. 2019;23(3):992–1001.
Wu S, Xie J, Shi H, Wang ZW. miR-492 promotes chemoresistance to CDDP and metastasis by targeting inhibiting DNMT3B and induces stemness in gastric cancer. Biosci Rep 2020, 40(3).
von Frowein J, Hauck SM, Kappler R, Pagel P, Fleischmann KK, Magg T, Cairo S, Roscher A, von Schweinitz D, Schmid I. MiR-492 regulates metastatic properties of hepatoblastoma via CD44. Liver International: Official J Int Association Study Liver. 2018;38(7):1280–91.
Liu M, An J, Huang M, Wang L, Tu B, Song Y, Ma K, Wang Y, Wang S, Zhu H, et al. MicroRNA-492 overexpression involves in cell proliferation, migration, and radiotherapy response of cervical squamous cell carcinomas. Mol Carcinog. 2018;57(1):32–43.
Liu S, He X, Di Y, Li Q, Li F, Ma Y, Chen L, Gao Y, Xu J, Yang S, et al. NamiRNA-enhancer network of miR-492 activates the NR2C1-TGF-β/Smad3 pathway to promote epithelial-mesenchymal transition of pancreatic cancer. Carcinogenesis. 2023;44(2):153–65.
Bu L, Baba H, Yoshida N, Miyake K, Yasuda T, Uchihara T, Tan P, Ishimoto T. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38(25):4887–901.
Bertero T, Oldham WM, Grasset EM, Bourget I, Boulter E, Pisano S, Hofman P, Bellvert F, Meneguzzi G, Bulavin DV, et al. Tumor-Stroma mechanics coordinate amino acid availability to sustain tumor growth and malignancy. Cell Metabol. 2019;29(1):124–e140110.
Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43(6):904–14.
Liu H, Luo J, Luan S, He C, Li Z. Long non-coding RNAs involved in cancer metabolic reprogramming. Cell Mol Life Sci. 2019;76(3):495–504.
Liu X, Gan B. LncRNA NBR2 modulates cancer cell sensitivity to phenformin through GLUT1. Cell Cycle (Georgetown Tex). 2016;15(24):3471–81.
Song J, Wu X, Liu F, Li M, Sun Y, Wang Y, Wang C, Zhu K, Jia X, Wang B, et al. Long non-coding RNA PVT1 promotes Glycolysis and tumor progression by regulating miR-497/HK2 axis in osteosarcoma. Biochem Biophys Res Commun. 2017;490(2):217–24.
Peng F, Wang JH, Fan WJ, Meng YT, Li MM, Li TT, Cui B, Wang HF, Zhao Y, An F, et al. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene. 2018;37(8):1062–74.
Yang J, Shi X, Yang M, Luo J, Gao Q, Wang X, Wu Y, Tian Y, Wu F, Zhou H. Glycolysis reprogramming in cancer-associated fibroblasts promotes the growth of oral cancer through the LncRNA H19/miR-675-5p/PFKFB3 signaling pathway. Int J Oral Sci. 2021;13(1):12.
Liu X, Wang X, Zhang J, Lam EK, Shin VY, Cheng AS, Yu J, Chan FK, Sung JJ, Jin HC. Warburg effect revisited: an epigenetic link between Glycolysis and gastric carcinogenesis. Oncogene. 2010;29(3):442–50.
Shi T, Ma Y, Cao L, Zhan S, Xu Y, Fu F, Liu C, Zhang G, Wang Z, Wang R, et al. B7-H3 promotes aerobic Glycolysis and chemoresistance in colorectal cancer cells by regulating HK2. Cell Death Dis. 2019;10(4):308.
Cong J, Wang X, Zheng X, Wang D, Fu B, Sun R, Tian Z, Wei H. Dysfunction of natural killer cells by FBP1-Induced Inhibition of Glycolysis during lung cancer progression. Cell Metabol. 2018;28(2):243–e255245.
Yang J, Wang C, Zhao F, Luo X, Qin M, Arunachalam E, Ge Z, Wang N, Deng X, Jin G, et al. Loss of FBP1 facilitates aggressive features of hepatocellular carcinoma cells through the Warburg effect. Carcinogenesis. 2017;38(2):134–43.
Li B, Qiu B, Lee DS, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TP, Keith B, Nissim I, et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature. 2014;513(7517):251–5.
Ju D, Liang Y, Hou G, Zheng W, Zhang G, Dun X, Wei D, Yan F, Zhang L, Lai D, et al. FBP1 /miR-24-1/enhancer axis activation blocks renal cell carcinoma progression via Warburg effect. Front Oncol. 2022;12:928373.
Acknowledgements
Figure 1 was created with BioRender.com
Funding
This work was supported by Shenyang Science and Technology Project of China. (Grant number: 20-205-4-096).
Author information
Authors and Affiliations
Contributions
Xiang and Gang are responsible for literature study, conception and manuscript drafting. Jianping gave comments, revised the manuscript and provided funding for this work. All authors reviewed the manuscript and revised it critically before submission. All authors have seen and approved the final version of the manuscript and agreed to account for all aspects of the work.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Ren, X., Liu, G. & Zhou, J. Nuclear-activating miRNAs: unveiling the intricacies of subcellular miRNA function and regulation in cancer and immunity disease. Cancer Cell Int 25, 147 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03760-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03760-8