Open access peer-reviewed chapter

Multiple Genetic Polymorphisms within microRNA Targets and Homologous microRNA-Binding Sites: Two More Factors Influencing microRNA-Mediated Regulation of Gene Expression

Written By

Miruna Giurgiu, Robert Kaltenbach, Franziska Ahrend, Summer Weeks, Holly Clifton, Martin Bouldo, Vitaly Voloshin, Jiling Zhong, Siegfried Harden and Alexander Kofman

Submitted: 24 June 2023 Reviewed: 28 June 2023 Published: 14 August 2023

DOI: 10.5772/intechopen.1002250

From the Edited Volume

Advances in Genetic Polymorphisms

Nouha Bouayed Abdelmoula and Balkiss Abdelmoula

Chapter metrics overview

139 Chapter Downloads

View Full Metrics

Abstract

miRNA-mRNA interaction depends on multiple factors such as 3’UTR isoforms, the cell and tissue-specific expression levels of RNA-binding proteins, the sequence context around the mRNA target site, and other mechanisms. Genetic polymorphisms within miRNAs and their target sites appear to be among the most important ones because they influence the mode and outcome of miRNA-mRNA interaction universally and irreversibly. SNP disruption of miRNAs and their binding sites, as well as conformational changes preventing the access of the miRNA to its target site, are adopted as the most credible mechanistic explanations of SNP-mediated effects. The occurrence of multiple SNPs within the same miRNA-binding site implies their combinatorial mode of action. The presence of the repetitive (homologous) binding sites for the same miRNA on its mRNA target may both enhance the miRNA targeting and provide for the backup target site instead of the one disrupted by SNP, thus rescuing the miRNA functionality. While being underexplored, the multiple genetic polymorphisms within the miRNA-binding sites, as well as homologous miRNA-binding sites, may be considered as additional factors influencing miRNA-mediated regulation of gene expression.

Keywords

  • microRNA
  • mRNA
  • target
  • single-nucleotide polymorphism
  • homologous microRNA-binding sites

1. Introduction

Since the beginning of the 2000s, miRNAs stay in the focus of every aspect of medical and biomedical research [1, 2]. miRNAs are involved in a wide range of biological processes such as cell differentiation [3, 4, 5, 6, 7] and reprogramming [8], cellular senescence [9] and cell death [10, 11, 12, 13, 14, 15, 16], tissue and organ development [5, 17, 18, 19, 20] and regeneration [21, 22, 23, 24, 25, 26], cell signaling [27, 28], oxidative stress and metabolism [3, 29, 30, 31, 32, 33, 34, 35, 36, 37], mitochondrial dysfunctions [22, 38, 39], hormonal regulation [16, 40, 41] and adaptive responses [17], brain function [42, 43], inflammation [31, 44, 45, 46], immune response and the effects of the microbiome [46, 47, 48, 49, 50, 51, 52, 53], viral infections and latency [43, 54, 55, 56, 57, 58, 59, 60, 61], DNA damage and repair [22, 62, 63, 64, 65, 66, 67], genomic balance [68] and genomic instability [65, 69], etc.

miRNAs deregulation promotes the biological processes resulting in various human diseases [41, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84] including oncogenesis [6, 7, 35, 37, 54, 59, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120]. miRNAs are recognized as important factors that are involved in tumor cell invasiveness [121], metastases [90, 121, 122, 123, 124, 125, 126, 127, 128, 129], cancer cachexia [130], drug resistance [131, 132, 133, 134, 135, 136], sensitivity to chemotherapy [65, 137, 138], and radiosensitivity [138, 139, 140]. miRNAs are actively explored as both therapeutic agents [32, 112, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164] and targets [28, 42, 161, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174]. A growing number of reports point to the usefulness of miRNAs, and specifically circulating miRNAs [175, 176] as diagnostic and prognostic biomarkers [51, 70, 71, 73, 110, 112, 155, 160, 161, 174, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223] related to clinical and forensic [177, 224] studies, as well as normal physiological conditions, such as, diet [225] and increased physical activity [226, 227].

Deep sequencing and computational approaches indicate that miRNA genes comprise about 0.5–1% of the predicted genes in animals and humans [228]. One mRNA can be targeted by hundreds of miRNAs [84, 229], and a single miRNA can potentially recognize hundreds of different target transcripts [229, 230], which often share pathways to ensure their impact [52]. Therefore, miRNAs may be considered as global regulators of gene expression [231] with the vast targetome [52, 174] regulating about 90% of all protein-coding genes [232, 233, 234] and interactome-complex networking and cooperation between themselves [235, 236], as well as other regulatory molecules, in particular, transcription factors and noncoding RNAs [237, 238, 239].

Identification and characterization of the factors influencing miRNA functionality are essential to elucidate the mechanisms of miRNA activity and to explain and predict the effects of miRNAs for clinical applications. Genetic variations of both miRNAs and their target sites may have a significant impact on the efficacy of miRNA targeting. It is growingly recognized that naturally occurring variations in miRNAs and their target genes contribute to phenotypic complexity and may be associated with human pathologies [240, 241], including cancer [231, 242, 243, 244]. We present information about two underexplored SNP-related mechanisms influencing miRNA-mRNA interaction: the multiple SNPs within the single miRNA target site and the multiple (homologous) target sites for the same miRNA.

Advertisement

2. miRNA properties and biogenesis

miRNAs belong to the category of small noncoding RNA molecules [2, 245] ranging between 19 and 24 nucleotides in length [229]. miRNAs have been detected in all biological species, including viruses [55, 56, 57, 58, 59, 60, 246]. Of note, bacteria do have short RNA sequences with the miRNA-similar functions, yet they are not considered as true miRNAs [247]. Bioinformatics analysis indicates the existence of miRNAs derived from transposable elements [248, 249]. Although miRNAs are usually located in the cytoplasm, several studies have detected a regulatory role of miRNAs in other cell compartments such as mitochondria (mitomiRs), nucleus, endoplasmic reticulum, granules, and P-bodies [38, 232]. Nuclear miRNAs participate in the regulation of miRNA biogenesis and function [250].

Circulating miRNA may be released by different types of cells and delivered into recipient cells for functional purposes, acting as cell-to-cell signaling mediators [251]. miRNA may be freed from dying cells or selectively sorted into the secreted small vesicles called exosomes [158, 229, 252, 253, 254, 255]. Such exosomal miRNAs are regarded as promising clinical biomarkers (see above). miRNA can undergo both vertical and horizontal transmissions among distinct species, remarkably through feeding between plants and animals [229, 245].

The biogenesis of canonical animal miRNAs starts with the transcription of the pri-miRNA by RNA Pol II [256, 257]. The pri-miRNA has the length of about several thousands of nucleotides [258], the 7-methylguanosine, and 3′- poly(A) tail [259]. The pri-miRNA forms a stem-loop structure, which is recognized and cleaved by Drosha (endonuclease RNAse III) and two molecules of its partner protein, DGCR8 (DiGeorge syndrome critical region 8, named Pasha in flies and nematodes), which results in the formation of pre-miRNA. The pre-miRNA is further processed by the Dicer (another endonuclease RNAse III). The Dicer’s partner protein TRBP (transactivation response element RNA-binding protein) is essential for miRNA processing in flies. The resulting short RNA duplex is associated with one of the AGO proteins to form the RISC, which is essential in destabilizing the target mRNA molecule or inhibiting its translation (for detailed review of miRNA biogenesis see [229, 260, 261, 262, 263]). Noncanonical miRNAs are produced without Drosha- or Dicer-catalyzed cleavage [84, 261, 264]. Each step of miRNA biogenesis is tightly regulated [260, 263]. Like all RNA molecules, miRNAs undergo posttranscriptional modifications, which may affect their properties [234, 260, 262, 265, 266]. Recent studies point to the existence of sequence variants in miRNAs called isomiRs, which differ from the annotated miRNAs by altered sequences due to the various posttranscriptional modifications [267, 268].

The core miRNA function is posttranscriptional regulation of the expression of targeted genes. It is achieved through miRNA-mediated RNA silencing [269]. miRNAs, as a part of the RISC, trigger the various forms of the translational repression of the target mRNA via blocking its ribosomal loading and initiation of translation, inhibiting translation elongation, and causing protein degradation [234, 261, 262]. It may also cause mRNA deadenylation and decay [234], which is typical for plants, whereas translational repression is more characteristic for animal cells [261, 270, 271]. In contrary to mRNA degradation, translational repression is reversible and can be employed for rapid response to internal or external cues [271].

While target repression is universally recognized as the dominant mode of miRNA action, some reports indicate that miRNAs can stabilize the targeted transcript [234, 272, 273, 274, 275, 276]. MiRNAs can also regulate gene expression both indirectly by targeting the mRNAs of transcription factors and directly after being transported back to the nucleus and binding to the complementary sequence in the promoter [232, 234, 250, 262, 276]. miRNAs can also perform other noncanonical functions acting as ligands for toll-like receptors [277].

Advertisement

3. MiRNA target recognition

miRNAs bind to the complementary sequences of the host mRNA as a part of the RISC with its core component Ago proteins [229]. In plants, miRNAs show perfect or near-perfect complementarity to their target mRNA. In mammalian cells, as well as in other eukaryotes, miRNAs bind to mRNA sequences with imperfect complementarity [229, 278]. The 5′-end of miRNA exhibits the so-called seed sequence (Figure 1), which is complementary to six or more nucleotides at the 3’UTR of mRNA [2, 233, 262, 279].

Figure 1.

miRNA-mRNA interaction in animal cells (developed by F. Ahrend).

Canonical miRNA-binding sites are positioned in the 3’UTR of the targeted mRNA sequence and classified upon the extent and location of matching miRNA nucleotides: 6mers perfectly pair to nucleotides 2–7 on the 5′-end of miRNA, 7-merA1 and 7mer8—additional pairing with miRNA nucleotide 1 or 8 respectively, and 8mer sites match miRNA nucleotides 1–8 [261]. The efficacy of these sites is usually augmented by additional pairing at the 3′ end of miRNAs. In addition to the binding site at the miRNA 5′ end, pairing around the binding sites contributes to the targeting efficacy [233, 261, 280]. The so-called 3′-supplementary sites with atypical elaboration of the 6mer, 7mer, and 8mer sites (≥3–4 pairs), and 3′-compensatory sites stretching for more than 4–5 pairs, while the seed region is mismatched are called atypical sites [84, 233, 261]. The supplementary pairing is believed to enhance the miRNA-mRNA interaction with the greater specificity. However, in animal cells, the high complementarity beyond the seed may cause TDMD [281], the common mechanism of destabilization of the short RNA molecules including miRNAs, which is observed in many diseases [84]. In general, the miRNA-mRNA complementarity with nucleotides at 3′ end or with nucleotides in the center (centered pairing) in the absence of the perfectly matched seed nucleotides is believed to be much less effective in regulating gene expression [84, 233, 261, 282, 283, 284]. The noncanonical seed-matching sites may also be in the protein-coding regions of the target mRNAs and even in the 5’UTR mRNA sequence [84, 232, 233, 285, 286].

Advertisement

4. Factors influencing miRNA targeting

miRNA-mRNA interaction depends on a range of factors. The posttranscriptional modification resulting in miRNA methylation causes structural changes that affect AGO binding [232]. miRNA sponges [287, 288, 289] bind miRNAs, thus preventing them from interacting with mRNA targets. miRNAs competition and cross talk with RNA-binding proteins [84, 232, 233, 236, 270], whose expression levels may depend on the cellular context and vary between the tissues [261, 290], also add up to the spectrum of various regulatory outcomes beside the dominating mechanism of miRNA targeting with moderate target repression [84]. miRNA targeting is known to depend on 3’UTR isoforms: 3’UTR size, alternative cleavage, and the location of target sequences within AU-rich regions [233, 261, 282, 284, 290, 291, 292].

The extent of complementary identity between miRNA and its mRNA targets appears to be one of the crucial factors, which determine the mode of target repression. The target cleavage by RISC is typical for plants, where the pairwise alignment is characterized by high complementarity. In opposite, the limited seed matching in animals often leads to translational inhibition [270]. It has been suggested that the above-mentioned noncanonical stabilization of the targeted transcript by miRNAs may depend on the complementarity between the miRNA and its target mRNA [275]. On the other hand, the extensive base-pairing may also cause TDMD [84, 293]. In this regard, genetic polymorphisms within miRNAs and their target sites appear to be among the most important mechanisms that ubiquitously and irreversibly define the mode and outcome of miRNA-mRNA interaction.

Advertisement

5. miRNAs and genetic polymorphisms

SNPs are genetic variations (nucleotide substitutions and indels) in DNA sequence [294, 295]. SNPs within the mRNA noncoding regions do not change the protein sequence, but they may significantly alter gene expression by affecting transcription, RNA processing, translation, and interaction of mRNA with noncoding regulatory RNAs [296]. While most SNPs are likely to be functionally neutral, some of them may represent causative links with human diseases [297]. SNPs in miRNA-precursor flanking regions, promoters of miRNA-encoding genes, and in the genes involved in miRNA biogenesis (transcription and RNA processing) may result in higher or lower miRNA expression profiles [298, 299, 300, 301, 302], which, in turn, represent the unique disease-specific signatures that can identify cancer types [231240243303]. SNPs within mature miRNA sequences, specifically in their seed regions, dissimilarities in mature miRNAs due to variable cleavage sites for Drosha and Dicer [243, 301, 302, 304], and polymorphisms within or nearby miRNA-binding sites on genes targets, may enhance or weaken the interaction between miRNA and its target transcripts and contribute to phenotypic variations and disease susceptibility [230240241, 267, 294, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321].

Within the mRNA targets, SNPs can either create or destroy miRNA-binding sites. It may impair miRNA ability to target oncogenes and, in opposite, render tumor-suppressor genes susceptible to miRNA-mediated inhibition (Figure 2). SNPs located both within and beyond the seed-target regions [322] can affect miRNA-mRNA interaction by destabilizing the mRNA target molecule, slowing down its ability for ribosomal loading, and altering the secondary RNA structure, which in turn, may have an impact on the availability of miRNA-binding sites [284, 290, 323]. Disruption or creation of miRNA-binding sites is recognized as one of the most credible mechanistic explanations of SNP-mediated effects [240, 241].

Figure 2.

The hypothetical effects of SNP on miRNA-mRNA interaction and cancer development. 1a—miRNA suppresses the oncogenic mRNA, 1b—SNP disrupts the target site resulting in oncogene overexpression, 2a—miRNA does not control the expression of the tumor suppressor gene, and 2b—SNP creates the miRNA-binding site resulting in the inhibition of the tumor suppressor gene expression (developed by H. Clifton and F. Ahrend).

Advertisement

6. Multiple SNPs within miRNA-binding sites

A considerable number of genome-wide association studies demonstrated links between the SNPs within the miRNA target sites and predisposition to various human diseases. The attention was mostly devoted to SNPs, whose population frequency was well above 1%. Still, miRNA target sites may harbor multiple SNPs, although most of them are of low population frequency [324]. Even though the probability of their coincidental occurrence within the site is low, the frequency of some SNPs may vary significantly due to their ethnic disparities [325]. It has been suggested that, at least for some genes, it is not a separately taken SNP, but the combinatorial effect of several SNPs that may determine the outcome and efficacy of miRNA-mediated regulation of gene expression [326]. The location of SNPs (seed-corresponding region, centered, or 3′ end position) should also be taken into consideration.

Thus, the presence of additional, even rare (low frequency) SNPs within miRNA target sites, may modify (weaken or enhance) the effects of the SNPs, which occur at higher frequencies. Consequently, multiple SNPs may increase the probability of the site disruption, but may be also neutral, and even enhancing (Figure 3). This situation is less typical for the protein-coding regions due to the selection pressure resulting in the fewer SNPs. In the case of the overlapping miRNA-binding sites, the outcome is less clear due to the co-targeting effect—additive repression by more than one miRNA [84, 232]. Beside this, the increased complementarity between miRNA and its target in animals may result in miRNA decay (see above about TDMD).

Figure 3.

The examples of disrupting, enhancing, and neutral SNPs within a single target site. The image shows the fragment of the human PDGFRA DNA sequence corresponding to the mRNA 3’UTR region, the miRNA-10a-5P binding site. 1—Enhancing through the formation of the G·U wobble base pair (rs1302330738), 2—Neutral switch from the G·U wobble base pair to the classical Watson-Crick G·C bond (rs1227726044), 3—Neutral switch from the G·U wobble base pair to the classical Watson-Crick A-T bond, 4Neutral switch from the classical Watson-Crick G·C bond to the G·U wobble base pair (rs1322316972), 5—Disrupting (rs1724902856, rs1724902491, rs1314937650), and 6—Enhancing through the formation of the G·U wobble base pair (rs918309834). Note: The G·U wobble base pair has comparable thermodynamic stability to that of Watson-Crick base pairs and is known as a fundamental unit of RNA secondary structure in every class of RNA in organisms of all three phylogenetic domains [327]. However, it is still under the discussion whether G·U is characteristic for the canonical mRNA seed match [328] (developed by M. Giurgiu).

SNPs within the single target site may be independent (Figure 4) and mutually exclusive if they are overlapping (Figure 5). Of note, SNP frequencies in the databases reflect the experimental NextGen sequencing data, and the possible SNP overlap is not taken into consideration. If mutually exclusive SNPs are represented by the overlapping indel and nucleotide substitution, the latter one is “shadowed” by the indel (Figure 5), and its real population frequency may be higher than that presented in the database.

Figure 4.

The fragment of the human PDGFRA DNA sequence corresponding to the mRNA 3’UTR region, the miRNA-212-3P binding site. SNPs rs1577756917, rs1387790378, rs984854314, rs1560496903, and rs1454435576 represent independent events (developed by M. Giurgiu).

Figure 5.

The fragment of the human PDGFRA DNA sequence corresponding to the mRNA 3’UTR region, the miRNA-4444 binding site. SNPs rs1724893276 and rs1413062472, as well as rs1724893401, represent mutually exclusive events. rs1724893276 is an indel, and in the case of the deletion, neither rs1413062472 nor s172489 3401 (nucleotide substitutions) will be present (developed by M. Giurgiu).

Advertisement

7. Homologous miRNA-binding sites

It has been reported that some mRNAs may have more than one binding (seed-matching) site for the miRNAs [329, 330, 331, 332, 333, 334, 335, 336]. These sites have the analogous seed-binding motif, but the different nucleotide content outside (Figure 6). Such multiple sites are proposed to act synergistically [330, 333, 334, 335, 337, 338], which, in turn, may depend on the distance between the sites [330, 338], as well as the activity and structural variations of miRNA-Ago complexes [336, 339]. It would be rational to suggest that if SNPs disrupt one of the miRNA-binding sites, the other sites for the same miRNA could preserve the miRNA-mediated control of gene expression. Such homologous miRNA-binding sites are present in almost all human genes, and their numbers are significant and directly proportional to the length of the 3’UTR [340].

Figure 6.

The repeated (homologous) target sites for miRNA-X (developed by A. Kofman).

Advertisement

8. Bioinformatic tools

Whereas miRNAs and SNPs are both subjects of intense studies, there are few resources that allow to investigate the potential effects of SNPs on miRNA functionality.

PolymiRTS (Polymorphism in miRNAs and their Target Sites) is a database of naturally occurring DNA variations in miRNAs seed regions and miRNAs target genes [341]. PolymiRTS database was created by scanning 3’UTRs of mRNAs in humans and mice for SNPs in miRNA target sites. The database contains the results of genome-wide association studies about the links between the specific SNPs and human diseases. The PolymiRTS database also includes polymorphisms in target sites that have been supported by a variety of experimental methods and polymorphisms in miRNA seed regions.

MiRSNP is another database of human polymorphisms altering miRNA-binding sites [342]. It is a collection of 414,510 SNPs that are predicted to be able to affect (decrease/break or enhance/create) miRNA-mRNA binding.

MiRdSNP [343] is a database supplemented with the tools allowing the proximity searches between miRNA target sites and disease-associated SNPs (dSNPs) by gene name, miRbase ID, target prediction algorithm, disease, and any nucleotide distance between dSNPs and miRNA target sites. The web interface displays detailed sequence views showing the relationship among dSNPs, miRNA target sites, and SNPs. An interactive visualization tool shows the chromosomal distribution of dSNPs, miRNA target sites from TargetScan, and SNPs. The limitation of MiRdSNP is that the database contains only manually curated dSNPs on the 3’UTRs of human genes from available publications in PubMed, and the SNPs are only those that are disease-associated.

MSSD [344] is another manually curated database that provides comprehensive experimentally supported associations among miRNAs, SNPs, and human diseases. It has the same limitations as MiRdSNP.

miRNAhrend (https://franziskaahrend.pythonanywhere.com/) is the online-available package (beta version), which allows users to predict miRNA-target sites in any transcript upon the presence of seed-matching 6mers, which is verified by the complementarity-based alignment. The tool allows to assess how the presence of SNPs changes (“disappear/emerge”) the landscape of the target sites and their corresponding miRNAs, identify the homologous miRNA-binding sites, and track down the exact position of the sites within any studied RNA sequence.

Advertisement

9. Conclusions

The ability of miRNAs to regulate the variety of genes in all tissues, as well as their presence in all biological species and their promise for clinical applications as novel diagnostics and therapeutics prompt further exploration of the factors influencing miRNA activity and functions. It is possible to assume that the presence of SNPs within miRNAs and their binding sites represents a powerful mechanism that can influence all biological processes in which miRNAs are involved. However, the growing number of reports indicates that the presence of SNPs cannot be interpreted as unalterable situation defining the biological outcome of miRNA targeting. The combinatorial effect of multiple SNPs located within the same target site, as well as beyond the target site, yet within the region where additional complementary matches between miRNA and its target can be formed, the circumstances where some SNPs may be mutually exclusive, and the homologous sites, which hypothetically can be utilized by miRNAs as an alternative target if the other sites are disrupted or become inaccessible, all these additional factors related to miRNA functionalities are anticipated of being studied with the development of the appropriate biocomputing tools and mathematical models.

Advertisement

Abbreviations

miRNAmicroRNA
mRNAmessenger RNA
SNPsingle-nucleotide polymorphism
Polpolymerase
pri-miRNAprimary miRNA transcript
pre-miRNAprecursor miRNA
AGOargonaute proteins
RISCRNA-induced silencing complex
UTRuntranslated region
3′3-prime
5′5-prime
Indelinsertion/deletion
PDGFRAplatelet-derived growth factor receptor alpha
TDMDtarget-directed miRNA degradation

References

  1. 1. Arteaga-Vazquez M, Caballero-Perez J, Vielle-Calzada JP. A family of microRNAs present in plants and animals. Plant Cell. 2006;18(12):3355-3369
  2. 2. Ambros V. The functions of animal microRNAs. Nature. 2004;431(7006):350-355
  3. 3. Singh V. Intracellular metabolic reprogramming mediated by micro-RNAs in differentiating and proliferating cells under non-diseased conditions. Molecular Biology Reports. 2021;48(12):8123-8140
  4. 4. Galagali H, Kim JK. The multifaceted roles of microRNAs in differentiation. Current Opinion in Cell Biology. 2020;67:118-140
  5. 5. Zare A et al. Epigenetic modification factors and microRNAs network associated with differentiation of embryonic stem cells and induced pluripotent stem cells toward cardiomyocytes: A Review. Life (Basel). 2023;13(2):569. DOI: 10.3390/life13020569
  6. 6. Souza OF, Popi AF. Role of microRNAs in B-cell compartment: Development, proliferation and Hematological diseases. Biomedicines. 2022;10(8):2004. DOI: 10.3390/biomedicines10082004
  7. 7. Katsaraki K et al. MicroRNAs: Tiny regulators of gene expression with pivotal roles in Normal B-cell development and B-cell chronic lymphocytic Leukemia. Cancers (Basel). 2021;13(4):593. DOI: 10.3390/cancers13040593
  8. 8. Pascale E et al. MicroRNA roles in cell reprogramming mechanisms. Cells. 2022;11(6):940. DOI: 10.3390/cells11060940
  9. 9. Lettieri-Barbato D et al. MicroRNAs, long non-coding RNAs, and circular RNAs in the redox control of cell senescence. Antioxidants (Basel). 2022;11(3):480. DOI: 10.3390/antiox11030480
  10. 10. Ferris WF. The role and interactions of programmed cell death 4 and its regulation by microRNA in transformed cells of the gastrointestinal tract. Frontiers in Oncology. 2022;12:903374
  11. 11. Lamberti MJ et al. Damage-associated molecular patterns modulation by microRNA: Relevance on immunogenic cell death and cancer treatment outcome. Cancers (Basel). 2021;13(11):2566. DOI: 10.3390/cancers13112566
  12. 12. Palumbo S et al. Emerging roles of microRNA in modulating cell-death processes in malignant glioma. Journal of Cellular Physiology. 2014;229(3):277-286
  13. 13. Chen CH, Guo M, Hay BA. Identifying microRNA regulators of cell death in drosophila. Methods in Molecular Biology. 2006;342:229-240
  14. 14. Bai X et al. Role of microRNA-34b-5p in cancer and injury: How does it work? Cancer Cell International. 2022;22(1):381
  15. 15. Azam INA et al. Roles of microRNAs in regulating apoptosis in the pathogenesis of endometriosis. Life (Basel). 2022;12(9):1321. DOI: 10.3390/life12091321
  16. 16. Matarrese P et al. The sex-related interplay between TME and cancer: On the critical role of Estrogen, MicroRNAs and autophagy. Cancers (Basel). 2021;13(13):3287. DOI: 10.3390/cancers13133287
  17. 17. DeVeale B, Swindlehurst-Chan J, Blelloch R. The roles of microRNAs in mouse development. Nature Reviews Genetics. 2021;22(5):307-323
  18. 18. Raza SHA et al. The role of MicroRNAs in muscle tissue development in beef cattle. Genes (Basel). 2020;11(3):295. DOI: 10.3390/genes11030295
  19. 19. Boshtam M et al. Crosstalk of transcriptional regulators of adaptive immune system and microRNAs: An insight into differentiation and development. Cells. 2023;12(4):635. DOI: 10.3390/cells12040635
  20. 20. Zolboot N et al. MicroRNAs instruct and maintain cell type diversity in the nervous system. Frontiers in Molecular Neuroscience. 2021;14:646072
  21. 21. Mahtal N et al. MicroRNAs in kidney injury and disease. Nature Reviews Nephrology. 2022;18(10):643-662
  22. 22. Rodrigues SC, Cardoso RMS, Duarte FV. Mitochondrial microRNAs: A putative role in tissue regeneration. Biology (Basel). 2021;9(12):486. DOI: 10.3390/biology9120486
  23. 23. Cione E et al. Liver damage and microRNAs: An update. Current Issues in Molecular Biology. 2022;45(1):78-91
  24. 24. Maries L et al. MicroRNAs-the heart of post-myocardial infarction Remodeling. Diagnostics (Basel). 2021;11(9):1675. DOI: 10.3390/diagnostics11091675
  25. 25. Theofilis P et al. The impact of proangiogenic microRNA modulation on blood flow recovery following hind limb ischemia. A systematic review and meta-analysis of animal studies. Vascular Pharmacology. 2021;141:106906
  26. 26. Tanase DM et al. Current knowledge of MicroRNAs (miRNAs) in acute coronary syndrome (ACS): ST-elevation myocardial infarction (STEMI). Life (Basel). 2021;11(10):1057. DOI: 10.3390/life11101057
  27. 27. Vaghf A et al. The role of microRNAs in diseases and related signaling pathways. Molecular Biology Reports. 2022;49(7):6789-6801
  28. 28. Humphries BA, Wang Z, Yang C. MicroRNA regulation of the small rho GTPase regulators-complexities and opportunities in targeting cancer metastasis. Cancers (Basel). 2020;12(5):1092. DOI: 10.3390/cancers12051092
  29. 29. Tarlton JMR, Patterson S, Graham A. MicroRNA sequences modulated by Beta cell lipid metabolism: Implications for type 2 diabetes mellitus. Biology (Basel). 2021;10(6):534. DOI: 10.3390/biology10060534
  30. 30. Wallace SR, Pagano PJ, Kracun D. MicroRNAs in the regulation of NADPH oxidases in vascular diabetic and ischemic pathologies: A case for alternate inhibitory strategies? Antioxidants (Basel). 2023;12(1):70. DOI: 10.3390/antiox12010070
  31. 31. Kolodziej F et al. MicroRNAs as the sentinels of redox and hypertrophic signalling. International Journal of Molecular Sciences. 2022;23(23):14716. DOI: 10.3390/ijms232314716
  32. 32. Gibson MS, Noronha-Estima C, Gama-Carvalho M. Therapeutic metabolic reprograming using microRNAs: From cancer to HIV infection. Genes (Basel). 2022;13(2):273. DOI: 10.3390/genes13020273
  33. 33. Virga F et al. MicroRNA-mediated metabolic shaping of the tumor microenvironment. Cancers (Basel). 2021;13(1):127. DOI: 10.3390/cancers13010127
  34. 34. Vezza T et al. MicroRNAs and oxidative stress: An intriguing crosstalk to Be exploited in the Management of Type 2 diabetes. Antioxidants (Basel). 2021;10(5):802. DOI: 10.3390/antiox10050802
  35. 35. Shiah SG, Chou ST, Chang JY. MicroRNAs: Their role in metabolism, tumor microenvironment, and therapeutic implications in head and neck squamous cell carcinoma. Cancers (Basel). 2021;13(22):5604. DOI: 10.3390/cancers13225604
  36. 36. Azizi M, Othman I, Naidu R. The role of MicroRNAs in lung cancer metabolism. Cancers (Basel). 2021;13(7):1716. DOI: 10.3390/cancers13071716
  37. 37. Wai Hon K et al. Insights into the role of microRNAs in colorectal cancer (CRC). Metabolism. Cancers (Basel). 2020;12(9):2462. DOI: 10.3390/cancers12092462
  38. 38. Ortega MA et al. The regulatory role of mitochondrial MicroRNAs (MitomiRs) in breast cancer: Translational implications present and future. Cancers (Basel). 2020;12(9):2443. DOI: 10.3390/cancers12092443
  39. 39. Tan WL et al. An insight into the associations between microRNA expression and mitochondrial functions in cancer cell and cancer stem cell. Molecular Biology Reports. 2023;50(6):5395-5405
  40. 40. Macias S, Michlewski G, Cáceres JF. Hormonal regulation of MicroRNA biogenesis. Molecular Cell. 2009;36(2):172-173
  41. 41. Kim IK et al. The role of Epicardial adipose tissue-derived MicroRNAs in the regulation of cardiovascular disease: A narrative review. Biology (Basel). 2023;12(4):498. DOI: 10.3390/biology12040498
  42. 42. Brennan GP, Henshall DC. MicroRNAs as regulators of brain function and targets for treatment of epilepsy. Nature Reviews Neurology. 2020;16(9):506-519
  43. 43. Cao DD, Li L, Chan WY. MicroRNAs: Key regulators in the central nervous system and their implication in neurological diseases. International Journal of Molecular Sciences. 2016;17(6):842. DOI: 10.3390/ijms17060842
  44. 44. Brites D. Regulatory function of microRNAs in microglia. Glia. 2020;68(8):1631-1642
  45. 45. Lee A, Kim SN. Serum MicroRNA on inflammation: A literature review of mouse model studies. Biomarkers. 2020;25(7):513-524
  46. 46. Maranini B et al. microRNAs and inflammatory immune response in SARS-CoV-2 infection: A narrative review. Life (Basel). 2022;12(2):288. DOI: 10.3390/life12020288
  47. 47. Bi K et al. MicroRNAs regulate intestinal immunity and gut microbiota for gastrointestinal health: A comprehensive review. Genes (Basel). 2020;11(9):1075. DOI: 10.3390/genes11091075
  48. 48. Allegra A et al. Interactions between the MicroRNAs and microbiota in cancer development: Roles and therapeutic opportunities. Cancers (Basel). 2020;12(4):805. DOI: 10.3390/cancers12040805
  49. 49. Zhou H et al. MicroRNAs with multiple targets of immune checkpoints, as a potential sensitizer for immune checkpoint inhibitors in breast cancer treatment. Cancers (Basel). 2023;15(3):824. DOI: 10.3390/cancers15030824
  50. 50. Mourenza A et al. Understanding microRNAs in the context of infection to find new treatments against human bacterial pathogens. Antibiotics (Basel). 2022;11(3):356. DOI: 10.3390/antibiotics11030356
  51. 51. Sarshar M et al. Fecal microRNAs as innovative biomarkers of intestinal diseases and effective players in host-microbiome interactions. Cancers (Basel). 2020;12(8):2174. DOI: 10.3390/cancers12082174
  52. 52. Cho S, Tai JW, Lu LF. MicroRNAs and their Targetomes in tumor-immune communication. Cancers (Basel). 2020;12(8):2025. DOI: 10.3390/cancers12082025
  53. 53. De Silva S, Tennekoon KH, Karunanayake EH. Interaction of gut microbiome and host microRNAs with the occurrence of colorectal and breast cancer and their impact on patient immunity. Oncotargets and Therapy. 2021;14:5115-5129
  54. 54. Kofman AV et al. The p53-microRNA-34a axis regulates cellular entry receptors for tumor-associated human herpes viruses. Medical Hypotheses. 2013;81(1):62-67
  55. 55. Cullen BR. Viral and cellular messenger RNA targets of viral microRNAs. Nature. 2009;457(7228):421-425
  56. 56. Zhang L, Yu J, Liu Z. MicroRNAs expressed by human cytomegalovirus. Virology Journal. 2020;17(1):34
  57. 57. Zhan S, Wang Y, Chen X. RNA virus-encoded microRNAs: Biogenesis, functions and perspectives on application. ExRNA. 2020;2(1):15
  58. 58. Abdalla AE et al. Human cytomegalovirus-encoded MicroRNAs: A master regulator of latent infection. Infection, Genetics and Evolution. 2020;78:104119
  59. 59. Kandeel M. Oncogenic viruses-encoded microRNAs and their role in the progression of cancer: Emerging targets for antiviral and anticancer therapies. Pharmaceuticals (Basel). 2023;16(4):485. DOI: 10.3390/ph16040485
  60. 60. Yu M et al. microRNA, a subtle indicator of human cytomegalovirus against host immune cells. Vaccines (Basel). 2022;10(2):144. DOI: 10.3390/vaccines10020144
  61. 61. Panigrahi M, Palmer MA, Wilson JA. MicroRNA-122 regulation of HCV infections: Insights from studies of miR-122-independent replication. Pathogens. 2022;11(9):1005. DOI: 10.3390/pathogens11091005
  62. 62. Li Y et al. The role of MicroRNA in DNA damage response. Frontiers in Genetics. 2022;13:850038
  63. 63. Visser H, Thomas AD. MicroRNAs and the DNA damage response: How is cell fate determined? DNA Repair (Amst). 2021;108:103245
  64. 64. Orafidiya F et al. Crosstalk between long non coding RNAs, microRNAs and DNA damage repair in prostate cancer: New therapeutic opportunities? Cancers (Basel). 2022;14(3):755. DOI: 10.3390/cancers14030755
  65. 65. Gajek A et al. Current implications of microRNAs in genome stability and stress responses of ovarian cancer. Cancers (Basel). 2021;13(11):2690. DOI: 10.3390/cancers13112690
  66. 66. Szatkowska M, Krupa R. Regulation of DNA damage response and homologous recombination repair by microRNA in human cells exposed to ionizing radiation. Cancers (Basel). 2020;12(7):1838. DOI: 10.3390/cancers12071838
  67. 67. Pellegrini L et al. MicroRNAs in cancer treatment-induced cardiotoxicity. Cancers (Basel). 2020;12(3):704. DOI: 10.3390/cancers12030704
  68. 68. Shi X, Yang H, Birchler JA. MicroRNAs play regulatory roles in genomic balance. BioEssays. 2023;45(2):e2200187
  69. 69. Huppi K et al. Genomic instability and mouse microRNAs. Toxicology Mechanisms and Methods. 2011;21(4):325-333
  70. 70. Devara D, Choudhary Y, Kumar S. Role of MicroRNA-502-3p in human diseases. Pharmaceuticals (Basel). 2023;16(4):532. DOI: 10.3390/ph16040532
  71. 71. Zhao Y et al. New insights into the functions of MicroRNAs in cardiac fibrosis: From mechanisms to therapeutic strategies. Genes (Basel). 2022;13(8):1390. DOI: 10.3390/genes13081390
  72. 72. Stunf, Pukl S. MicroRNA of epithelial to mesenchymal transition in Fuchs' endothelial corneal dystrophy. Genes (Basel). 2022;13(10):1711. DOI: 10.3390/genes13101711
  73. 73. Ruiz-Manriquez LM et al. A brief review on the regulatory roles of MicroRNAs in cystic diseases and their use as potential biomarkers. Genes (Basel). 2022;13(2):191. DOI: 10.3390/genes13020191
  74. 74. Ramirez AE et al. MicroRNA: A linking between astrocyte dysfunction, mild cognitive impairment, and neurodegenerative diseases. Life (Basel). 2022;12(9):1439. DOI: 10.3390/life12091439
  75. 75. Moriondo G et al. Obstructive sleep Apnea: A look towards micro-RNAs as biomarkers of the future. Biology (Basel). 2023;12(1):66. DOI: 10.3390/biology12010066
  76. 76. Lee C, Han J, Jung Y. Pathological contribution of extracellular vesicles and their MicroRNAs to progression of chronic liver disease. Biology (Basel). 2022;11(5):637. DOI: 10.3390/biology11050637
  77. 77. Iulian, Stanciugelu S et al. Osteoarthritis and microRNAs: Do they provide novel insights into the pathophysiology of this degenerative disorder? Life (Basel). 2022;12(11):1914. DOI: 10.3390/life12111914
  78. 78. Homorogan C et al. Uncovering the roles of MicroRNAs in major depressive disorder: From candidate diagnostic biomarkers to treatment response indicators. Life (Basel). 2021;11(10):1073. DOI: 10.3390/life11101073
  79. 79. De Benedittis G et al. Emerging role of microRNAs and long non-coding RNAs in Sjogren's syndrome. Genes (Basel). 2021;12(6):903. DOI: 10.3390/genes12060903
  80. 80. Chiti E et al. MicroRNAs in hypertrophic, Arrhythmogenic and dilated cardiomyopathy. Diagnostics (Basel). 2021;11(9):1720. DOI: 10.3390/diagnostics11091720
  81. 81. Barbiera A et al. Nutrition and microRNAs: Novel insights to fight sarcopenia. Antioxidants (Basel). 2020;9(10):951. DOI: 10.3390/antiox9100951
  82. 82. Martins B et al. Extracellular vesicles and MicroRNA: Putative role in diagnosis and treatment of diabetic retinopathy. Antioxidants (Basel). 2020;9(8):705. DOI: 10.3390/antiox9080705
  83. 83. De Palma FDE et al. The multifaceted roles of MicroRNAs in cystic fibrosis. Diagnostics (Basel). 2020;10(12):1102. DOI: 10.3390/diagnostics10121102
  84. 84. Suzuki HI. Roles of MicroRNAs in disease biology. JMA Journal. 2023;6(2):104-113
  85. 85. Kaller M et al. Analysis of the p53/microRNA network in cancer. Advances in Experimental Medicine and Biology. 2022;1385:187-228
  86. 86. Dayakar A, Shanmukha KD, Kalangi SK. Spectrum of microRNAs and their target genes in cancer: Intervention in diagnosis and therapy. Molecular Biology Reports. 2022;49(7):6827-6846
  87. 87. Semina EV et al. MicroRNAs in cancer: From gene expression regulation to the metastatic niche reprogramming. Biochemistry (Mosc). 2021;86(7):785-799
  88. 88. Angius A et al. Modulatory role of microRNAs in triple negative breast cancer with basal-like phenotype. Cancers (Basel). 2020;12(11):3298. DOI: 10.3390/cancers12113298
  89. 89. Alshamrani AA. Roles of microRNAs in ovarian cancer tumorigenesis: Two decades later, what have we learned? Frontiers in Oncology. 2020;10:1084
  90. 90. Sell MC et al. MicroRNAs in cancer metastasis: Biological and therapeutic implications. Expert Reviews in Molecular Medicine. 2023;25:e14
  91. 91. Pekarek L et al. An overview of the role of MicroRNAs on carcinogenesis: A focus on cell cycle, angiogenesis and metastasis. International Journal of Molecular Sciences. 2023;24(8):7268. DOI: 10.3390/ijms24087268
  92. 92. Pan W et al. p53/MicroRNA-34 axis in cancer and beyond. Heliyon. 2023;9(4):e15155
  93. 93. Sevcikova A et al. Clinical significance of microRNAs in hematologic malignancies and hematopoietic stem cell transplantation. Cancers (Basel). 2023;15(9):2658. DOI: 10.3390/cancers15092658
  94. 94. Tran F et al. MicroRNA-gene interactions impacted by toxic metal(oid)s during EMT and carcinogenesis. Cancers (Basel). 2022;14(23):5818. DOI: 10.3390/cancers14235818
  95. 95. Sobolewski C, Dubuquoy L, Legrand N. MicroRNAs, Tristetraprolin family members and HuR: A complex interplay controlling cancer-related processes. Cancers (Basel). 2022;14(14):3516. DOI: 10.3390/cancers14143516
  96. 96. Setlai BP et al. MicroRNA interrelated epithelial mesenchymal transition (EMT) in glioblastoma. Genes (Basel). 2022;13(2):244. DOI: 10.3390/genes13020244
  97. 97. Rajtmajerova M et al. Long non-coding RNA and microRNA interplay in colorectal cancer and their effect on the tumor microenvironment. Cancers (Basel). 2022;14(21):5450. DOI: 10.3390/cancers14215450
  98. 98. Prinz C et al. Emerging role of microRNA dysregulation in diagnosis and prognosis of extrahepatic cholangiocarcinoma. Genes (Basel). 2022;13(8):1479. DOI: 10.3390/genes13081479
  99. 99. Pecorelli A, Valacchi G. Oxidative-stress-sensitive microRNAs in UV-promoted development of melanoma. Cancers (Basel). 2022;14(13):3224. DOI: 10.3390/cancers14133224
  100. 100. Koustas E et al. The emerging role of MicroRNAs and autophagy mechanism in pancreatic cancer progression: Future therapeutic approaches. Genes (Basel). 2022;13(10):1868. DOI: 10.3390/genes13101868
  101. 101. Wang X et al. Effects of CAF-derived MicroRNA on tumor biology and clinical applications. Cancers (Basel). 2021;13(13):3160. DOI: 10.3390/cancers13133160
  102. 102. Tomei S et al. Cancer stem cells are possible key players in regulating anti-tumor immune responses: The role of Immunomodulating molecules and MicroRNAs. Cancers (Basel). 2021;13(7):1674. DOI: 10.3390/cancers13071674
  103. 103. Morishita A et al. MicroRNAs in the pathogenesis of hepatocellular carcinoma: A review. Cancers (Basel). 2021;13(3):514. DOI: 10.3390/cancers13030514
  104. 104. Fitriana M et al. Roles of microRNAs in regulating cancer Stemness in head and neck cancers. Cancers (Basel). 2021;13(7):1742. DOI: 10.3390/cancers13071742
  105. 105. Kipkeeva F et al. MicroRNA in gastric cancer development: Mechanisms and biomarkers. Diagnostics (Basel). 2020;10(11):891. DOI: 10.3390/diagnostics10110891
  106. 106. Gluud M et al. MicroRNAs in the pathogenesis, diagnosis, prognosis and targeted treatment of cutaneous T-cell lymphomas. Cancers (Basel). 2020;12(5):1229. DOI: 10.3390/cancers12051229
  107. 107. Soond SM et al. Integrative p53, micro-RNA and Cathepsin protease Co-regulatory expression networks in cancer. Cancers (Basel). 2020;12(11):3454. DOI: 10.3390/cancers12113454
  108. 108. Aali M et al. Evaluating the role of microRNAs alterations in oral squamous cell carcinoma. Gene. 2020;757:144936
  109. 109. Wang N et al. MicroRNA-149: A review of its role in digestive system cancers. Pathology, Research and Practice. 2020;216(12):153266
  110. 110. Zietara KJ et al. The importance of selected dysregulated microRNAs in diagnosis and prognosis of childhood B-cell precursor acute lymphoblastic Leukemia. Cancers (Basel). 2023;15(2):428. DOI: 10.3390/cancers15020428
  111. 111. Teo AYT, Lim VY, Yang VS. MicroRNAs in the pathogenesis, prognostication and prediction of treatment resistance in soft tissue sarcomas. Cancers (Basel). 2020;12:1229. DOI: 10.3390/cancers12051229
  112. 112. Psilopatis I et al. The role of MicroRNAs in uterine Leiomyosarcoma diagnosis and treatment. Cancers (Basel). 2023;15(9):2420. DOI: 10.3390/cancers15092420
  113. 113. Maldonado E et al. Role of the mediator complex and MicroRNAs in breast cancer Etiology. Genes (Basel). 2022;13(2):234. DOI: 10.3390/genes13020234
  114. 114. Fogazzi V et al. The role of MicroRNAs in HER2-positive breast cancer: Where we are and future prospective. Cancers (Basel). 2022;14(21):5326. DOI: 10.3390/cancers14215326
  115. 115. Bocchetti M et al. MicroRNAs' crucial role in salivary gland Cancers' onset and prognosis. Cancers (Basel). 2022;14(21):5304. DOI: 10.3390/cancers14215304
  116. 116. Notarte KI et al. MicroRNA and other non-coding RNAs in Epstein-Barr virus-associated cancers. Cancers (Basel). 2021;13(15):3909. DOI: 10.3390/cancers13153909
  117. 117. Bevacqua E et al. Role of MicroRNAs in the development and progression of the four Medulloblastoma subgroups. Cancers (Basel). 2021;13(24):6323. DOI: 10.3390/cancers13246323
  118. 118. Sidorkiewicz I et al. Insulin resistance and endometrial cancer: Emerging role for microRNA. Cancers (Basel). 2020;12(9):2559. DOI: 10.3390/cancers12092559
  119. 119. Hitu L et al. MicroRNA in papillary thyroid carcinoma: A systematic review from 2018 to June 2020. Cancers (Basel). 2020;12(11):3118. DOI: 10.3390/cancers12113118
  120. 120. Khan P et al. MicroRNA-1: Diverse role of a small player in multiple cancers. Seminars in Cell & Developmental Biology. 2022;124:114-126
  121. 121. Grzywa TM, Klicka K, Wlodarski PK. Regulators at every step-how microRNAs drive tumor cell invasiveness and metastasis. Cancers (Basel). 2020;12(12):3709. DOI: 10.3390/cancers12123709
  122. 122. Siegl F et al. The significance of MicroRNAs in the molecular pathology of brain metastases. Cancers (Basel). 2022;14(14):3386. DOI: 10.3390/cancers14143386
  123. 123. Rodriguez, Calleja L et al. The p53 family members p63 and p73 roles in the metastatic dissemination: Interactions with microRNAs and TGFbeta pathway. Cancers (Basel). 2022;14(23):5948. DOI: 10.3390/cancers14235948
  124. 124. Haider MT, Smit DJ, Taipaleenmaki H. MicroRNAs: Emerging regulators of metastatic bone disease in breast cancer. Cancers (Basel). 2022;14(3):729. DOI: 10.3390/cancers14030729
  125. 125. Tang LB et al. Exosomal microRNAs: Pleiotropic impacts on breast cancer metastasis and their clinical perspectives. Biology (Basel). 2021;10(4):307. DOI: 10.3390/biology10040307
  126. 126. Oh-Hohenhorst SJ, Lange T. Role of metastasis-related microRNAs in prostate cancer progression and treatment. Cancers (Basel). 2021;13(17):4492. DOI: 10.3390/cancers13174492
  127. 127. Hammouz RY et al. MicroRNAs: Their role in metastasis, angiogenesis, and the potential for biomarker utility in bladder carcinomas. Cancers (Basel). 2021;13(4):891. DOI: 10.3390/cancers13040891
  128. 128. Wood SL, Brown JE. Personal medicine and bone metastases: Biomarkers, micro-RNAs and bone metastases. Cancers (Basel). 2020;12(8):2109. DOI: 10.3390/cancers12082109
  129. 129. Weidle UH et al. MicroRNAs involved in metastasis of hepatocellular carcinoma: Target candidates, functionality and efficacy in animal models and prognostic relevance. Cancer Genomics Proteomics. 2020;17(1):1-21
  130. 130. Santos JMO et al. The emerging role of MicroRNAs and other non-coding RNAs in cancer cachexia. Cancers (Basel). 2020;12(4):1004. DOI: 10.3390/cancers12041004
  131. 131. Pavlikova L et al. The roles of microRNAs in cancer multidrug resistance. Cancers (Basel). 2022;14(4):1090. DOI: 10.3390/cancers14041090
  132. 132. Mahinfar P et al. The role of microRNAs in multidrug resistance of glioblastoma. Cancers (Basel). 2022;14(13):3217. DOI: 10.3390/cancers14133217
  133. 133. Cuttano R, Afanga MK, Bianchi F. MicroRNAs and drug resistance in non-small cell lung cancer: Where are we now and where are we going. Cancers (Basel). 2022;14(23):5731. DOI: 10.3390/cancers14235731
  134. 134. Crudele F et al. The molecular networks of microRNAs and their targets in the drug resistance of colon carcinoma. Cancers (Basel). 2021;13(17):4355. DOI: 10.3390/cancers13174355
  135. 135. Cosentino G et al. Breast cancer drug resistance: Overcoming the challenge by capitalizing on MicroRNA and tumor microenvironment interplay. Cancers (Basel). 2021;13(15)
  136. 136. Lampis A et al. MicroRNAs as mediators of drug resistance mechanisms. Current Opinion in Pharmacology. 2020;54:44-50
  137. 137. Funamizu N et al. microRNAs associated with gemcitabine resistance via EMT, TME, and drug metabolism in pancreatic cancer. Cancers (Basel). 2023;15(4)
  138. 138. Masadah R et al. The role of microRNAs in the cisplatin- and radio-resistance of cervical cancer. Cancers (Basel). 2021;13(5)
  139. 139. Podralska M et al. Non-coding RNAs in cancer Radiosensitivity: MicroRNAs and lncRNAs as regulators of radiation-induced Signaling pathways. Cancers (Basel). 2020;12(6)
  140. 140. Bozzato AM et al. MicroRNAs related to TACE treatment response: A review of the literature from a radiological point of view. Diagnostics (Basel). 2022;12(2)
  141. 141. Sukocheva OA et al. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Seminars in Cancer Biology. 2022;86(Pt 2):358-375
  142. 142. Arghiani N, Shah K. Modulating microRNAs in cancer: Next-generation therapies. Cancer Biology & Medicine. 2021;19(3):289-304
  143. 143. Karkhane M et al. Oncogenesis and tumor inhibition by MicroRNAs and its potential therapeutic applications: A systematic review. MicroRNA. 2020;9(3):198-215
  144. 144. Chen L et al. Premature MicroRNA-based therapeutic: A “one-two punch” against cancers. Cancers (Basel). 2020;12(12)
  145. 145. Abd-Aziz N, Kamaruzman NI, Poh CL. Development of MicroRNAs as potential therapeutics against cancer. Journal of Oncology. 2020;2020:8029721
  146. 146. Samad AFA, Kamaroddin MF. Innovative approaches in transforming microRNAs into therapeutic tools. Wiley Interdisciplinary Reviews. RNA. 2023;14(1):e1768
  147. 147. Liu J et al. SALL4 and microRNA: The role of Let-7. Genes (Basel). 2021;12(9)
  148. 148. Li WJ et al. MicroRNA-34a: Potent tumor suppressor, cancer stem cell inhibitor, and potential anticancer therapeutic. Frontiers in Cell and Development Biology. 2021;9:640587
  149. 149. Li WJ et al. Prostate cancer stem cells, and therapeutic development. Cancers (Basel). 2022;14(18)
  150. 150. Kara G et al. miRacle of microRNA-driven cancer Nanotherapeutics. Cancers (Basel). 2022;14(15)
  151. 151. Genedy HH, Delair T, Montembault A. Chitosan Based MicroRNA Nanocarriers. Pharmaceuticals (Basel). 2022;15(9)
  152. 152. Romano G, Acunzo M, Nana-Sinkam P. microRNAs as Novel Therapeutics in Cancer. Cancers (Basel). 2021;13(7)
  153. 153. Reda, El Sayed S et al. MicroRNA therapeutics in cancer: Current advances and challenges. Cancers (Basel). 2021;13(11)
  154. 154. Shadbad MA et al. A systematic review on the therapeutic potentiality of PD-L1-inhibiting MicroRNAs for triple-negative breast cancer: Toward single-cell sequencing-guided biomimetic delivery. Genes (Basel). 2021;12(8)
  155. 155. Ohtsuka M et al. Circulating MicroRNAs in Gastrointestinal Cancer. Cancers (Basel). 2021;13:13
  156. 156. Niccolini B et al. Opportunities offered by graphene nanoparticles for MicroRNAs delivery for amyotrophic lateral sclerosis treatment. Materials (Basel). 2021;15(1)
  157. 157. Jung E et al. MicroRNA-based therapeutics for drug-resistant colorectal cancer. Pharmaceuticals (Basel). 2021;14(2)
  158. 158. Wong GL, Abu Jalboush S, Lo HW. Exosomal MicroRNAs and organotropism in breast cancer metastasis. Cancers (Basel). 2020;12(7)
  159. 159. Balachandran AA et al. Therapeutically significant MicroRNAs in primary and metastatic brain malignancies. Cancers (Basel). 2020;12(9)
  160. 160. Imedio L et al. MicroRNAs in rectal cancer: Functional significance and promising therapeutic value. Cancers (Basel). 2020;12(8)
  161. 161. Hsieh PL et al. MicroRNAs as Theranostics targets in Oral carcinoma stem cells. Cancers (Basel). 2020;12(2)
  162. 162. Fortunato O, Iorio MV. The therapeutic potential of MicroRNAs in cancer: Illusion or opportunity? Pharmaceuticals (Basel). 2020;13(12)
  163. 163. Forterre A et al. A comprehensive review of cancer MicroRNA therapeutic delivery strategies. Cancers (Basel). 2020;12(7)
  164. 164. Eckburg A et al. Oligonucleotides and microRNAs targeting telomerase subunits in cancer therapy. Cancers (Basel). 2020;12(9)
  165. 165. Liu D et al. Drugging the “undruggable” microRNAs. Cellular and Molecular Life Sciences. 2021;78(5):1861-1871
  166. 166. Chaudhry T, Coxon CR, Ross K. Trading places: Peptide and small molecule alternatives to oligonucleotide-based modulation of microRNA expression. Drug Discovery Today. 2022;27(11):103337
  167. 167. Van Meter EN, Onyango JA, Teske KA. A review of currently identified small molecule modulators of microRNA function. European Journal of Medicinal Chemistry. 2020;188:112008
  168. 168. Ma L et al. MicroRNAs as mediators of adipose thermogenesis and potential therapeutic targets for obesity. Biology (Basel). 2022;11(11)
  169. 169. Weidle UH et al. Gastric cancer: Identification of microRNAs inhibiting Druggable targets and mediating efficacy in preclinical In vivo models. Cancer Genomics Proteomics. 2021;18(4):497-514
  170. 170. Weidle UH et al. Down-regulated MicroRNAs in gastric carcinoma may Be targets for therapeutic intervention and replacement therapy. Anticancer Research. 2021;41(9):4185-4202
  171. 171. Weidle UH, Birzele F. Bladder cancer-related microRNAs with in vivo efficacy in preclinical models. Cancer Diagnosis & Prognosis. 2021;1(4):245-263
  172. 172. Doldi V, El Bezawy R, Zaffaroni N. MicroRNAs as epigenetic determinants of treatment response and potential therapeutic targets in prostate cancer. Cancers (Basel). 2021;13(10)
  173. 173. Cannataro R et al. Polyphenols in the Mediterranean diet: From dietary sources to microRNA modulation. Antioxidants (Basel). 2021;10(2)
  174. 174. Chehade M et al. Key MicroRNA's and their Targetome in adrenocortical cancer. Cancers (Basel). 2020;12(8)
  175. 175. Christou N et al. Circulating tumour cells, circulating tumour DNA and circulating tumour miRNA in blood assays in the different steps of colorectal cancer management, a review of the evidence in 2019. BioMed Research International. 2019;2019:5953036
  176. 176. Olivieri F et al. Circulating miRNAs and miRNA shuttles as biomarkers: Perspective trajectories of healthy and unhealthy aging. Mechanisms of Ageing and Development. 2017;165(Pt B):162-170
  177. 177. Rocchi A et al. MicroRNAs: An update of applications in forensic science. Diagnostics (Basel). 2020;11(1)
  178. 178. Regouc M et al. Non-coding microRNAs as novel potential tumor markers in testicular cancer. Cancers (Basel). 2020;12(3)
  179. 179. Glynn CL. Potential applications of microRNA profiling to forensic investigations. RNA. 2020;26(1):1-9
  180. 180. Ghosh RD, Pattatheyil A, Roychoudhury S. Functional landscape of dysregulated MicroRNAs in Oral squamous cell carcinoma: Clinical implications. Frontiers in Oncology. 2020;10:619
  181. 181. Varela N et al. The current state of MicroRNAs as restenosis biomarkers. Frontiers in Genetics. 2019;10:1247
  182. 182. Ahadi A. A systematic review of microRNAs as potential biomarkers for diagnosis and prognosis of gastric cancer. Immunogenetics. 2021;73(2):155-161
  183. 183. Yang IP et al. MicroRNAs as predictive biomarkers in patients with colorectal cancer receiving chemotherapy or chemoradiotherapy: A narrative literature review. Cancers (Basel). 2023;15(5)
  184. 184. Salamanna F et al. Sharing circulating micro-RNAs between osteoporosis and sarcopenia: A systematic review. Life (Basel). 2023;13(3)
  185. 185. Fawzy MS et al. MicroRNA-155 and disease-related Immunohistochemical parameters in cutaneous melanoma. Diagnostics (Basel). 2023;13(6)
  186. 186. Zhang J et al. Recent advances in the roles of MicroRNA and MicroRNA-based diagnosis in neurodegenerative diseases. Biosensors (Basel). 2022;12(12)
  187. 187. Wielogorska M et al. MicroRNA profile alterations in parathyroid carcinoma: Latest updates and perspectives. Cancers (Basel). 2022;14(4)
  188. 188. Vykoukal J et al. Contributions of circulating microRNAs for early detection of lung cancer. Cancers (Basel). 2022;14(17)
  189. 189. Shaw P et al. A clinical investigation on the Theragnostic effect of MicroRNA biomarkers for survival outcome in cervical cancer: A PRISMA-P compliant protocol for systematic review and comprehensive meta-analysis. Genes (Basel). 2022;13(3)
  190. 190. Le TAH, Lao TD. Circulating microRNAs as the potential diagnostic and prognostic biomarkers for nasopharyngeal carcinoma. Genes (Basel). 2022;13(7)
  191. 191. Kyriakidis I, Kyriakidis K, Tsezou A. MicroRNAs and the diagnosis of childhood acute lymphoblastic Leukemia: Systematic review, meta-analysis and Re-analysis with novel small RNA-Seq tools. Cancers (Basel). 2022;14(16)
  192. 192. Juracek J et al. Urinary microRNAs and their significance in prostate cancer diagnosis: A 5-year update. Cancers (Basel). 2022;14(13)
  193. 193. Davies M, Davey MG, Miller N. The potential of MicroRNAs as clinical biomarkers to aid ovarian cancer diagnosis and treatment. Genes (Basel). 2022;13(11)
  194. 194. Dabi Y et al. Clues for improving the pathophysiology knowledge for endometriosis using serum micro-RNA expression. Diagnostics (Basel). 2022;12(1)
  195. 195. Bevacqua E et al. The potential of MicroRNAs as non-invasive prostate cancer biomarkers: A systematic literature review based on a machine learning approach. Cancers (Basel). 2022;14(21)
  196. 196. Aghiorghiesei O et al. The world of Oral cancer and its risk factors viewed from the aspect of MicroRNA expression patterns. Genes (Basel). 2022;13(4)
  197. 197. Zhao Z et al. Fecal microRNAs, Fecal microRNA panels, or combinations of Fecal microRNAs with Fecal Hemoglobin for early detection of colorectal cancer and its precursors: A systematic review. Cancers (Basel). 2021;14(1)
  198. 198. Weidle UH, Birzele F, Nopora A. microRNAs promoting growth of gastric cancer xenografts and correlation to clinical prognosis. Cancer Genomics Proteomics. 2021;18(1):1-15
  199. 199. Turai PI et al. MicroRNAs, long non-coding RNAs, and circular RNAs: Potential biomarkers and therapeutic targets in Pheochromocytoma/Paraganglioma. Cancers (Basel). 2021;13(7)
  200. 200. Tito C et al. Circulating microRNAs from the molecular mechanisms to clinical biomarkers: A focus on the clear cell renal cell carcinoma. Genes (Basel). 2021;12(8)
  201. 201. Shaw P et al. A clinical update on the prognostic effect of microRNA biomarkers for survival outcome in nasopharyngeal carcinoma: A systematic review and meta-analysis. Cancers (Basel). 2021;13(17)
  202. 202. Sequeira JP et al. Unveiling the world of circulating and Exosomal microRNAs in renal cell carcinoma. Cancers (Basel). 2021;13(21)
  203. 203. Rzepka-Migut B, Paprocka J. Prospects and limitations related to the use of MicroRNA as a biomarker of epilepsy in children: A systematic review. Life (Basel). 2021;11(1)
  204. 204. Richard V et al. MicroRNAs in molecular classification and pathogenesis of breast Tumors. Cancers (Basel). 2021;13(21)
  205. 205. Prinz C, Mese K, Weber D. MicroRNA changes in gastric carcinogenesis: Differential dysregulation during helicobacter pylori and EBV infection. Genes (Basel). 2021;12(4)
  206. 206. Osan C et al. The connection between MicroRNAs and Oral cancer pathogenesis: Emerging biomarkers in Oral cancer management. Genes (Basel). 2021;12(12)
  207. 207. MacKenzie SM et al. Circulating microRNAs as diagnostic markers in primary Aldosteronism. Cancers (Basel). 2021;13(21)
  208. 208. Low SS et al. Recent Progress in nanomaterials modified electrochemical biosensors for the detection of MicroRNA. Micromachines (Basel). 2021;12(11)
  209. 209. Lao TD, Le TAH. Data integration reveals the potential biomarkers of circulating MicroRNAs in osteoarthritis. Diagnostics (Basel). 2021;11(3)
  210. 210. Jiang Y et al. MicroRNAs as potential biomarkers for exercise-based cancer rehabilitation in cancer survivors. Life (Basel). 2021;11(12)
  211. 211. Huang PS et al. Functional and clinical significance of dysregulated microRNAs in liver cancer. Cancers (Basel). 2021;13(21)
  212. 212. Gayosso-Gomez LV, Ortiz-Quintero B. Circulating MICRORNAS in blood and other body fluids as biomarkers for diagnosis, prognosis, and therapy response in lung cancer. Diagnostics (Basel). 2021:11(3):421. DOI: 10.3390/diagnostics11030421
  213. 213. Favier A et al. MicroRNA as epigenetic modifiers in endometrial cancer: A systematic review. Cancers (Basel). 2021;13(5)
  214. 214. Desantis V et al. MicroRNAs as a potential new preventive approach in the transition from asymptomatic to symptomatic multiple myeloma disease. Cancers (Basel). 2021;13(15)
  215. 215. Cione E et al. Exosome microRNAs in metabolic syndrome as tools for the early monitoring of diabetes and possible therapeutic options. Pharmaceuticals (Basel). 2021;14(12)
  216. 216. Chorley BN et al. Methodological considerations for measuring biofluid-based microRNA biomarkers. Critical Reviews in Toxicology. 2021;51(3):264-282
  217. 217. Cavallari I et al. The miR-200 family of microRNAs: Fine tuners of epithelial-mesenchymal transition and circulating cancer biomarkers. Cancers (Basel). 2021;13(23)
  218. 218. Borgia F et al. Involvement of microRNAs as a response to phototherapy and photodynamic therapy: A literature review. Antioxidants (Basel). 2021;10(8)
  219. 219. Andrikopoulou A et al. MicroRNAs as potential predictors of response to CDK4/6 inhibitor treatment. Cancers (Basel). 2021;13(16)
  220. 220. Al-Akhrass H, Christou N. The clinical assessment of MicroRNA diagnostic, prognostic, and Theranostic value in colorectal cancer. Cancers (Basel). 2021;13(12)
  221. 221. El Aamri M et al. Electrochemical biosensors for detection of MicroRNA as a cancer biomarker: Pros and cons. Biosensors (Basel). 2020;10(11)
  222. 222. Silaghi CA et al. The prognostic value of MicroRNAs in thyroid cancers-a systematic review and meta-analysis. Cancers (Basel). 2020;12(9)
  223. 223. Izzotti A et al. Predicting response to neoadjuvant therapy in colorectal cancer patients the role of messenger-and micro-RNA profiling. Cancers (Basel). 2020;12(6)
  224. 224. Maiese A et al. MicroRNAs as useful tools to estimate time since death. A systematic review of current literature. Diagnostics (Basel). 2021;11(1)
  225. 225. Witwer KW. XenomiRs and miRNA homeostasis in health and disease: Evidence that diet and dietary miRNAs directly and indirectly influence circulating miRNA profiles. RNA Biology. 2012;9(9):1147-1154
  226. 226. Soplinska A et al. MicroRNAs as biomarkers of systemic changes in response to endurance exercise-a comprehensive review. Diagnostics (Basel). 2020;10(10)
  227. 227. Dufresne S et al. A review of physical activity and circulating miRNA expression: Implications in cancer risk and progression. Cancer Epidemiology, Biomarkers & Prevention. 2018;27(1):11-24
  228. 228. Lewis BP et al. Prediction of mammalian microRNA targets. Cell. 2003;115(7):787-798
  229. 229. Li Z, Xu R, Li N. MicroRNAs from plants to animals, do they define a new messenger for communication? Nutrition & Metabolism (London). 2018;15:68
  230. 230. Landi D, Gemignani F, Landi S. Role of variations within microRNA-binding sites in cancer. Mutagenesis. 2012;27(2):205-210
  231. 231. Kasinski AL, Slack FJ. Epigenetics and genetics. MicroRNAs en route to the clinic: Progress in validating and targeting microRNAs for cancer therapy. Nature Reviews Cancer. 2011;11(12):849-864
  232. 232. de Rooij LA et al. The microRNA lifecycle in health and cancer. Cancers (Basel). 2022;14(23)
  233. 233. Bartel DP. MicroRNAs: Target recognition and regulatory functions. Cell. 2009;136(2):215-233
  234. 234. Liu J, Liu F. The yin and Yang function of microRNAs in insulin signalling and cancer. RNA Biology. 2021;18(1):24-32
  235. 235. Sindhu KJ, Venkatesan N, Karunagaran D. MicroRNA Interactome multiomics characterization for cancer research and personalized medicine: An expert review. OMICS. 2021;25(9):545-566
  236. 236. Komatsu S, Kitai H, Suzuki HI. Network regulation of microRNA biogenesis and target interaction. Cell. 2023;12(2)
  237. 237. Skrzypek K, Majka M. Interplay among SNAIL transcription factor, MicroRNAs, long non-coding RNAs, and circular RNAs in the regulation of tumor growth and metastasis. Cancers (Basel). 2020;12(1)
  238. 238. Poli V, Secli L, Avalle L. The Microrna-143/145 cluster in tumors: A matter of where and when. Cancers (Basel). 2020;12(3)
  239. 239. Perri P et al. A focus on regulatory networks linking MicroRNAs, transcription factors and target genes in neuroblastoma. Cancers (Basel). 2021;13(21)
  240. 240. Chen K et al. Polymorphisms in microRNA targets: A gold mine for molecular epidemiology. Carcinogenesis. 2008;29(7):1306-1311
  241. 241. Sethupathy P, Collins FS. MicroRNA target site polymorphisms and human disease. Trends in Genetics. 2008;24(10):489-497
  242. 242. Pelletier C, Weidhaas JB. MicroRNA binding site polymorphisms as biomarkers of cancer risk. Expert Review of Molecular Diagnostics. 2010;10(6):817-829
  243. 243. Ryan BM, Robles AI, Harris CC. Genetic variation in microRNA networks: The implications for cancer research. Nature Reviews Cancer. 2010;10(6):389-402
  244. 244. Skeeles LE et al. The impact of 3'UTR variants on differential expression of candidate cancer susceptibility genes. PLoS One. 2013;8(3):e58609
  245. 245. Ledda B et al. Small RNAs in eucaryotes: New clues for amplifying microRNA benefits. Cell & Bioscience. 2020;10:1
  246. 246. Casarotto M et al. Beyond MicroRNAs: Emerging role of other non-coding RNAs in HPV-driven cancers. Cancers (Basel). 2020;12(5)
  247. 247. Tjaden B et al. Target prediction for small, noncoding RNAs in bacteria. Nucleic Acids Research. 2006;34(9):2791-2802
  248. 248. Lee HE, Huh JW, Kim HS. Bioinformatics analysis of evolution and human disease related transposable element-derived microRNAs. Life (Basel). 2020;10(6)
  249. 249. Pegler JL et al. Miniature inverted-repeat transposable elements: Small DNA transposons that have contributed to plant MICRORNA gene evolution. Plants (Basel). 2023;12(5)
  250. 250. Hu X et al. Recent advances in the functional explorations of nuclear microRNAs. Frontiers in Immunology. 2023;14:1097491
  251. 251. Ortiz-Quintero B. Extracellular MicroRNAs as intercellular mediators and noninvasive biomarkers of cancer. Cancers (Basel). 2020;12(11)
  252. 252. Jimenez-Avalos JA, Fernandez-Macias JC, Gonzalez-Palomo AK. Circulating exosomal MicroRNAs: New non-invasive biomarkers of non-communicable disease. Molecular Biology Reports. 2021;48(1):961-967
  253. 253. Vu LT et al. microRNA exchange via extracellular vesicles in cancer. Cell Proliferation. 2020;53(11):e12877
  254. 254. Li J, Jiang X, Wang K. Exosomal miRNA: an alternative mediator of cell-to-cell communication. ExRNA. 2019;1(1):31
  255. 255. Li Z, Xu R, Li N. Correction to: MicroRNAs from plants to animals, do they define a new messenger for communication? Nutrition & Metabolism (London). 2018;15:74
  256. 256. Cai X, Hagedorn CH, Cullen BR. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA. 2004;10(12):1957-1966
  257. 257. Lee Y et al. MicroRNA genes are transcribed by RNA polymerase II. The EMBO Journal. 2004;23(20):4051-4060
  258. 258. Lee Y et al. MicroRNA maturation: Stepwise processing and subcellular localization. The EMBO Journal. 2002;21(17):4663-4670
  259. 259. Singh G, Storey KB. MicroRNA cues from nature: A roadmap to decipher and combat challenges in human health and disease? Cell. 2021;10(12)
  260. 260. Vilimova M, Pfeffer S. Post-transcriptional regulation of polycistronic microRNAs. Wiley Interdisciplinary Reviews. RNA. 2023;14(2):e1749
  261. 261. Bartel DP. Metazoan MicroRNAs. Cell. 2018;173(1):20-51
  262. 262. Annese T et al. microRNAs biogenesis, functions and role in tumor angiogenesis. Frontiers in Oncology. 2020;10:581007
  263. 263. Ha M, Kim VN. Regulation of microRNA biogenesis. Nature Reviews Molecular Cell Biology. 2014;15:509
  264. 264. Abdelfattah AM, Park C, Choi MY. Update on non-canonical microRNAs. Biomolecular Concepts. 2014;5(4):275-287
  265. 265. De Paolis V et al. Epitranscriptomics: A new layer of microRNA regulation in cancer. Cancers (Basel). 2021;13(13)
  266. 266. Gregorova J, Vychytilova-Faltejskova P, Sevcikova S. Epigenetic regulation of MicroRNA clusters and families during tumor development. Cancers (Basel). 2021;13(6)
  267. 267. Pifkova L, Vesela P, Slaby O. The development and significance of microRNA sequence variants in carcinogenesis. Klinická Onkologie. 2021;34(1):20-25
  268. 268. Orban TI. One locus, several functional RNAs-emerging roles of the mechanisms responsible for the sequence variability of microRNAs. Biologia Futura. 2023. DOI: 10.1007/s42977-023-00154-7
  269. 269. Wu D et al. Alternative splicing and MicroRNA: Epigenetic mystique in male reproduction. RNA Biology. 2022;19(1):162-175
  270. 270. Zhang B, Wang Q , Pan X. MicroRNAs and their regulatory roles in animals and plants. Journal of Cellular Physiology. 2007;210(2):279-289
  271. 271. Naeli P et al. The intricate balance between microRNA-induced mRNA decay and translational repression. The FEBS Journal. 2023;290(10):2508-2524
  272. 272. Henke JI et al. microRNA-122 stimulates translation of hepatitis C virus RNA. The EMBO Journal. 2008;27(24):3300-3310
  273. 273. Jopling CL et al. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005;309(5740):1577-1581
  274. 274. Li HC, Yang CH, Lo SY. Roles of microRNAs in hepatitis C virus replication and pathogenesis. Viruses. 2022;14(8)
  275. 275. Saraiya AA, Li W, Wang CC. Transition of a microRNA from repressing to activating translation depending on the extent of base pairing with the target. PLoS One. 2013;8(2):e55672
  276. 276. Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science. 2007;318(5858):1931-1934
  277. 277. Syed SN, Brune B. MicroRNAs as emerging regulators of Signaling in the tumor microenvironment. Cancers (Basel). 2020;12(4)
  278. 278. Hon LS, Zhang Z. The roles of binding site arrangement and combinatorial targeting in microRNA repression of gene expression. Genome Biology. 2007;8(8):R166
  279. 279. Reinhart BJ et al. MicroRNAs in plants. Genes & Development. 2002;16(13):1616-1626
  280. 280. Duan Y, Veksler-Lublinsky I, Ambros V. Critical contribution of 3′ non-seed base pairing to the in vivo function of the evolutionarily conserved let-7a microRNA. Cell Reports. 2022;39(4):110745
  281. 281. Han J, Mendell JT. MicroRNA turnover: A tale of tailing, trimming, and targets. Trends in Biochemical Sciences. 2023;48(1):26-39
  282. 282. Grimson A et al. MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Molecular Cell. 2007;27(1):91-105
  283. 283. Shin C et al. Expanding the microRNA targeting code: Functional sites with centered pairing. Molecular Cell. 2010;38(6):789-802
  284. 284. Garcia DM et al. Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nature Structural & Molecular Biology. 2011;18(10):1139-1146
  285. 285. Reczko M et al. Functional microRNA targets in protein coding sequences. Bioinformatics. 2012;28(6):771-776
  286. 286. Zhou X et al. Abundant conserved microRNA target sites in the 5′-untranslated region and coding sequence. Genetica. 2009;137(2):159-164
  287. 287. Jorgensen BG, Ro S. MicroRNAs and 'Sponging' competitive endogenous RNAs dysregulated in colorectal cancer: Potential as noninvasive biomarkers and therapeutic targets. International Journal of Molecular Sciences. 2022;23(4)
  288. 288. Zhou Q et al. Novel insights into MALAT1 function as a MicroRNA sponge in NSCLC. Frontiers in Oncology. 2021;11:758653
  289. 289. Cantile M et al. Functional interaction among lncRNA HOTAIR and MicroRNAs in cancer and other human diseases. Cancers (Basel). 2021;13(3)
  290. 290. Nam JW et al. Global analyses of the effect of different cellular contexts on microRNA targeting. Molecular Cell. 2014;53(6):1031-1043
  291. 291. Gu S et al. Functional SNP in 3'-UTR MicroRNA-binding site of ZNF350 confers risk for age-related cataract. Human Mutation. 2016;37(11):1223-1230
  292. 292. Flamand MN et al. A non-canonical site reveals the cooperative mechanisms of microRNA-mediated silencing. Nucleic Acids Research. 2017;45(12):7212-7225
  293. 293. Li L et al. Widespread microRNA degradation elements in target mRNAs can assist the encoded proteins. Genes & Development. 2021;35(23-24):1595-1609
  294. 294. Sethupathy P et al. Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 3′ untranslated region: A mechanism for functional single-nucleotide polymorphisms related to phenotypes. American Journal of Human Genetics. 2007;81(2):405-413
  295. 295. Genetic home reference. Retrieved from: http://ghr.nlm.nih.gov/handbook/genomicresearch/snp.
  296. 296. Gartner JJ et al. Whole-genome sequencing identifies a recurrent functional synonymous mutation in melanoma. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(33):13481-13486
  297. 297. Cline MS, Karchin R. Using bioinformatics to predict the functional impact of SNVs. Bioinformatics. 2011;27(4):441-448
  298. 298. Hu Z et al. Genetic polymorphisms in the precursor MicroRNA flanking region and non-small cell lung cancer survival. American Journal of Respiratory and Critical Care Medicine. 2011;183(5):641-648
  299. 299. Diederichs S, Haber DA. Sequence variations of microRNAs in human cancer: Alterations in predicted secondary structure do not affect processing. Cancer Research. 2006;66(12):6097-6104
  300. 300. Duan R, Pak C, Jin P. Single nucleotide polymorphism associated with mature miR-125a alters the processing of pri-miRNA. Human Molecular Genetics. 2007;16(9):1124-1131
  301. 301. Sun G et al. SNPs in human miRNA genes affect biogenesis and function. RNA. 2009;15(9):1640-1651
  302. 302. George GP, Mittal RD. MicroRNAs: Potential biomarkers in cancer. Indian Journal of Clinical Biochemistry. 2010;25(1):4-14
  303. 303. Greenberg E et al. Mutagen-specific mutation signature determines global microRNA binding. PLoS One. 2011;6(11):e27400
  304. 304. Wu H et al. Alternative processing of primary microRNA transcripts by Drosha generates 5′ end variation of mature microRNA. PLoS One. 2009;4(10):e7566
  305. 305. Calin GA, Croce CM. MicroRNA-cancer connection: The beginning of a new tale. Cancer Research. 2006;66(15):7390-7394
  306. 306. Clop A et al. A mutation creating a potential illegitimate microRNA target site in the myostatin gene affects muscularity in sheep. Nature Genetics. 2006;38(7):813-818
  307. 307. Hu Z, Bruno AE. The influence of 3'UTRs on MicroRNA function inferred from human SNP data. Comparative and Functional Genomics. 2011;2011:910769
  308. 308. Georges M, Coppieters W, Charlier C. Polymorphic miRNA-mediated gene regulation: Contribution to phenotypic variation and disease. Current Opinion in Genetics & Development. 2007;17(3):166-176
  309. 309. Borel C, Antonarakis SE. Functional genetic variation of human miRNAs and phenotypic consequences. Mammalian Genome. 2008;19(7-8):503-509
  310. 310. Lu M et al. An analysis of human microRNA and disease associations. PLoS One. 2008;3(10):e3420
  311. 311. Zhang R, Su B. MicroRNA regulation and the variability of human cortical gene expression. Nucleic Acids Research. 2008;36(14):4621-4628
  312. 312. Mencia A et al. Mutations in the seed region of human miR-96 are responsible for nonsyndromic progressive hearing loss. Nature Genetics. 2009;41(5):609-613
  313. 313. Harnprasopwat R et al. Alteration of processing induced by a single nucleotide polymorphism in pri-miR-126. Biochemical and Biophysical Research Communications. 2010;399(2):117-122
  314. 314. Slezak-Prochazka I et al. MicroRNAs, macrocontrol: Regulation of miRNA processing. RNA. 2010;16(6):1087-1095
  315. 315. Saunders MA, Liang H, Li WH. Human polymorphism at microRNAs and microRNA target sites. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(9):3300-3305
  316. 316. Gong J et al. Genome-wide identification of SNPs in microRNA genes and the SNP effects on microRNA target binding and biogenesis. Human Mutation. 2012;33(1):254-263
  317. 317. Marceca GP et al. Detecting and characterizing A-to-I microRNA editing in cancer. Cancers (Basel). 2021;13(7)
  318. 318. Ma R et al. MicroRNA polymorphism: A target for diagnosis and prognosis of hepatocellular carcinoma? Oncology Letters. 2021;21(4):324
  319. 319. Li Y et al. The associations and roles of microRNA single-nucleotide polymorphisms in cervical cancer. International Journal of Medical Sciences. 2021;18(11):2347-2354
  320. 320. Zhou C et al. Association of miR-146a, miR-149 and miR-196a2 polymorphisms with neuroblastoma risk in eastern Chinese population: A three-center case-control study. Bioscience Reports. 2019;39(6)
  321. 321. Wang K et al. A functional variation in pre-microRNA-196a is associated with susceptibility of esophageal squamous cell carcinoma risk in Chinese Han. Biomarkers. 2010;15(7):614-618
  322. 322. Sun G, Li H, Rossi JJ. Sequence context outside the target region influences the effectiveness of miR-223 target sites in the RhoB 3'UTR. Nucleic Acids Research. 2010;38(1):239-252
  323. 323. Kertesz M et al. The role of site accessibility in microRNA target recognition. Nature Genetics. 2007;39(10):1278-1284
  324. 324. Budmark A et al. Single nucleotide variations within and around microRNA-binding sites. Cancer Research. 2018;78(13):490-490
  325. 325. Berardi C et al. Multi-ethnic analysis reveals soluble L-selectin may be post-transcriptionally regulated by 3'UTR polymorphism: The multi-ethnic study of atherosclerosis (MESA). Human Genetics. 2015;134(4):393-403
  326. 326. Gibson G. Rare and common variants: Twenty arguments. Nature Reviews Genetics. 2012;13(2):135-145
  327. 327. Hermann T, Westhof E. Non-Watson-crick base pairs in RNA-protein recognition. Chemistry & Biology. 1999;6(12):R335-R343
  328. 328. Smalheiser NR, Torvik VI. Complications in mammalian microRNA target prediction. Methods in Molecular Biology. 2006;342:115-127
  329. 329. Kuc C et al. Rainbow trout exposed to benzo[a]pyrene yields conserved microRNA binding sites in DNA methyltransferases across 500 million years of evolution. Scientific Reports. 2017;7(1):16843
  330. 330. Jopling CL, Schutz S, Sarnow P. Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell Host & Microbe. 2008;4(1):77-85
  331. 331. Ivashchenko A et al. The properties of binding sites of miR-619-5p, miR-5095, miR-5096, and miR-5585-3p in the mRNAs of human genes. BioMed Research International. 2014;2014:720715
  332. 332. Koscianska E et al. Cooperation meets competition in microRNA-mediated DMPK transcript regulation. Nucleic Acids Research. 2015;43(19):9500-9518
  333. 333. Thibault PA et al. Regulation of hepatitis C virus genome replication by Xrn1 and MicroRNA-122 binding to individual sites in the 5' untranslated region. Journal of Virology. 2015;89(12):6294-6311
  334. 334. Nieder-Rohrmann A et al. Cooperative enhancement of translation by two adjacent microRNA-122/Argonaute 2 complexes binding to the 5′ untranslated region of hepatitis C virus RNA. The Journal of General Virology. 2017;98(2):212-224
  335. 335. Trobaugh DW et al. Cooperativity between the 3′ untranslated region microRNA binding sites is critical for the virulence of eastern equine encephalitis virus. PLoS Pathogens. 2019;15(10):e1007867
  336. 336. Briskin D, Wang PY, Bartel DP. The biochemical basis for the cooperative action of microRNAs. Proceedings of the National Academy of Sciences of the United States of America. 2020;117(30):17764-17774
  337. 337. Rinck A et al. The human transcriptome is enriched for miRNA-binding sites located in cooperativity-permitting distance. RNA Biology. 2013;10(7):1125-1135
  338. 338. Saetrom P et al. Distance constraints between microRNA target sites dictate efficacy and cooperativity. Nucleic Acids Research. 2007;35(7):2333-2342
  339. 339. Broderick JA et al. Argonaute protein identity and pairing geometry determine cooperativity in mammalian RNA silencing. RNA. 2011;17(10):1858-1869
  340. 340. Clifton H et al. The abundance of homologous MicroRNA-binding sites in the human c-MET mRNA 3’UTR. Biomedical Journal of Scientific & Technical Research. 2022;42(3):33543-33549
  341. 341. Bhattacharya A, Ziebarth JD, Cui Y. PolymiRTS database 3.0: Linking polymorphisms in microRNAs and their target sites with human diseases and biological pathways. Nucleic Acids Research. 2014;42(Database issue):D86-D91
  342. 342. Liu C et al. MirSNP, a database of polymorphisms altering miRNA target sites, identifies miRNA-related SNPs in GWAS SNPs and eQTLs. BMC Genomics. 2012;13:661
  343. 343. Bruno AE et al. miRdSNP: A database of disease-associated SNPs and microRNA target sites on 3'UTRs of human genes. BMC Genomics. 2012;13:44
  344. 344. Yue M et al. MSDD: A manually curated database of experimentally supported associations among miRNAs, SNPs and human diseases. Nucleic Acids Research. 2018;46(D1):D181-D185

Written By

Miruna Giurgiu, Robert Kaltenbach, Franziska Ahrend, Summer Weeks, Holly Clifton, Martin Bouldo, Vitaly Voloshin, Jiling Zhong, Siegfried Harden and Alexander Kofman

Submitted: 24 June 2023 Reviewed: 28 June 2023 Published: 14 August 2023