Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2017

Open Access 01.12.2017 | Review

Nuclear microRNAs in normal hemopoiesis and cancer

verfasst von: John E.J. Rasko, Justin J.-L. Wong

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2017

Abstract

Since the discovery of microRNAs (miRNAs) in the early 1990s, these small molecules have been increasingly recognized as key players in the regulation of critical biological processes. They have also been implicated in many diverse human diseases. The canonical function of miRNAs is to target the 3′ untranslated region (3′ UTR) of cytoplasmic messenger RNA to post-transcriptionally regulate mRNA and protein levels. It has now been shown that miRNAs can also bind to the promoter regions of genes or primary miRNA transcripts to regulate gene expression. Such observations have indicated the presence of miRNAs in the nucleus and implied additional non-canonical functions. Nevertheless, the role(s) of nuclear miRNAs in normal hemopoiesis and cancer remains elusive despite a burgeoning literature. Herein, we review current knowledge concerning the abundance and/or functions of nuclear miRNAs during blood cell development and cancer biology. We also discuss ongoing challenges in order to provoke further studies into identifying key roles for nuclear miRNAs in the development of other cell lineages and human cancers.
Abkürzungen
3′ UTR
3′ untranslated region
Ago
Argonaute
HPSE
Heparanase
lncRNAs
Long non-coding RNAs
miRNA
MicroRNA
mRNA Messenger RNA pre-miRNA
Precursor miRNA
pri-miRNA
Primary miRNA
RISC
RNA-induced silencing complex

Background

Non-coding RNAs are RNA molecules that are not translated into proteins but are nevertheless functional. They include long non-coding RNAs, intronic RNAs, circular RNAs, competing endogenous RNAs, microRNAs (miRNAs) and Piwi-interacting RNAs that are known to regulate gene expression at both the transcriptional and post-transcriptional levels [15]. Of all non-coding RNA species, miRNAs are best characterized in terms of their biogenesis and functions.
miRNAs are approximately 22 nucleotides in length. They play major roles in numerous biological processes including cell differentiation, lineage specification, reprogramming, immune response and the cell cycle [610]. Almost 30,000 miRNAs in 223 animal and plant species have been annotated in the miRNA database, miRBase (Release 21, June 2014). Even viruses, particularly herpesviruses, encode miRNAs to enhance their replication potential [11, 12]. The biological importance of miRNAs is further highlighted by the deregulation of miRNA expression in many diverse human diseases including cardiovascular [13], neuronal [14, 15], inflammatory [15], dermatological [16], hepatological [17] and malignant diseases [1820]. Given their key roles in many normal and disease-related processes, it is not surprising that miRNAs are enthusiastically viewed as potential druggable targets. To achieve this goal and anticipate side effects, it is important to understand their mechanisms of action and functions.
It is generally recognized that most mature miRNAs are localized in the cytoplasm along with the four catalytic Argonaute (Ago) proteins, where they contribute to the RNA-induced silencing complex (RISC) [2123]. It is via this RISC complex that miRNAs regulate gene expression by targeting messenger RNAs (mRNAs). The majority of binding sites for miRNAs are within the 3′ untranslated region (3′ UTR) of target mRNAs in animals, whereas they are often within coding regions in plants [24]. A mature miRNA can bind to the 3′ UTR of a target mRNA based on partial sequence complementarity between the two to initiate one of several mechanisms to reduce mRNA and/or protein levels. These mechanisms include repression of translational elongation, impairment of translation initiation, and decapping and deadenylation of mRNA, which are reviewed extensively elsewhere [2527].
Notably, numerous nuclear miRNAs have also been reported. These miRNAs may have diverse known or unknown non-canonical functions. Herein, we discuss how nuclear-localized miRNAs, although synthesized through biogenesis pathways identical to cytoplasmic miRNAs, could be shuttled back and retained in the nucleus to exert functions that differ from canonical miRNA actions. We focus especially on our current knowledge concerning the distribution and roles of these miRNAs in hemopoietic and cancer cells.

Evidence of nuclear-localized miRNAs

Systematic profiling analyses performed on nuclear and cytoplasmic-fractionated RNA samples have concluded that the majority of miRNAs are present in the nucleus [28, 29]. These results indicate that most, if not all, miRNAs have the capacity to shuttle between the nucleus and cytoplasm. This finding is supported by the localization of Ago proteins in the nucleus and a recent discovery that a component of the RISC complexes, TNRC6A, is a nuclear and cytoplasmic shuttling protein that facilitates Ago nuclear transport. [30, 31] In vitro knockdown of Importin 1 and 8 has also been shown to reduce the nuclear localization of Ago proteins and/or miRNAs, indicating that these transporter proteins mediate the translocation of Ago/miRNAs into the nucleus (see reviews by Liang et al. [32] and Roberts [33], for details) [32, 33].
In addition to nuclear import mechanisms, there are also miRNA-intrinsic aspects that facilitate nuclear localization. The first evidence was provided by the predominant localization of miR-29b in the nuclei of HeLa and NIH3T3 cells, as directed by a hexanucleotide motif (AGUGUU) at its 3′ terminus [34]. A related miRNA, miR-29a, lacked this hexanucleotide motif and is enriched in the cytoplasm [34]. Notably, a synthetic siRNA harboring the miR-29a sequence engineered to include the AGUGUU motif could be directed into the nucleus, indicating the importance of this motif for nuclear localization [34]. Hwang and co-workers further identified variants of this motif that is associated with the nuclear localization of miRNAs, including UGUGUU, ACUGUU, AGAGUU, AGUCUU, AGUGAU, AGUGUA, AGNGUN [35]. An independent study by Jeffries and co-workers confirmed the presence of these motifs in seven other nuclear-localized miRNAs: miR-30b, miR-30c, miR-19a, miR-374a, miR-374b, miR-590-5p and miR-193b [36].
However, these motifs alone may not be sufficient to direct nuclear localization of miRNAs [28]. For example, miR-92b that possesses a hexanucleotide motif identical to the nuclear-localized miR-29b has been reported to be cytoplasmically enriched [28]. A recent study also failed to identify a relationship between nuclear or cytoplasmic enrichment of miRNAs and their seed sequences, suggesting that seed identity itself is insufficient to determine their predominant localization [36]. As yet unknown sequence-independent factors may exist to retain, detain or reimport specific miRNAs in the nucleus, and they may also be cell-type specific.
Notwithstanding the lack of clarity regarding mechanisms of nuclear miRNA localization, numerous studies have demonstrated their enrichment and functions in myriad mammalian cell types [3745]. The functions of these nuclear miRNAs include regulation of gene and long non-coding RNA expression [37, 38, 46], controlling the biogenesis of other miRNAs [41] and fine-tuning the expression of mRNA expression in the cytoplasm [43] (Fig. 1). Many of these functions are relevant to hemopoiesis and cancer as outlined in the following sections.

Nuclear-localized miRNAs in hemopoietic cells

Hemopoiesis is one of the first processes in which the functions of miRNAs were elucidated [47, 48]. Lineage commitment towards blood cell production begins with the hemopoietic stem cell escaping quiescence and the stepwise acquisition of specific myeloid or lymphoid identities [49, 50]. The process involves gradual changes in the expression patterns of hundreds of proteins, including transcription factors [49, 50] and cell cycle regulators [51], many of which are direct or indirect targets of miRNAs.
More than a decade ago, three miRNAs, miR-181a, miR-223 and miR-142 were recognized as key players in myeloid and lymphoid cell differentiation [47]. miR-181a is preferentially expressed in the B-lineage, miR-223 in the myeloid lineage and mir-142 in both B and myeloid lineages [47]. Remarkably, enforced expression of miR-181a led to a doubling of B cells while persistent expression of miR-223 or miR-142 increased the number of T cells by 30–40% [47]. Subsequently, miR-181a has also been found to be crucial for T cell development and function, modulating T cell receptor signalling, in part through its role in the downregulation of multiple phosphatases including SHP2, PTPN22, DUSP5 and DUSP6 [52]. Other confirmed targets of miR-181a include TCR, CD69 and BCL-2, all of which are regulated during T cell development [53].
Given the substantial literature on the role of miRNAs in hemopoiesis, studies that determined the cellular localization of miRNAs in hemopoietic cells are relatively few. Most studies have focused on the canonical roles of miRNAs in the post-transcriptional regulation of genes involved in hemopoiesis. The best examples of nuclear miRNA functions have been reported in the context of granulopoiesis (Table 1).
Table 1
Nuclear-localized miRNAs in normal hemopoiesis
Nuclear miRNA
Lineage/cell type
Function/activity
Significance
Reference
miR-223
Myeloid
Binds to the promoter region of NF1A to induce transcriptional silencing
Repression of NF1A to promote granulopoiesis
[42]
miR-706
Myeloid (LinSca+Kit+ hemopoietic stem/progenitor cells, promyelocytes, myelocytes and granulocytes), MPRO, EL4, MEL, A20
Detained in the nucleus to derepress target mRNA (e.g. Stat1)
Increased expression of transcription factor (e.g. Stat1) to promote granulopoiesis
[43, 57]
miR-690
Myeloid (LinSca+Kit+ hemopoietic stem/progenitor cells, promyelocytes, myelocytes and granulocytes), MPRO, EL4, MEL, A20
Unknown
Unknown
[43]
miR-709
Myeloid (LinSca+Kit+ hemopoietic stem/progenitor cells, promyelocytes, myelocytes and granulocytes), MPRO, EL4, MEL, A20
Unknown
Unknown
[43]
miR-467a*
LinSca+Kit+ hemopoietic stem/progenitor cells, MPRO, EL4, A20
Unknown
Unknown
[43]
The myeloid specific miRNA, miR-223, has been well characterized as playing an essential role in the control of granulopoiesis. In the cytoplasm of myeloid progenitors, miR-223 targets transcription factors MEF2C and NF1A that usually promote myeloid progenitor cell proliferation [54, 55]. The inhibition of cell proliferation in myeloid progenitors coincides with their differentiation into granulocytes. Notably, miR-223 also targets seed sequences in the NF1A promoter to induce transcriptional gene silencing via recruitment of the polycomb repressive complex and a consequent increase in DNA methylation levels [42]. These data indicate that both nuclear and cytoplasmic miR-223 work synergistically to silence NF1A during granulopoiesis [42, 54].
Our group has recently reported the enrichment of miRNAs including miR-690, miR-706 and miR-709 in the nucleus of primary LinSca+Kit+ hemopoietic stem/progenitor cells, promyelocytes, myelocytes and granulocytes [43]. We and others have also found miR-690 and/or miR-709 to be nuclear-enriched in various human and mouse cell lines including MPRO, EL4, MEL, A20, L929 and HEK-293T indicating that their functions may be localized to the nucleus [41, 43]. One of these miRNAs, miR-709, has been reported to bind with perfect complementarity to pri-miR-15a and pri-miR-16-1 and inhibit the biogenesis of these miRNAs [41]. This finding has established a previously unanticipated role of nuclear miRNAs in regulating or fine-tuning the expression of other miRNAs.
Our in silico analysis predicted many putative primary miRNA (pri-miRNA) sequences that could be targeted by miR-690, miR-706 or miR-709 with perfect complementarity [43]. Relevant to myelopoiesis, miR-706 is predicted to bind perfectly to a hemopoeitic-specific miR-142-3p, which is known to play an essential role in granulocyte homeostasis and maturation [43, 56]. Nevertheless, we were not able to demonstrate an increase of pri-miR-142-3p processing following inhibition of miR-706 with a hairpin inhibitor against this miRNA [43]. Our result indicates the complexity of factors that determine miRNA binding to its targets in the nucleus, which are not likely to be dependent on sequences alone.
Importantly, we further reported that miRNAs may be retained in the nucleus to fine-tune the expression of mRNA targets [43]. For example, miR-706 enrichment in the nucleus is associated with decreased cytoplasmic miR-706 expression. Consequently, the expression of its targets such as the myeloid transcription factors, Stat1, increases to promote myeloid differentiation [43, 57].

Nuclear-localized miRNAs in cancer

The nuclear localization of miRNAs in cancer cells is well documented. Cancer cell lines including the 5-8F nasopharyngeal carcinoma cells, the HCT116 colorectal cancer cells and the THP-1 acute monocytic leukaemia cells harbor hundreds of miRNAs that are enriched in the nucleus of these cells [28, 29, 58]. The nuclear-specific functions of many of these miRNAs remain elusive, but they are likely to regulate or fine-tune the expression of cancer-associated genes. For example, miR-10a, which is nuclear-localized in both the HCT116 and THP-1 cell lines [29, 58], has been reported to inhibit the transcription of Hoxd4 in the breast cancer cell lines, MCF7 and MDA-MB-231 [39]. Nuclear-localized miR-10a binds with near perfect complementarity to the promoter of this tumor invasion and metastasis-promoting gene to trigger its silencing via hypermethylation and trimethylation of histone 3 lysine 27 at its promoter [39]. This example demonstrates that nuclear enrichment of miR-10a may provide a therapeutic opportunity to modulate gene expression relevant to cancer metastasis.
Other nuclear-localized miRNAs have been reported to promote transcriptional activation of oncogenes by binding to their promoters. Examples include miR-483 that binds to the promoter of IGF2 to increase its expression in Wilms’ tumors [59] and miR-558 that binds to the promoter of heparanase (HPSE) to enhance its expression, resulting in enhanced tumor growth in neuroblastoma cells [44] (Table 2).
Table 2
Function of nuclear-localized miRNAs relevant to human cancers
Nuclear miRNA
Function
Cancer cells
Reference
miR-10a
Inhibit Hoxd4 to control cancer metastasis
Breast cancer cell lines: MCF7 and MDA-MB-231
[39]
miR-483
Activate IGF2 to enhance tumorigenesis
Wilms’ tumors
[59]
miR-558
Activate HPSE to promote tumor growth
Neuroblastoma cell lines: SK-N-SH, SK-N-AS, SH-SY5Y and SK-N-BE(2)
[44]
miR-373
Increase expression of CDH1 tumor suppressor gene
Prostate cancer cell line: PC3
[38]
miR-124
Activate tumor suppressor gene, P27
Breast and ovarian cancer cell lines: MDA-MB-231, HeyA8, SKOV3.ip1, SKBr3, OVCAR-3, BxPC-3 and L3.6pl, MIA PaCa 2, Panc1, U87, SNB19 and LN229
[60].
miR-205
Activate interleukin tumor suppressor genes, IL24 and IL32
Prostate cancer cell lines: PC3, LNCaP and Du145
[45].
miR-774
Promote transcription of Ccnb1
Mouse prostate adenocarcinoma cell line: TRAMP C1
[61]
miR-370
Activate tumor suppressor gene, P21
Bladder cancer cell lines: T24 and EJ
[76]
miR-1180
Activate tumor suppressor gene, P21
Bladder cancer cell lines: T24 and EJ
[76]
miR-1236
Activate tumor suppressor gene, P21
Bladder cancer cell lines: T24 and EJ
[76]
miR-939
Repression of Bcl-xl anti-apoptosis gene
Human neuroblastoma cell line: SH-SY5Y
[62]
Nuclear-localized miRNAs can also protect against tumorigenesis by promoting the activation of tumor suppressor genes. For example, miR-373 binds to the promoter of CDH1 to increase its expression in the prostate cancer cell line, PC3 [38]. miR-124 promotes the activation of P27, leading to G1 arrest in myriad breast and ovarian cancer cell lines [60]. miR-205 induces the expression of the interleukin tumor suppressor genes, IL24 and IL32, by targeting specific regions of their promoters [45]. Additional examples are provided in Table 2.
The mechanisms by which binding of miRNAs to gene promoters results in transcriptional activation or silencing have also been described. They include increased or decreased levels of histone modifications associated with gene activation [39, 45, 61], altered RNA Polymerase II activity [38, 61], enhanced recruitment of transcription factors [59] and inhibition of transcription factor binding due to the presence of decoy miRNAs [62].
Besides coding genes, cancer-associated nuclear long non-coding RNAs (lncRNAs), MALAT-1 and XIST, have been reported as a target of miR-9 and miR-210, respectively [63, 64]. The HOTAIR lncRNA, which localizes to both the nucleus and cytoplasm, has also been reported as a target of miR34a in prostate cancer cells [65]. However, it is unknown whether this interaction occurs in the nucleus. The GENCODE Consortium has mapped over 10,000 lncRNAs as putative miRNA targets, many of which are nuclear-enriched [66]. Further experimental validations will no doubt identify more nuclear lncRNAs as miRNA targets that are relevant to cancer.
Surprisingly, no report has previously reviewed the role of nuclear miRNAs in haematological malignancies despite the known roles of miRNAs as oncogenes or tumor suppressor genes [18, 67]. miR-15a and miR-16-1 are established tumor suppressor miRNAs in chronic lymphocytic leukaemia. Their biogenesis is known to be regulated by the nuclear-localized miR-709 in liver cells [41, 67]. Whether or not miR-709 fine-tunes the expression of miR-15a and miR-16 in chronic lymphocytic leukaemia remains to be determined. As described in the previous section of this review, nuclear miR-223 has a key role in normal granulopoiesis. A decrease in miR-223 expression level is associated with CEBPA-mutated acute myeloid leukaemia [68]. Inhibition of the miR-223 target gene, E2F1, by CEBPA is pivotal to prevent leukemogenesis that results from E2F1-mediated expression of the oncogene tribble (Trib) 2 gene [68, 69]. As such both miR-223 and CEBPA regulate the expression of E2F1 in myeloid cells under normal physiological condition. Whether aberrant nuclear detention of miR-223 can occur to derepress E2F1 in CEBPA mutant leukaemia demands further investigation.

Challenges in determining the functions of nuclear miRNAs

The role of nuclear-localized miRNAs has been relatively neglected as evidenced by the extensive literature on miRNAs, including those relevant to hemopoiesis and cancer. Indeed, the majority of studies have focused on the action of miRNAs in post-transcriptional gene regulation. Several reports have considered miRNAs’ role in transcriptional regulation of specific mRNAs only when these miRNAs did not act canonically via 3′ UTR targeting [39, 61]. Others have focused on the transcriptional regulation of specific genes by miRNAs because these genes are known to be transcriptionally regulated by other synthetic or endogenous double-stranded small RNAs [37, 38]. In order to promote more rapid elucidation of the nuclear-associated roles of thousands of miRNAs, studies should specifically seek to identify nuclear miRNAs and their functions, notwithstanding the technical challenges.
Most studies reporting nuclear-localized miRNAs have utilized nuclear and cytoplasmic fractionation methods to determine their localization. Inevitably, the difficulty in obtaining a perfectly pure nuclear fraction has provoked scepticism concerning the accuracy of reported studies. Ensuring the removal of the cytoplasmic fraction prior to collection of the nuclear fraction is crucial and not always possible with some cells. It is also important to perform validations to show the absence of contaminating factors by western blot and RT-qPCR in conjunction with microscopy-based detection of nuclear miRNA labelled with fluorescence probes. These experimental procedures are laborious and often require cell-type specific optimizations to obtain reliable results. This is particularly relevant for commercially available nuclear and cytoplasmic extraction kits typically optimized for use with commonly studied cell lines such as HeLa. In our recent work, we undertook considerable optimization steps beyond the manufacturer’s protocol to obtain good quality nuclear and cytoplasmic fractions from primary cells [43].
Upon identification of nuclear miRNAs, one next logical step is to determine where they bind. Bioinformatic tools have been developed to predict targets of miRNAs within gene promoters or pri-miRNAs [70, 71]. Nearly 800,000 miRNA seed sequences match over 27,000 promoter sequences [71]. miRNAs can also target gene promoters through non-seed related complementarity [45], indicating that miRNA binding to promoter regions may be more widespread than previously thought.
However, similar to the complexity of 3′ UTR targeting by miRNAs, predicted miRNA-promoter/pri-miRNA pairing often did not result in the expected functional changes or occur in a cell-type specific manner [38, 43]. Thus, cause-effect experiments are necessary to confirm that the binding of miRNAs to genomic sequences in the nucleus results in functional consequences.
Experiments designed to reduce the expression of miRNAs that localized specifically to the nucleus are technically challenging. Indeed, our recent report shows the dominant cytoplasmic localization of anti-miR-706 when transfected into myeloid cells [43]. In a study that reported the inhibition of miR-15a and miR-16 processing by miR-709 in mouse L929 liver cells, modest upregulation of miR-15a and miR-16 (<2-fold) was detected in anti-miR-709 transfected cells [41]. It has not been shown whether anti-miR-709 entered the nucleus to inhibit miRNA processing. Thus, a direct role of nuclear miR-709 in controlling the expression of other miRNAs remains elusive.
A recent study has reported the utility of a snoRNA-based vector (snoVector) that allows efficient nuclear retention of RNA molecules processed from this vector [72]. RNA sequences such as lncRNAs, coding mRNAs and precursor miRNAs (pre-miRNAs) can be inserted into the snoVector. They can subsequently be processed into functional RNAs via the endogenous snoRNA processing machinery. RNA molecules expressed using snoVector have been reported to be nuclear-enriched, including those that are typically localized to the cytoplasm [72]. A similar vector (snoMEN) created via manipulation of the human C/D small nuclear RNA HBII-180C has also been reported to target nuclear RNA [73]. It has been utilized to express interfering RNAs that effectively reduced the expression of complementary sequences including nuclear pre-mRNA and pri-miRNA [74, 75]. Thus, it may be possible to use snoVector or SnoMEN to constrain anti-miRNA sequences to the nucleus. This step may facilitate functional characterization following specific and efficient repression of nuclear miRNAs.
Nonetheless, it is important to recognize that miRNAs have been reported to shuttle from the cytoplasm into the nucleus. So, even if repression of specific nuclear miRNAs is achieved, the observed functional loss may be due to the overall depletion of cytoplasmic miRNAs that are inhibited when they enter the nucleus. In such a case, it would be very challenging to discern the specific role of a given nuclear-localized miRNA. Optimal experiment design should exclude changes in cytoplasmic miRNA expression as a contributor to any phenotypic alteration. Alternatively, despite experimental hurdles, it is necessary to distinguish the function of any given nuclear miRNA from its cytoplasmic counterpart.

Conclusions

The importance of miRNAs in hemopoiesis and cancer through the post-transcriptional regulation of the expression of relevant genes has been well-established. In recent years, non-canonical roles for nuclear-localized miRNAs have been uncovered. While there is evidence that nuclear miRNAs regulate the transcription of specific genes in hemopoietic and cancer cells, these examples are relatively few in the context of the vast published literature on miRNAs in these cell types. In this review, we have described nuclear miRNAs that promote granulopoieis and those that enhance the expression of tumor suppressor genes and oncogenes. The roles of nuclear miRNAs in the vast majority of myeloid and lymphoid cells remain to be determined. Their roles in cancer, including post-transcriptional silencing of tumor suppressor genes, is as yet unreported. Therefore, examination of the functions of these nuclear miRNAs should prove to be a fruitful area for further research.

Acknowledgements

Not applicable.

Funding

JEJR and JJ-LW received funding from the National Health and Medical Research Council of Australia (Grant Nos. 1061906 to JEJR, 1080530 and 1128175 to JEJR and JJ-LW, and 1126306 to JJ-LW). JEJR is funded by the Cancer Council of NSW, Cure the Future and an anonymous foundation. JJ-LW holds a Fellowship from the Cancer Institute of NSW.

Availability of data and materials

Data sharing is not applicable to this article as no dataset was generated or analyzed in this review.

Authors’ contributions

JEJR and JJ-LW wrote the manuscript. Both authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Sanchez-Mejias A, Tay Y. Competing endogenous RNA networks: tying the essential knots for cancer biology and therapeutics. J Hematol Oncol. 2015;8:30.CrossRefPubMedPubMedCentral Sanchez-Mejias A, Tay Y. Competing endogenous RNA networks: tying the essential knots for cancer biology and therapeutics. J Hematol Oncol. 2015;8:30.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Chen L-L. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17:205–11.CrossRefPubMed Chen L-L. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17:205–11.CrossRefPubMed
6.
Zurück zum Zitat Bueno M, Malumbres M. MicroRNAs and the cell cycle. BBA-Mol Basis Dis. 2011;1812:592–601.CrossRef Bueno M, Malumbres M. MicroRNAs and the cell cycle. BBA-Mol Basis Dis. 2011;1812:592–601.CrossRef
7.
Zurück zum Zitat Clancy JL, Patel HR, Hussein SMI, Tonge PD, Cloonan N, Corso AJ, Li M, Lee D-S, Shin J-Y, Wong JJL, Bailey CG, Benevento M, Munoz J, Chuah A, Wood D, Rasko JEJ, Heck AJR, Grimmond SM, Rogers IM, Seo J-S, Wells CA, Puri MC, Nagy A, Preiss T. Small RNA changes en route to distinct cellular states of induced pluripotency. Nat Commun. 2014;5:5522.CrossRefPubMed Clancy JL, Patel HR, Hussein SMI, Tonge PD, Cloonan N, Corso AJ, Li M, Lee D-S, Shin J-Y, Wong JJL, Bailey CG, Benevento M, Munoz J, Chuah A, Wood D, Rasko JEJ, Heck AJR, Grimmond SM, Rogers IM, Seo J-S, Wells CA, Puri MC, Nagy A, Preiss T. Small RNA changes en route to distinct cellular states of induced pluripotency. Nat Commun. 2014;5:5522.CrossRefPubMed
8.
Zurück zum Zitat Ivey KN, Srivastava D. MicroRNAs as regulators of differentiation and cell fate decisions. Cell Stem Cell. 2010;7:36–41.CrossRefPubMed Ivey KN, Srivastava D. MicroRNAs as regulators of differentiation and cell fate decisions. Cell Stem Cell. 2010;7:36–41.CrossRefPubMed
9.
Zurück zum Zitat Leonardo TR, Schultheisz HL, Loring JF, Laurent LC. The functions of microRNAs in pluripotency and reprogramming. Nat Cell Biol. 2012;14:1114–21.CrossRefPubMed Leonardo TR, Schultheisz HL, Loring JF, Laurent LC. The functions of microRNAs in pluripotency and reprogramming. Nat Cell Biol. 2012;14:1114–21.CrossRefPubMed
11.
Zurück zum Zitat Nukui M, Mori Y, Murphy EA. A human herpesvirus 6A-encoded microRNA: role in viral lytic replication. J Virol. 2015;89:2615–27.CrossRefPubMed Nukui M, Mori Y, Murphy EA. A human herpesvirus 6A-encoded microRNA: role in viral lytic replication. J Virol. 2015;89:2615–27.CrossRefPubMed
12.
Zurück zum Zitat Yan Q, Li W, Tang Q, Yao S, Lv Z, Feng N, Ma X, Bai Z, Zeng Y, Qin D, Lu C. Cellular microRNAs 498 and 320d regulate herpes simplex virus 1 induction of Kaposi’s sarcoma-associated herpesvirus lytic replication by targeting RTA. PLoS ONE. 2013;8:e55832.CrossRefPubMedPubMedCentral Yan Q, Li W, Tang Q, Yao S, Lv Z, Feng N, Ma X, Bai Z, Zeng Y, Qin D, Lu C. Cellular microRNAs 498 and 320d regulate herpes simplex virus 1 induction of Kaposi’s sarcoma-associated herpesvirus lytic replication by targeting RTA. PLoS ONE. 2013;8:e55832.CrossRefPubMedPubMedCentral
13.
14.
Zurück zum Zitat Haramati S, Chapnik E, Sztainberg Y, Eilam R, Zwang R, Gershoni N, McGlinn E, Heiser PW, Wills A-M, Wirguin I, Rubin LL, Misawa H, Tabin CJ, Brown R, Chen A, Hornstein E. miRNA malfunction causes spinal motor neuron disease. Proc Natl Acad Sci U S A. 2010;107:13111–6.CrossRefPubMedPubMedCentral Haramati S, Chapnik E, Sztainberg Y, Eilam R, Zwang R, Gershoni N, McGlinn E, Heiser PW, Wills A-M, Wirguin I, Rubin LL, Misawa H, Tabin CJ, Brown R, Chen A, Hornstein E. miRNA malfunction causes spinal motor neuron disease. Proc Natl Acad Sci U S A. 2010;107:13111–6.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Andersen HH, Duroux M, Gazerani P. MicroRNAs as modulators and biomarkers of inflammatory and neuropathic pain conditions. Neurobiol Dis. 2014;71:159–68.CrossRefPubMed Andersen HH, Duroux M, Gazerani P. MicroRNAs as modulators and biomarkers of inflammatory and neuropathic pain conditions. Neurobiol Dis. 2014;71:159–68.CrossRefPubMed
16.
Zurück zum Zitat Jinnin M. Recent progress in studies of miRNA and skin diseases. J Dermatol. 2015;42:551–8.CrossRefPubMed Jinnin M. Recent progress in studies of miRNA and skin diseases. J Dermatol. 2015;42:551–8.CrossRefPubMed
18.
Zurück zum Zitat Gordon JEA, Wong JJL, Rasko JEJ. MicroRNAs in myeloid malignancies. Brit J Haematol. 2013;162:162–76.CrossRef Gordon JEA, Wong JJL, Rasko JEJ. MicroRNAs in myeloid malignancies. Brit J Haematol. 2013;162:162–76.CrossRef
21.
Zurück zum Zitat Mourelatos Z, Dostie J, Paushkin S, Sharma A, Charroux B, Abel L, Rappsilber J, Mann M, Dreyfuss G. miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs. Genes Dev. 2002;16:720–8.CrossRefPubMedPubMedCentral Mourelatos Z, Dostie J, Paushkin S, Sharma A, Charroux B, Abel L, Rappsilber J, Mann M, Dreyfuss G. miRNPs: a novel class of ribonucleoproteins containing numerous microRNAs. Genes Dev. 2002;16:720–8.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science. 2001;293:1146–50.CrossRefPubMed Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science. 2001;293:1146–50.CrossRefPubMed
23.
24.
Zurück zum Zitat Millar AA, Waterhouse PM. Plant and animal microRNAs: similarities and differences. Funct Integr Genomics. 2005;5:129–35.CrossRefPubMed Millar AA, Waterhouse PM. Plant and animal microRNAs: similarities and differences. Funct Integr Genomics. 2005;5:129–35.CrossRefPubMed
25.
Zurück zum Zitat Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 2013;12:847–65.CrossRefPubMedPubMedCentral Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 2013;12:847–65.CrossRefPubMedPubMedCentral
26.
27.
Zurück zum Zitat Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16:421–33.CrossRefPubMed Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16:421–33.CrossRefPubMed
28.
Zurück zum Zitat Liao J-Y, Ma L-M, Guo Y-H, Zhang Y-C, Zhou H, Shao P, Chen Y-Q, Qu L-H. Deep sequencing of human nuclear and cytoplasmic small RNAs reveals an unexpectedly complex subcellular distribution of miRNAs and tRNA 3′ trailers. PLoS ONE. 2010;5:e10563.CrossRefPubMedPubMedCentral Liao J-Y, Ma L-M, Guo Y-H, Zhang Y-C, Zhou H, Shao P, Chen Y-Q, Qu L-H. Deep sequencing of human nuclear and cytoplasmic small RNAs reveals an unexpectedly complex subcellular distribution of miRNAs and tRNA 3′ trailers. PLoS ONE. 2010;5:e10563.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Park CW, Zeng Y, Zhang X, Subramanian S, Steer CJ. Mature microRNAs identified in highly purified nuclei from HCT116 colon cancer cells. RNA Biol. 2010;7:606–14.CrossRefPubMedPubMedCentral Park CW, Zeng Y, Zhang X, Subramanian S, Steer CJ. Mature microRNAs identified in highly purified nuclei from HCT116 colon cancer cells. RNA Biol. 2010;7:606–14.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Nishi K, Nishi A, Nagasawa T, Ui-Tei K. Human TNRC6A is an Argonaute-navigator protein for microRNA-mediated gene silencing in the nucleus. RNA. 2013;19:17–35.CrossRefPubMedPubMedCentral Nishi K, Nishi A, Nagasawa T, Ui-Tei K. Human TNRC6A is an Argonaute-navigator protein for microRNA-mediated gene silencing in the nucleus. RNA. 2013;19:17–35.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Liang H, Zhang J, Zen K, Zhang C-Y, Chen X. Nuclear microRNAs and their unconventional role in regulating non-coding RNAs. Protein Cell. 2013;4:325–30.CrossRefPubMedPubMedCentral Liang H, Zhang J, Zen K, Zhang C-Y, Chen X. Nuclear microRNAs and their unconventional role in regulating non-coding RNAs. Protein Cell. 2013;4:325–30.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Hwang H-W, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science. 2007;315:97–100.CrossRefPubMed Hwang H-W, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science. 2007;315:97–100.CrossRefPubMed
35.
Zurück zum Zitat Hwang H-W, Wentzel EA, Mendell JT. Nucleotide motifs providing localization elements and methods of use. Geneva: World Intellectual Property Organization. 2007. Patent No. WO 2007/149521 A2. Hwang H-W, Wentzel EA, Mendell JT. Nucleotide motifs providing localization elements and methods of use. Geneva: World Intellectual Property Organization. 2007. Patent No. WO 2007/149521 A2.
37.
Zurück zum Zitat Kim DH, Sætrom P, Snøve O, Rossi JJ. MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci U S A. 2008;105:16230–5.CrossRefPubMedPubMedCentral Kim DH, Sætrom P, Snøve O, Rossi JJ. MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci U S A. 2008;105:16230–5.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Place RF, Li L-C, Pookot D, Noonan EJ, Dahiya R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A. 2008;105:1608–13.CrossRefPubMedPubMedCentral Place RF, Li L-C, Pookot D, Noonan EJ, Dahiya R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A. 2008;105:1608–13.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Tan Y, Zhang B, Wu T, Skogerbø G, Zhu X, Guo X, He S, Chen R. Transcriptional inhibiton of Hoxd4 expression by miRNA-10a in human breast cancer cells. BMC Mol Biol. 2009;10:12.CrossRefPubMedPubMedCentral Tan Y, Zhang B, Wu T, Skogerbø G, Zhu X, Guo X, He S, Chen R. Transcriptional inhibiton of Hoxd4 expression by miRNA-10a in human breast cancer cells. BMC Mol Biol. 2009;10:12.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Younger ST, Corey DR. Transcriptional gene silencing in mammalian cells by miRNA mimics that target gene promoters. Nucleic Acids Res. 2011;39:5682–91.CrossRefPubMedPubMedCentral Younger ST, Corey DR. Transcriptional gene silencing in mammalian cells by miRNA mimics that target gene promoters. Nucleic Acids Res. 2011;39:5682–91.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Tang R, Li L, Zhu D, Hou D, Cao T, Gu H, Zhang J, Chen J, Zhang C-Y, Zen K. Mouse miRNA-709 directly regulates miRNA-15a/16-1 biogenesis at the posttranscriptional level in the nucleus: evidence for a microRNA hierarchy system. Cell Res. 2011;22:504–15.CrossRefPubMedPubMedCentral Tang R, Li L, Zhu D, Hou D, Cao T, Gu H, Zhang J, Chen J, Zhang C-Y, Zen K. Mouse miRNA-709 directly regulates miRNA-15a/16-1 biogenesis at the posttranscriptional level in the nucleus: evidence for a microRNA hierarchy system. Cell Res. 2011;22:504–15.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Zardo G, Ciolfi A, Vian L, Starnes LM, Billi M, Racanicchi S, Maresca C, Fazi F, Travaglini L, Noguera N, Mancini M, Nanni M, Cimino G, Lo-Coco F, Grignani F, Nervi C. Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood. 2012;119:4034–46.CrossRefPubMed Zardo G, Ciolfi A, Vian L, Starnes LM, Billi M, Racanicchi S, Maresca C, Fazi F, Travaglini L, Noguera N, Mancini M, Nanni M, Cimino G, Lo-Coco F, Grignani F, Nervi C. Polycombs and microRNA-223 regulate human granulopoiesis by transcriptional control of target gene expression. Blood. 2012;119:4034–46.CrossRefPubMed
43.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Qu H, Zheng L, Pu J, Mei H, Xiang X, Zhao X, Li D, Li S, Mao L, Huang K, Tong Q. miRNA-558 promotes tumorigenesis and aggressiveness of neuroblastoma cells through activating the transcription of heparanase. Hum Mol Genet. 2015;24:2539–51.CrossRefPubMed Qu H, Zheng L, Pu J, Mei H, Xiang X, Zhao X, Li D, Li S, Mao L, Huang K, Tong Q. miRNA-558 promotes tumorigenesis and aggressiveness of neuroblastoma cells through activating the transcription of heparanase. Hum Mol Genet. 2015;24:2539–51.CrossRefPubMed
45.
Zurück zum Zitat Majid S, Dar AA, Saini S, Yamamura S, Hirata H, Tanaka Y, Deng G, Dahiya R. MicroRNA-205-directed transcriptional activation of tumor suppressor genes in prostate cancer. Cancer. 2010;116:5637–49.CrossRefPubMedPubMedCentral Majid S, Dar AA, Saini S, Yamamura S, Hirata H, Tanaka Y, Deng G, Dahiya R. MicroRNA-205-directed transcriptional activation of tumor suppressor genes in prostate cancer. Cancer. 2010;116:5637–49.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, Kjems J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011;30:4414–22.CrossRefPubMedPubMedCentral Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, Kjems J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011;30:4414–22.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Chen C-Z, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83–6.CrossRefPubMed Chen C-Z, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83–6.CrossRefPubMed
48.
Zurück zum Zitat Garzon R, Croce CM. MicroRNAs in normal and malignant hematopoiesis. Curr Opin Hematol. 2008;15:352–8.CrossRefPubMed Garzon R, Croce CM. MicroRNAs in normal and malignant hematopoiesis. Curr Opin Hematol. 2008;15:352–8.CrossRefPubMed
49.
Zurück zum Zitat Rosenbauer F, Tenen DG. Transcription factors in myeloid development: balancing differentiation with transformation. Nat Rev Immunol. 2007;7:105–17.CrossRefPubMed Rosenbauer F, Tenen DG. Transcription factors in myeloid development: balancing differentiation with transformation. Nat Rev Immunol. 2007;7:105–17.CrossRefPubMed
50.
Zurück zum Zitat Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, McConkey ME, Habib N, Yosef N, Chang CY, Shay T, Frampton GM, Drake ACB, Leskov I, Nilsson B, Preffer F, Dombkowski D, Evans JW, Liefeld T, Smutko JS, Chen J, Friedman N, Young RA, Golub TR, Regev A, Ebert BL. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell. 2011;144:296–309.CrossRefPubMedPubMedCentral Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, McConkey ME, Habib N, Yosef N, Chang CY, Shay T, Frampton GM, Drake ACB, Leskov I, Nilsson B, Preffer F, Dombkowski D, Evans JW, Liefeld T, Smutko JS, Chen J, Friedman N, Young RA, Golub TR, Regev A, Ebert BL. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell. 2011;144:296–309.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Passeguë E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med. 2005;202:1599–611.CrossRefPubMedPubMedCentral Passeguë E, Wagers AJ, Giuriato S, Anderson WC, Weissman IL. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med. 2005;202:1599–611.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Li Q-J, Chau J, Ebert PJR, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P, Klein LO, Davis MM, Chen C-Z. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–61.CrossRefPubMed Li Q-J, Chau J, Ebert PJR, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P, Klein LO, Davis MM, Chen C-Z. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–61.CrossRefPubMed
53.
Zurück zum Zitat Neilson JR, Zheng GXY, Burge CB, Sharp PA. Dynamic regulation of miRNA expression in ordered stages of cellular development. Genes Dev. 2007;21:578–89.CrossRefPubMedPubMedCentral Neilson JR, Zheng GXY, Burge CB, Sharp PA. Dynamic regulation of miRNA expression in ordered stages of cellular development. Genes Dev. 2007;21:578–89.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C, Bozzoni I. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPα regulates human granulopoiesis. Cell. 2005;123:819–31.CrossRefPubMed Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C, Bozzoni I. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPα regulates human granulopoiesis. Cell. 2005;123:819–31.CrossRefPubMed
55.
Zurück zum Zitat Johnnidis JB, Harris MH, Wheeler RT, Stehling-Sun S, Lam MH, Kirak O, Brummelkamp TR, Fleming MD, Camargo FD. Regulation of progenitor cell proliferation and granulocyte function by microRNA-223. Nature. 2008;451:1125–9.CrossRefPubMed Johnnidis JB, Harris MH, Wheeler RT, Stehling-Sun S, Lam MH, Kirak O, Brummelkamp TR, Fleming MD, Camargo FD. Regulation of progenitor cell proliferation and granulocyte function by microRNA-223. Nature. 2008;451:1125–9.CrossRefPubMed
56.
Zurück zum Zitat Fan H-B, Liu Y-J, Wang L, Du T-T, Dong M, Gao L, Meng Z-Z, Jin Y, Chen Y, Deng M, Yang H-T, Jing Q, Gu A-H, Liu T-X, Zhou Y. miR-142-3p acts as an essential modulator of neutrophil development in zebrafish. Blood. 2014;124:1320–30.CrossRefPubMed Fan H-B, Liu Y-J, Wang L, Du T-T, Dong M, Gao L, Meng Z-Z, Jin Y, Chen Y, Deng M, Yang H-T, Jing Q, Gu A-H, Liu T-X, Zhou Y. miR-142-3p acts as an essential modulator of neutrophil development in zebrafish. Blood. 2014;124:1320–30.CrossRefPubMed
57.
Zurück zum Zitat Hegde VL, Nagarkatti P, Nagarkatti M. MicroRNAs and their role in the generation of myeloid derived suppressor cells (MDSC) by cannabidiol in vivo. J Immunol. 2012;188:48.16. Hegde VL, Nagarkatti P, Nagarkatti M. MicroRNAs and their role in the generation of myeloid derived suppressor cells (MDSC) by cannabidiol in vivo. J Immunol. 2012;188:48.16.
58.
Zurück zum Zitat Taft RJ, Simons C, Nahkuri S, Oey H, Korbie DJ, Mercer TR, Holst J, Ritchie W, Wong JJL, Rasko JEJ, Rokhsar DS, Degnan BM, Mattick JS. Nuclear-localized tiny RNAs are associated with transcription initiation and splice sites in metazoans. Nat Struct Mol Biol. 2010;17:1030–4.CrossRefPubMed Taft RJ, Simons C, Nahkuri S, Oey H, Korbie DJ, Mercer TR, Holst J, Ritchie W, Wong JJL, Rasko JEJ, Rokhsar DS, Degnan BM, Mattick JS. Nuclear-localized tiny RNAs are associated with transcription initiation and splice sites in metazoans. Nat Struct Mol Biol. 2010;17:1030–4.CrossRefPubMed
59.
Zurück zum Zitat Liu M, Roth A, Yu M, Morris R, Bersani F, Rivera MN, Lu J, Shioda T, Vasudevan S, Ramaswamy S, Maheswaran S, Diederichs S, Haber DA. The IGF2 intronic miR-483 selectively enhances transcription from IGF2 fetal promoters and enhances tumorigenesis. Genes Dev. 2013;27:2543–8.CrossRefPubMedPubMedCentral Liu M, Roth A, Yu M, Morris R, Bersani F, Rivera MN, Lu J, Shioda T, Vasudevan S, Ramaswamy S, Maheswaran S, Diederichs S, Haber DA. The IGF2 intronic miR-483 selectively enhances transcription from IGF2 fetal promoters and enhances tumorigenesis. Genes Dev. 2013;27:2543–8.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Seviour EG, Sehgal V, Lu Y, Luo Z, Moss T, Zhang F, Hill SM, Liu W, Maiti SN, Cooper L, Azencot R, Lopez-Berestein G, Rodriguez-Aguayo C, Roopaimoole R, Pecot C, Sood AK, Mukherjee S, Gray JW, Mills GB, Ram PT. Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene. 2016;35:691–701.CrossRefPubMed Seviour EG, Sehgal V, Lu Y, Luo Z, Moss T, Zhang F, Hill SM, Liu W, Maiti SN, Cooper L, Azencot R, Lopez-Berestein G, Rodriguez-Aguayo C, Roopaimoole R, Pecot C, Sood AK, Mukherjee S, Gray JW, Mills GB, Ram PT. Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene. 2016;35:691–701.CrossRefPubMed
61.
Zurück zum Zitat Huang V, Place RF, Portnoy V, Wang J, Qi Z, Jia Z, Yu A, Shuman M, Yu J, Li L-C. Upregulation of cyclin B1 by miRNA and its implications in cancer. Nucleic Acids Res. 2012;40:1695–707.CrossRefPubMed Huang V, Place RF, Portnoy V, Wang J, Qi Z, Jia Z, Yu A, Shuman M, Yu J, Li L-C. Upregulation of cyclin B1 by miRNA and its implications in cancer. Nucleic Acids Res. 2012;40:1695–707.CrossRefPubMed
62.
Zurück zum Zitat Cui C, Yu J, Huang S, Zhu H, Huang Z. Transcriptional regulation of gene expression by microRNAs as endogenous decoys of transcription factors. Cell Physiol Biochem. 2014;33:1698–714.CrossRefPubMed Cui C, Yu J, Huang S, Zhu H, Huang Z. Transcriptional regulation of gene expression by microRNAs as endogenous decoys of transcription factors. Cell Physiol Biochem. 2014;33:1698–714.CrossRefPubMed
63.
Zurück zum Zitat Leucci E, Patella F, Waage J, Holmstrøm K, Lindow M, Porse B, Kauppinen S, Lund AH. microRNA-9 targets the long non-coding RNA MALAT1 for degradation in the nucleus. Sci Rep. 2013;3:2535.CrossRefPubMedPubMedCentral Leucci E, Patella F, Waage J, Holmstrøm K, Lindow M, Porse B, Kauppinen S, Lund AH. microRNA-9 targets the long non-coding RNA MALAT1 for degradation in the nucleus. Sci Rep. 2013;3:2535.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Fasanaro P, Greco S, Lorenzi M, Pescatori M, Brioschi M, Kulshreshtha R, Banfi C, Stubbs A, Calin GA, Ivan M, Capogrossi MC, Martelli F. An integrated approach for experimental target identification of hypoxia-induced miR-210. J Biol Chem. 2009;284:35134–43.CrossRefPubMedPubMedCentral Fasanaro P, Greco S, Lorenzi M, Pescatori M, Brioschi M, Kulshreshtha R, Banfi C, Stubbs A, Calin GA, Ivan M, Capogrossi MC, Martelli F. An integrated approach for experimental target identification of hypoxia-induced miR-210. J Biol Chem. 2009;284:35134–43.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Chiyomaru T, Yamamura S, Fukuhara S, Yoshino H, Kinoshita T, Majid S, Saini S, Chang I, Tanaka Y, Enokida H, Seki N, Nakagawa M, Dahiya R. Genistein inhibits prostate cancer cell growth by targeting miR-34a and oncogenic HOTAIR. PLoS ONE. 2013;8:e70372.CrossRefPubMedPubMedCentral Chiyomaru T, Yamamura S, Fukuhara S, Yoshino H, Kinoshita T, Majid S, Saini S, Chang I, Tanaka Y, Enokida H, Seki N, Nakagawa M, Dahiya R. Genistein inhibits prostate cancer cell growth by targeting miR-34a and oncogenic HOTAIR. PLoS ONE. 2013;8:e70372.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012;28:2062–3.CrossRefPubMedPubMedCentral Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012;28:2062–3.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Fabbri M, Garzon R, Andreeff M, Kantarjian HM, Garcia-Manero G, Calin GA. MicroRNAs and noncoding RNAs in hematological malignancies: molecular, clinical and therapeutic implications. Leukemia. 2008;22:1095–105.CrossRefPubMed Fabbri M, Garzon R, Andreeff M, Kantarjian HM, Garcia-Manero G, Calin GA. MicroRNAs and noncoding RNAs in hematological malignancies: molecular, clinical and therapeutic implications. Leukemia. 2008;22:1095–105.CrossRefPubMed
68.
Zurück zum Zitat Pulikkan JA, Dengler V, Peramangalam PS, Peer Zada AA, Müller-Tidow C, Bohlander SK, Tenen DG, Behre G. Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia. Blood. 2010;115:1768–78.CrossRefPubMedPubMedCentral Pulikkan JA, Dengler V, Peramangalam PS, Peer Zada AA, Müller-Tidow C, Bohlander SK, Tenen DG, Behre G. Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia. Blood. 2010;115:1768–78.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Rishi L, Hannon M, Salomè M, Hasemann M, Frank A-K, Campos J, Timoney J, O’Connor C, Cahill MR, Porse B, Keeshan K. Regulation of Trib2 by an E2F1-C/EBPα feedback loop in AML cell proliferation. Blood. 2014;123:2389–400.CrossRefPubMed Rishi L, Hannon M, Salomè M, Hasemann M, Frank A-K, Campos J, Timoney J, O’Connor C, Cahill MR, Porse B, Keeshan K. Regulation of Trib2 by an E2F1-C/EBPα feedback loop in AML cell proliferation. Blood. 2014;123:2389–400.CrossRefPubMed
70.
71.
73.
Zurück zum Zitat Ono M, Yamada K, Avolio F, Scott MS, van Koningsbruggen S, Barton GJ, Lamond AI. Analysis of human small nucleolar RNAs (snoRNA) and the development of snoRNA modulator of gene expression vectors. Mol Biol Cell. 2010;21:1569–84.CrossRefPubMedPubMedCentral Ono M, Yamada K, Avolio F, Scott MS, van Koningsbruggen S, Barton GJ, Lamond AI. Analysis of human small nucleolar RNAs (snoRNA) and the development of snoRNA modulator of gene expression vectors. Mol Biol Cell. 2010;21:1569–84.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Ono M, Yamada K, Endo A, Avolio F, Lamond AI. Analysis of human protein replacement stable cell lines established using snoMEN-PR vector. PLoS ONE. 2013;8:e62305.CrossRefPubMedPubMedCentral Ono M, Yamada K, Endo A, Avolio F, Lamond AI. Analysis of human protein replacement stable cell lines established using snoMEN-PR vector. PLoS ONE. 2013;8:e62305.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Ono M, Yamada K, Avolio F, Afzal V, Bensaddek D, Lamond AI. Targeted knock-down of miR21 primary transcripts using snoMEN vectors induces apoptosis in human cancer cell lines. PLoS ONE. 2015;10:e0138668.CrossRefPubMedPubMedCentral Ono M, Yamada K, Avolio F, Afzal V, Bensaddek D, Lamond AI. Targeted knock-down of miR21 primary transcripts using snoMEN vectors induces apoptosis in human cancer cell lines. PLoS ONE. 2015;10:e0138668.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Wang C, Chen Z, Ge Q, Hu J, Li F, Hu J, Xu H, Ye Z, Li L-C. Up-regulation of p21WAF1/CIP1 by miRNAs and its implications in bladder cancer cells. FEBS Lett. 2014;588:4654–64.CrossRefPubMed Wang C, Chen Z, Ge Q, Hu J, Li F, Hu J, Xu H, Ye Z, Li L-C. Up-regulation of p21WAF1/CIP1 by miRNAs and its implications in bladder cancer cells. FEBS Lett. 2014;588:4654–64.CrossRefPubMed
Metadaten
Titel
Nuclear microRNAs in normal hemopoiesis and cancer
verfasst von
John E.J. Rasko
Justin J.-L. Wong
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2017
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-016-0375-x

Weitere Artikel der Ausgabe 1/2017

Journal of Hematology & Oncology 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.