Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Review

Principles and innovative technologies for decrypting noncoding RNAs: from discovery and functional prediction to clinical application

verfasst von: Yu-Meng Sun, Yue-Qin Chen

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

Noncoding RNAs (ncRNAs) are a large segment of the transcriptome that do not have apparent protein-coding roles, but they have been verified to play important roles in diverse biological processes, including disease pathogenesis. With the development of innovative technologies, an increasing number of novel ncRNAs have been uncovered; information about their prominent tissue-specific expression patterns, various interaction networks, and subcellular locations will undoubtedly enhance our understanding of their potential functions. Here, we summarized the principles and innovative methods for identifications of novel ncRNAs that have potential functional roles in cancer biology. Moreover, this review also provides alternative ncRNA databases based on high-throughput sequencing or experimental validation, and it briefly describes the current strategy for the clinical translation of cancer-associated ncRNAs to be used in diagnosis.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ncRNA
Noncoding RNA
lncRNA
Long noncoding RNA
circRNA
Circular RNA
mRNA
Message RNA
H3K4me3
Trimethylation of lysine 4 of histone 3
H3K36me3
Trimethylation of lysine 36 of histone 3
lincRNA
Long intergenic ncRNA
H3K4m1
Monomethylation of lysine 4 of histone 3
H3K27ac
Histone H3 acetylation at lysine 27
eRNA
Enhancer-derived RNA
3D
3 Dimensions
pre-RNA
Precursor RNA
snoRNA
Small nucleolar RNA
rRNA
Ribosomal RNA
tRNA
Transfer RNA
sno-lncRNA
SnoRNA-related lncRNA
rRF
Ribosomal RNA-derived fragment
tsRNA
tRNA-derived small RNA
sdRNA
snoRNA-derived RNA
ORF
Open reading frame
SRA
Steroid receptor RNA activator
Xist
X inactive specific transcript
lnx3
Ligand of numb-protein X 3
Pol II
RNA polymerase II
snoRBP
snoRNA binding protein
RNA-seq
RNA sequencing
XPO1
Exportin 1
CAMSAP1
Calmodulin-regulated spectrin-associated protein 1
EIF3J
Eukaryotic translation initiation factor 3 subunit J
PAIP2
Poly(A)-binding protein-interacting protein 2
miRNA
MicroRNA
siRNA
Small interfering RNA
piRNA
Piwi-interacting RNA
Ago
Argonaute
RISCs
RNA-induced silencing complexes
RBP
RNA-binding protein
vRNA
Vault RNA
VTRNA
Vault RNA gene
Pol III
RNA polymerase III
Dicer
Ribonuclease III
cDNA
Complementary DNA
HOTAIR
HOX antisense intergenic RNA
NGS
Next-generation sequencing
PRC2
Polycomb repressive complex
scRNA-seq
Single-cell RNA sequencing
FACS
Fluorescence-activated cell sorting
DP-seq
Designed primer-based sequencing
MMLV RT
Moloney murine leukemia virus reverse transcriptase
PCR
Polymerase chain reaction
SUPeR-seq
Single-cell universal poly(A)-independent sequencing
RamDA-seq
Random displacement amplification sequencing
RT-RamDA
RT with random displacement amplification
NSR
Not-so-random primer
Neat1
Nuclear paraspeckle assembly transcript 1
GRO-seq
Global run-on sequencing
RT
Reverse transcription
GRID-seq
Global RNA interactions with DNA by deep sequencing
DSG
Disuccinimidyl glutarate
ssRNA
Single-stranded RNA
dsDNA
A double-stranded DNA
ChAR-seq
Chromatin-associated RNA sequencing
caRNA
Chromatin-associated RNA
TSSs
Transcription star sites
snRNA
Small nuclear RNA
CLASH
Cross-linking ligation and sequencing of hybrids
RIPPLiT
RNA immunoprecipitation and proximity ligation in tandem
MARIO
Mapping the RNA interactome in vivo
PARIS
Psoralen analysis of RNA interactions and structures
LIGR-seq
Ligation of interacting RNA followed by high-throughput sequencing
SPLASH
Sequencing of psoralen cross-linked, ligated, and selected hybrids
RIC
RNA in situ conformation sequencing
ISH
In situ hybridization
smFISH
Single-molecule fluorescence ISH
FISSEQ
Fluorescent in situ RNA sequencing
STARmap
Spatially resolved transcript amplicon readout mapping
CeFra-seq
Biochemical cell fractionation combined with RNA-seq
APEX
Engineered ascorbate peroxidase
RIP
RNA immunoprecipitation
CLIP-seq
Immunoprecipitation followed by deep sequencing
ChIRP-seq
Chromatin isolation by RNA purification followed by high-throughput sequencing

Background

More than half a century after being considered as the central component in the central dogma of biology, RNA has been accepted to play various essential roles in different biological processes [14]. With recent developments in sequencing methods and information analysis, an increasing number of novel ncRNAs have been identified, including long noncoding RNAs (lncRNAs) [5, 6], circular RNAs (circRNAs) [7, 8], and novel small ncRNAs [911]. Growing studies have uncovered the characteristics of these ncRNAs, including their origins, mechanisms of generation, structures, and potential functions [6, 8, 12], which can be summarized into a principle for the identification of known species of ncRNAs or even novel ncRNA discovery. As many ncRNAs exhibit highly tissue-specific expression patterns and important roles in biological processes related to cancer [1319], ncRNAs have been considered as ideal therapeutic targets for cancer diagnosis and treatment [2022]. Due to the enormous transcription potential of mammalian genomes and multiple mechanisms of ncRNA generation [8, 9, 23, 24], the ncRNA world is still full of infinite mysteries, in which unknown species of RNAs could play important roles. Technological innovation makes it possible to discover more novel functional ncRNAs.
This review focuses on the principles and innovative technologies currently available for the discovery of novel ncRNAs or functional ncRNAs within specific subcellular compartments. The particular classes of ncRNAs that are either novel transcripts or “old dogs” performing “new tricks” are especially emphasized. Moreover, this review also provides an overview of ncRNA-associated databases and applications of cancer-related ncRNA identification for therapeutic strategies.

Principle for novel ncRNA discovery

Early sequencing data revealed that the mammalian genome encodes many thousands of noncoding transcripts, especially those that resemble message RNAs (mRNAs) in length and splicing structure but cannot code for proteins, revealing that the world of RNA genes is far more complex than originally imagined [25]. Here, we summarized the features into a principle that could be used for the identification of known species of ncRNAs or even for novel ncRNA discovery.

Chromatin signatures for novel ncRNA discovery

The definition of genes has become a major hurdle following the sequencing of the human genome. As histones can be modified in different ways that are indicative of the underlying DNA functional state [2629], chromatin modifications of the corresponding genomic region could represent important biological information for the identification and classification of noncoding transcripts. The increased occurrence of trimethylation of lysine 4 of histone 3 (H3K4me3) at the promoter regions of transcripts and trimethylation of lysine 36 of histone 3 (H3K36me3) along the entire transcribed region is a signature for active transcription; these occurrences are always found at active sites of mRNA transcription [27, 28]. By searching for H3K4me3/H3K36me3 signatures that failed to overlap with known genes, there was the identification of approximately 2500 regions in the human genome and approximately 1600 regions in the mouse genome that were actively transcribed [30, 31]. However, the vast majority of these intergenic regions with H3K4me3/H3K36me3 signatures produced multi-exonic RNAs that had a little capability to encode a conserved protein; they were termed as long intergenic ncRNAs (lincRNAs) (Fig. 1a) [30, 32]. A fraction of genes encoding ncRNAs display monomethylation of lysine 4 of histone 3 (H3K4m1) and histone H3 acetylation at lysine 27 (H3K27ac), which cover their initiation sites, indicating that they are transcribed from activated enhancers as enhancer-derived RNAs (eRNAs) (Fig. 1a) [29, 33]. Although both lincRNAs and eRNAs are categorized as lncRNAs because of their lengths, distinguishing different classes of ncRNAs based on distinct chromatin modifications is necessary because specific ncRNAs generated from given gene regulatory elements could function in classic modes [34, 35]. For example, eRNAs are thought to play an important role in regulating the 3D architecture of chromosomes near their site of transcription [34].
With developments in sequencing technologies and bioinformatics analysis, novel ncRNAs generated from alternative splicing processing or degradation of their parent RNAs have been discovered [8, 9, 36]. This kind of ncRNA does not have independent genomic regions or transcriptional regulatory elements and can be produced following parent gene transcription or degradation. Therefore, it is unable to accurately identify and describe the characteristics of these kinds of ncRNAs at the level of chromatin modification. As a typical example, circRNAs are mainly generated from alternative splicing of precursor RNA (pre-RNA), and then, they form covalently closed loop structures [8, 37]. Exonic circRNAs are produced from back-spliced exons of precursor linear RNAs, including mRNAs and lncRNAs, and they account for a major portion of the circRNA family. In addition, the intron lariats escaping from degradation can also form intronic circRNAs. Although there are some other variant forms of circRNAs, such as circular formats of small nucleolar RNAs (snoRNAs) and P RNA [38], the majority of circRNAs in humans are mainly produced from actively transcribed mRNA and lncRNA genes with H3K4me3-H3K36me3 signatures [39, 40]. Interestingly, the junction site sequences of circRNAs, such as circSTATB1 in mice, have been discovered to be inserted into an enhancer with active H3K4me1 signatures (Fig. 1a) [41]. The H3K4me1 modifications suggest that the functions of circRNAs in the regulation of enhancer and genome structure by forming pseudogenes, which may provide evidence for further classification of circSTATB1 as a retrotransposed circRNA (Fig. 1a) [41]. Although chromatin modifications cannot be used in the discovery of circRNAs, the modification signatures may be useful for more detailed classification of circRNAs.
In addition to circRNAs, there are many other novel ncRNAs that are generated from the degradation of typical transcripts from well-known genomic regions [9, 11, 42]. The excised intron-derived lncRNAs with snoRNA-like ends (sno-lncRNAs) are formed when one intron contains two snoRNA genes [42]. After splicing, the sequences between two snoRNAs escape degradation, resulting in the accumulation of certain lncRNAs. Another example is novel functional small ncRNAs, such as small ribosomal RNA-derived fragments (rRFs) [11], tRNA-derived small RNAs (tsRNAs) [9], and snoRNA-derived RNAs (sdRNAs) [10], which are derived from “old dogs” including ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), and snoRNAs. An increasing number of discoveries of novel ncRNAs have indicated the limitation of chromatin modification signatures in novel ncRNA identification. However, chromatin signatures are still an available tool of ncRNA classification for efficient investigation of their functions.

Principles for evaluating coding potential

As ncRNAs, especially lncRNAs and circRNAs, are likely to contain open reading frames (ORFs) purely by chance, it has been a challenge to determine whether a transcript is noncoding [43]. As a growing number of studies have shown that several lncRNAs and circRNAs can produce functional micropeptides [4447], it is necessary to evaluate the RNA coding potential of novel ncRNAs.
The lack of evolutionary conservation in identified ORFs is evidence for the absence of coding potential of ncRNAs [48, 49]. Novikova et al. reported that a human lncRNA, SRA, has different isoforms that either function at the ncRNA level or produce proteins, and there is higher evolutionary stabilization of the RNA structural core than that of the translational product under evolutionary pressure [50]. Another example is Xist, a lncRNA involved in X chromosome inactivation in mammals that originates from the protein-coding gene Lnx3 [51]. Interestingly, the Lnx3 gene is still a protein-coding gene in opossum; however, it has been transformed into a noncoding transcript with frame-shifting mutations in later vertebrates [51]. In addition, the lack of homology to known protein domains and the inability to template significant protein production are the other important factors that are needed to be considered [48, 49]. These principles have been generalized to classify ncRNA coding potential by scoring conserved ORFs across diverse species with computational methods [52, 53], by searching for homology using protein-domain databases [54], and by sequencing ncRNAs associated with polyribosomes [55].
Table 1
Characteristics of diverse sequencing methods
Classification
Techniques
Short description
Strengths of the approach
Weakness
Ref
Microarrays
Tiling arrays
A method based on probes for discovering transcripts from specific genomic regions.
This approach can provide in-depth analysis of transcripts from target regions of genome.
Suffer from potential noise as a result of weak binding or cross-hybridization of transcripts to probes.
[56]
Microarrays
A method based on a large number of oligonucleotide probes for performing quick global or parallel expression analysis of transcriptome.
Small size and high-throughput capabilities.
This method is not able to discover novel transcripts.
[57]
RNA-seq
RNA-seq
A technique that is currently the most widespread sequencing technology for both detecting RNA expression and discovering novel RNAs.
The method provides a global high-throughput detection amd identification of RNAs greater than 200 nt.
Its standard procedure is not suitable for detection of RNAs less than 200 nt. It also suffer from sequence errors at the reverse-transcription step or primer bias.
[58]
RNA capture sequencing
A derivative technology combining RNA-seq with tilling arrays.
The method can specifically elevate the sequencing depth of target regions.
Suffer from disadvantages of both tiling arrays and RNA-seq.
[59]
scRNA-seq
Smart-seq
A scRNA-seq method based on a full-length cDNA amplification strategy.
Provide a full-length cDNA amplification of polyadenylated RNAs.
The limitations are lack of strand-specific identification, inability to read transcripts longer than 4 kb and only for polyadenylated RNAs.
[60]
DP-seq
A scRNA-seq method using heptamer primers.
Suitable for smaller size samples or transcripts longer than 4 kb. this approach also suppresses highly expressed rRNAs in the cDNA library.
Captured RNAs are limited to polyadenylated RNAs.
[61]
Quartz-seq
A scRNA-seq method which reduces back ground noise.
Reduce background noise by using specially suppression PCR primers to reduce side products.
The method is limited to detecting polyadenylated RNAs.
[62]
SUPeR-seq
A single-cell universal polyadenylated tail-independent RNA sequencing.
Detect polyadenylated and nonpolyadenylated RNAs. Minimal rRNAs contamination.
Relatively low sensitivity for nonpolyadenylated RNAs.
[63]
RamDA-seq
A full-length total RNA-sequencing method for analyzing single cells.
High sensitivity for nonpolyadenylated RNAs. It can also uncover the dynamics of recursive splicing.
Unknown
[64]
Small RNA-seq
Small RNA-seq
A type of RNA-seq that discriminate small RNA from larger RNA to better evaluate and discover novel small RNAs.
Specifically detect and discover small or intermediate-sized RNAs with target sizes.
Adapter ligation bias lead to reverse transcription bias or amplification bias.
[65]
Single-cell small-RNA sequencing
Small-seq
A method which detect small RNAs in a single cell.
The method can detect small RNAs in a single cell.
The limination may be similar to small RNA-seq.
[66]
Nascent RNA-seq
GRO-seq
A method labeling nascent RNAs with 5Br-UTP and immunoprecipitating RNAs for sequencing.
Detect nascent RNAs and provide a genome-wide view of the location, orientation, and density of Pol II-engaged transcripts.
The method is confounded by contamination due to nonspecific binding, which could possibly result in experimental bias.
[67]
SLAM-seq
A method distinguishing nascent RNA from total RNA via s4U-to-C conversion induced by nucleophilic substitution chemistry.
It is an enrichment-free method which can avoid contamination induced by affinity purification.
The oxidation condition caused certain oxidative damage to guanine, which may impact the accurancy of sequencing.
[68]
TimeLapse-seq
A method distinguishing nascent RNA from total RNA via s4U-to-C conversion induced by an oxidative nucleophilic aromatic substitution reaction.
It is an enrichment-free method which can avoid contamination induced by affinity purification.
The oxidation condition caused certain oxidative damage to guanine, which may impact the accurancy of sequencing.
[69]
AMUC-seq
A method distinguishing nascent RNA from total RNA via transforming s4U into a cytidine derivative using acrylonitrile.
More efficient and reliable because it has a minimal influence on the base-pairing manner of other nucleosides.
Unknown
[70]
Identification of RNA-chromatin interaction
GRID-seq
A method that aims to comprehensively detect and determine the localization of all potential chromatin-interacting RNAs.
Use a bivalent linker to ligate RNA to DNA in situ and provide exact profiles of RNA-chromatin interactome.
Usable sequence length for mapping RNA is 18–23 bp. However, short sequence length can result in ambiguity in mapping.
[71]
iMARGI
A method providing a in situ mapping of RNA-genome interactome.
iMARGI needs less number of input cells and is suitable for paired-end sequencing.
Unknown
[72]
ChAR-seq
A chromatin-associated RNA sequencing that maps genome-wide RNA-to-DNA contacts.
Uncover chromosome-specific dosage compensation ncRNAs, and genome-wide trans-associated RNAs.
The method needs more than 100 million input cells.
[73]
Identification of RNA-RNA interaction
CLASH
A relatively early method that uses UV cross-linking to capture direct RNA-RNA hybridization.
Avoid noise from protein intermediate-mediated interactions.
This method only detects the RNA-RNA interactions base on proteins.
[74]
RIPPLiT
A transcriptome-wide method for probing the 3D conformations of RNAs stably associated with defined proteins.
The method can capture 3D RNP structural information independent of base pairing.
This method only detects the RNA-RNA interactions base on proteins.
[75]
MARIO
A method identifying RNA-RNA interactions in the vicinity of all RNA-binding proteins using a biotin-linked reagent.
This method can identify RNA-RNA interactions in the vicinity of all RNA-binding proteins.
The method only detects the RNA-RNA interactions base on proteins.
[76]
PARIS
Psoralen analysis of RNA interactions and structures with high throughput and resolution.
Directly measure RNA-RNA interactions independent of proteins in living cells.
Unknown
[77]
LIGR-seq
A method for the global-scale mapping RNA-RNA interactions in vivo.
Provide global-scale mapping RNA-RNA interactions independent of proteins in vivo
Unknown
[78]
SPLASH
A method providing pairwise RNA-RNA partnering information genome-wide.
Map pairwise RNA interactions in vivo with high sensitivity and specificity, genome-wide.
Unknown
[79]
RIC-seq
RNA in situ conformation sequencing technology for the global mapping of intra- and intermolecular RNA-RNA interactions.
The method performs RNA proximity ligation in situ and can facilitate the generation of 3D RNA interaction maps.
Unknown
[80]
RNA proximity sequencing
A method based on massive-throughput RNA barcoding of particles in water-in-oil emulsion droplets.
This method can detect multiple RNAs in proximity to each other without ligation and is fit for studying the spatial organization of RNAs in the nucleus.
Unknown
[81]
RNAs in protein complexes or subcellular structures
FISSEQ
A method that offers in situ information of RNAs at high-throughput levels.
Provide information of RNAs at high-throughput levels. Visualization.
Unknown
[82]
CeFra-seq
A method that physically isolates subcellular compartments and identifies their RNAs.
The methods have high sensitivity for low-abundance transcripts.
The method is limited to isolation protocols and the purity of resulting isolates.
[83]
APEX-RIP
A method can map organelle-associated RNAs in living cells via proximity biotinylation combined with protein-RNA crosslinking.
The technique can offer high specificity and sensitivity in targeting the transcriptome of membrane-bound organelles.
Unknown
[84]
However, the coding potential of some novel ncRNAs, especially circRNAs, could fail to be determined with the principle mentioned above. Most circRNAs derived from mRNA back-splicing lose translational capacity because of the lack of effective ORFs or ribosome entry approaches, while a few circRNAs from coding or noncoding transcripts could also obtain novel ORFs and may be translated into new proteins [47, 85]. The deficiency of coding-potential evaluation could be due to the incomplete circRNAs databases across diverse species, the complex mechanism of ribosome entry and translational initiation of circRNAs [86], and the lack of databases that document the information of new peptides or proteins transcribed from novel templates containing the sequences of circRNA junction sites. Ribosome profiling has provided a strategy to identify ribosome occupancy on RNA, which has been proposed to be an available method for distinguishing noncoding transcripts from coding ones [55]. Nevertheless, some transcripts playing clear roles as ncRNAs have been detected in ribosomes, indicating that an association of RNA with a ribosome alone cannot be taken as evidence of protein-coding potential [87, 88]. These ribosome-associated ncRNAs may serve as translational regulators or may produce nonfunctional translation noise [89, 90]. Thus, experimental technologies such as mass spectrometry proteomics have been used to improve the accuracy of noncoding transcript definition [91].

Characteristics of known ncRNAs

With the development of sequencing methods and information analysis, a vast number of diverse types of ncRNAs have been identified, such as microRNAs (miRNAs), lncRNAs, circRNAs, and novel small ncRNAs derived from well-known RNAs. Understanding the characteristics of the known ncRNAs would be helpful for novel ncRNA discovery.
NcRNAs are very heterogeneous in terms of their length and conformation [92]. They can be separated into 3 categories: (1) small ncRNAs (< 50 nt), including miRNAs (19–25 nt) [93], small interfering RNAs (siRNAs, 19–29 nt) [94], piwi-interacting RNAs (piRNAs, 25–31 nt) [95], and other functional small RNAs such as transcription initiation RNAs (tiRNAs, 17–18 nt) [96], tsRNAs (14–36 nt) [9], sdRNAs (17–24 nt or > 27 nt) [10], and sectional rRFs (15-81 nt) [11]; (2) intermediate-sized ncRNAs (50–500 nt), including 5S rRNAs (~120 nt) [97], 5.8S rRNA (~150 nt) [98], tRNAs (76–90 nt) [99], snoRNAs (60–300 nt) [100], and small nuclear RNAs (snRNAs, ~150 nt) [101]; (3) long noncoding transcripts greater than 500 nt, including linear lncRNAs [30] and circular circRNAs [40].
Most large ncRNAs, including lncRNAs and circRNAs, have been reported to be tissue-specific and expressed at relatively low levels [24, 102104]. Different types of ncRNAs have distinct structures that maintain their stability. The most abundant lncRNAs are transcribed by RNA polymerase II (Pol II), and then, they undergo mRNA-like posttranscriptional processes, leading to 5′-caps and polyadenylated tails at their 3′ ends [30]. However, studies of novel ncRNA identification that were not based on polyadenylated tails have shown the existence of nonpolyadenylated ncRNAs such as sno-lncRNAs with snoRNA-like ends and circRNAs (Fig. 1b, c) [42]. Several sno-lncRNAs have been reported to stabilize their structures by interacting with classical snoRNA binding proteins (snoRBPs) via the classical stem-loop structures of snoRNAs (Fig. 1b) [105]. In addition, circRNAs are processed to form covalently closed loop structures without open terminals, which makes them resistant to degradation by exonucleases, causing them to have relatively high stability (Fig. 1c) [8]. In contrast, most eRNAs are nonpolyadenylated transcripts that have shorter half-lives than polyadenylated lncRNAs and are difficult to discover according to their even lower levels in organisms [24, 106].
Intermediate-sized and small ncRNAs possess specifically structural features as well, such as the conversed stem-box structures of snoRNAs (C/D box or H/ACA box) [100], unique 5′-caps of snRNAs (5′-trimethylguanosine caps or 5′-monomethylphosphate caps) [101, 107], the cloverleaf-like secondary structure of tRNA [99], and hairpin loop of miRNA precursor. Most types of intermediate-sized and small ncRNAs do not have specific modification at the 5′ or 3′ ends, and they maintain their stabilities via binding specific proteins to form complexes. For example, snoRNAs stabilize their structures by interacting with classical snoRBPs via the classical stem-loop structures [108]. Another example is miRNA, whose precursor yileds a miRNA:miRNA duplex with Dicer processing [109]. In most cases, only one strand of the deplex is usually incorporated into the RNA-induced silencing complex (RISC) to exist and function, and the other free strand is normally degraded. Together, RNA structures could affect their expression levels in cells, which always influences the discovery of potential novel ncRNAs.

Principle and strategy for identification of novel ncRNAs

Nowadays, increased types of ncRNAs have been detected and identified by the development of next-generation sequencing (NGS) [58], which can be roughly divided into the process sections of sample preprocessing, library preparation, sequencing, and bioinformatics. Importantly, it shoud be noted that the ways of RNA isolation and library preparation greatly affect the detection of target species of ncRNAs.
Organic reagent method using isothiocyanate/phenol/chloroform or Trizol (Invitrogen) is an universial RNA extraction way to obtain total RNA containing small and intermediate-sized RNA. However, it has been reported that phenol contamination has influences on RNA yields and subsequent sequencing [110]. Spin column chromatography using commerial kits without phenol can avoid this contamination and obtain relatively high-quality RNA from the same samples. However, silica-based spin column chromatography fails to efficiently capture RNA shorter than 200 nt, which leads to massive loss of small and intermediate-sized ncRNAs and makes the way unsuitable for small RNA-seq [111, 112]. In contrast, the ways using spin column that can capture all RNA greater than 10 nt can be selected when we aim to obtain total ncRNAs or specifically enrich small ncRNAs. Choosing appropriate ways of RNA extraction is important for identification of novel ncRNAs with a specific size.
Library with appropriate RNA selection/depletion is also pivotal in the detection of specific types of ncRNAs. In library preparation for mRNA sequencing, RNAs with polyadenylated tails are specifically isolated by hybridization with poly(dT) oligomers from nonpolyadenylated RNAs which include a vast number of rRNAs. As a part of lncRNAs do not have polyadenylated tails, polyadenylated tail selection can only capture mRNA-like lncRNAs [113]. As for total lncRNA sequencing, library preparation is generally dependent on rRNA depletion methods. Next, the filtered RNAs are fragmented, reverse transcribed into cDNA by random primers, and undergo end repair, sequencing adaptor ligation, and size selection for subsequent sequencing. In this way, not only lncRNA but also mRNA, circRNA, and a part of intermediate-sized ncRNAs can be detected. However, reverse transcription (RT) by random primering and size selection leads to the deficiency of small ncRNAs such as miRNAs [114]. Depletion of linear RNAs by Rnase R treatment for circRNA sequencing and separation of RNAs with specific size by gel electrophoresis can specifically enrich target types of ncRNAs for RNA-seq, which are as far as possible to reduce interference signal from other transcripts. In addition, due to the shortened size, small RNA is hard to be successfully acquired through cDNA synthesis (first or second cDNA synthesis) by random priming and be always removed by size selection after sequencing adaptor ligation [114]. Thus, in small RNA-seq, both ends of the RNA fragments are firstly ligated to the adapters and followed by the cDNA synthesis and library construction. We also need to pay attention to the effects of RNA modifications on library preparation, which usually influence adapter ligation. For example, 5′ caps of snRNAs shoud be removed before adapter ligation. Selecting appropriate methods of library preparation is also important for identification of novel ncRNAs [101, 107].
It is worth noting that alternative splicing processes enable great complexity in transcripts from the same genomic regions [115]. For linear ncRNAs, various isoforms can be relatively easy to identify by RNA-seq. Nevertheless, despite the identification of circRNAs based on the junction site, extra sequence identification is still needed to determine the actual sequences of circRNAs because of potential circRNA variants being generated from a single gene locus [116]. This issue results from alternative splicing that occurs within circRNAs with multiple exons (Fig. 1c) [116]. All four basic types of canonical alternative splicing were found to occur in circRNAs as well: cassette exon, intron retention, alternative 5′ splicing and alternative 3′ splicing (Fig. 1c) [116]. For example, the human XPO1 gene locus has been demonstrated to contain a circRNA-predominant cassette exon, the CAMSAP1 gene locus generates two cirRNA isoforms with or without a retained intron, and the human EIF3J and PAIP2 gene loci can also produce circRNAs containing both exon and intron sequences [104, 117, 118]. Other factors, such as read-through transcription and the fusion of genes derived from chromatin rearrangement, also generate read-through circRNAs and fusion circRNAs, respectively, which increase the diversity of ncRNAs [119, 120].
Traditionally well-known small noncoding RNAs, including miRNAs, siRNAs, and piRNAs, function in concert with the Argonaute (Ago) family of proteins to regulate gene expression at the level of transcription, mRNA stability, or translation [121, 122]. Interestingly, sdRNAs were initially discovered from an analysis of small RNAs associated with human Ago1 and Ago2 revealed by immunoprecipitation and RNA-seq (Fig. 1d) [10]. In addition, a number of novel small ncRNAs derived from both rRNAs (rRFs) and tRNAs (tsRNAs) have also been found to be enriched in RNA-induced silencing complexes (RISCs), and they function in a miRNA-like pathway (Fig. 1d) [9, 11, 36]. Immunoprecipitation of members of the Ago family proteins followed by small RNA-seq has revealed a series of novel small ncRNAs that play roles in RNA-induced target gene silencing. These data suggested that functional ncRNAs in well-known complexes should have more extensive sources and that transcripts derived from canonical DNA regions could have functions in addition to their classical ones by interacting with nonclassical RNA binding proteins (RBPs) or being located in novel complexes. This method of identifying RNA found in specific complexes or associating with subcellular components followed by RNA-seq represents an ideal way to discover new species of functional small ncRNAs. For example, the Vault complex, a novel ribonucleoprotein that probably functions in the nuclear export of large molecules, was isolated and characterized in 1986 [123]. By analyzing the components of Vaults, researchers discovered a novel and single species of small ncRNAs that is 86-141 nt in length, which was termed Vault RNAs (vRNAs) [124]. VRNAs that are derived from VTRNA genes by RNA polymerase III (Pol III) have been reported to be associated with multidrug resistance and, interestingly, also be the origin of miRNA-like small ncRNAs processed by Dicer [125]. Another example of identification or RNAs in complexes is snoRNAs, whose canonical functions are generally considered to guide the pseudouridylation and 2′-O-methylation of rRNA in the nucleolus [126]. However, in situ global RNA interactions with DNA identified by immunoprecipitation and RNA-seq showed that snoRNAs represent a vast population and a high enrichment in the chromatin-bound fractions, suggesting the other potential functions of these well-known small ncRNAs located in the nonclassical complexes [71, 127, 128].
Lack of sequence conservation, low level or high tissue-specific expression pattern, or derivation from canonical DNA sequences are potential factors that make the discovery and identification of novel ncRNAs difficult. We provided the identification principle of recently discovered functional ncRNAs, which would be a referential principle for novel ncRNA discovery. Importantly, recent technological developments, especially specific sequencing technological developments, have provided multiple approaches for the discovery and study novel ncRNAs.

Approaches for discovering ncRNAs

Most ncRNAs, such as lncRNAs and circRNAs, have the characteristics of spatiotemporal specificity and low expression levels, which make it difficult to identify them [24, 102104]. Therefore, it is necessary for us to purposefully choose the appropriate methods in sample preparation and sequencing techniques. Here, we will review innovative and novel sequencing methods that significantly improve the process of RNA identification and investigation, placing special emphasis on their advantages and limitations (Table 1).

Tiling arrays and microarrays

Tiling array is an alternative and classic method for discovering RNA [56]. This approach hybridizes complementary DNAs (cDNAs) to microarray slides containing tiled oligonucleotide probes that are designed to hybridize with nonrepetitive sequences of specific genomic regions or the entire genome [56]. For example, tiling arrays were used to specifically identify the potential transcripts from four human HOX gene clusters with 400,000 probes, leading to the discovery of intergenic ncRNAs, including the well-known lncRNA HOX antisense intergenic RNA (HOTAIR) [129]. Tiling arrays can also provide in-depth analysis of alternative splicing, polymorphism, and novel transcription site identification by elevating the resolutions of designed probes [56, 130]. Nevertheless, because microarrays suffer from potential noise as a result of weak binding or cross-hybridization of transcripts to probes, tiling arrays have been replaced by NGS technologies and now preferably serve as a supplemental step for RNA-seq to increase the sequencing depth of target regions.
Microarray is an important method for performing quick global or parallel expression analysis of the transcriptome in different cell/tissue types, experimental systems, developmental stages, or pathological conditions [57]. This classic method consists of a large number of oligonucleotide probes spotted on a solid surface that are then allowed to hybridize to target sequences from samples, which are further detected by fluorescently labeled target sequences. The intensity of fluorescence is used to quantify target sequences. Their small size and high-throughput capabilities have brought microarrays to the forefront of RNomic research. However, this approach can only detect RNAs whose sequences are known and have specific hybridization probes; this method is not able to discover novel transcripts.

RNA-seq

RNA-seq is currently the most widespread sequencing technology for both detecting RNA expression and discovering novel species of ncRNAs (Fig. 2a) [24, 58]. In addition, this approach can also be used to identify single nucleotide polymorphisms, alternative splicing isoforms, gene fusion events, and novel splice junctions [131134]. RNA-seq is based on the conversion of RNA into a pool of cDNA with either oligo (dT) primers or random primers, depending on the purpose of the sequencing. However, because cDNA libraries prepared with oligo (dT) selectively enrich for polyadenylated RNA and simultaneously deplete nonpolyadenylated and partially degraded transcripts, RNA-seq with random primers for cDNA synthesis on rRNA-depleted transcripts is currently a more widely used approach. Analysis of human or mouse cell types using RNA-seq revealed the presence of more than 8000 human and over 1000 mouse long intergenic ncRNAs (lincRNAs), the majority of which had not been previously identified [32, 135]. Interestingly, in one study using RNA-seq for the specific identification of nonpolyadenylated RNA, a novel species of lncRNAs with snoRNA-like ends was discovered to be produced from excised introns [42, 105]. Moreover, the first identification of large numbers of circRNAs in humans and mice occurred following the combination of RNA-seq and RNase R treatment, which uncovered the effective presence of 1950 human and 1903 mouse circRNAs in human cell lines (HEK293 and leukocytes) and mouse tissues such as the brains and fetal heads [7]. In addition, RNA-seq with specific preparation for small RNA identification is also the primary approach for discovering and detecting miRNAs, snoRNAs, piRNAs, and other novel small ncRNAs, including IRFs, tsRNA, and sdRNAs [10, 126, 136139].
There is a derivative technology based on RNA-seq, RNA capture sequencing, which is combined with tiling arrays to elevate the sequencing depth of target regions [59]. In brief, tiling arrays are performed first with specific oligonucleotide probes to enrich cDNAs from specific genomic regions. Second, the hybridized cDNAs are eluted and then sequenced by RNA-seq. RNA capture sequencing increases the sequencing depth in specific genomic regions compared to RNA-seq and has uncovered multiple unannotated isoforms of both mRNAs and ncRNAs, including a novel alternative splicing transcript of HOTAIR that lacks the binding domain for the polycomb repressive complex (PRC2) [59].
Over the years, many technologies based on basic RNA-seq have been developed to identify RNAs at the transcriptome scale, some of which will be discussed in the following sections. It is inferred that advanced algorithms for analysis of sequencing data are also likely to promote transcriptome analysis. Nevertheless, RNA-seq may suffer from disadvantages such as the introduction of sequence errors at the reverse-transcription step or primer bias, which require further optimization [140].

Small RNA-seq and single-cell small-RNA sequencing

Because sample preparation for RNA-seq is not suitable for small RNAs, such as reverse transcription with random priming (short RNA species yield even shorter cDNAs that are not long enough for efficient alignment), small RNA-seq with modified library preparation, such as miRNA-seq, was developed [65, 114, 141]. Small RNA-seq is a type of RNA-seq that discriminate small RNAs from larger RNAs to better evaluate and discover novel small RNAs [65]. In this method, small RNAs are fractionated by gel electrophoresis, and then, universal adapters are ligated to the both ends of RNA fragments, which are acted as primer binding sites during reverse transcription and PCR amplification. Previous studies using small RNA-seq detect specific expression profiles of miRNAs in various biological processes and cancer; reveal asymmetric processing of small RNAs from rRNAs, snoRNAs, snRNA, and tRNAs; and even provide evidence for human miRNA-offset RNAs [65, 142, 143]. Although adapter ligation bias which lead to reverse transcription bias or amplification bias still need to be optimized [144, 145], small RNA-seq currently remains a high-efficiency way to detect and discover novel small ncRNAs.
A recent study provided a method to detect small ncRNAs in a single cell and the method was named as Small-seq [66]. In brief, single cell is lysed, and 5.8S rRNA is masked with a complementary oligonucleotide during adapter ligation. Then 3′ adapters are ligated to small RNAs, and unligated adapters are subsequently digested. The 5′ adapters containing a unique molecular identifier (UMI) are ligated, and reverse transcription is carried out. In the original article, the method captured a complex set of small RNAs, including miRNAs, fragments of tRNAs, and snoRNAs [66].

Single-cell RNA sequencing (scRNA-seq)

The fundamental unit of an organism is a single cell. Along with in-depth studies on development and disease occurrence, there is a growing sense that some single cells possess nonnegligible abilities that can affect organic growth or lead to the downfall of the entire organism [146]. It is helpful for researchers to further understand the mechanisms of growth or disease progression by revealing the gene expression pattern of specific single cells. However, the sample sizes from a single cell are insufficient for general RNA-seq, which has led to the development of scRNA-seq methods (Fig. 2b(I)). In addition, scRNA-seq techniques are also appropriate for other small samples, such as limited clinical patient samples or cells sorted with fluorescence-activated cell sorting (FACS) [61, 147].
Previous scRNA-seq techniques include Smart-seq [60, 148], designed primer-based sequencing (DP-seq) [61], and Quartz-seq [62], and each of them exhibits prominent advantages and disadvantages. Smart-seq is a method based on a full-length cDNA amplification strategy (Fig. 2b(II)) [60]. In this approach, polyadenylated RNAs are reverse transcribed into a pool of cDNAs by oligo (dT) primers and Moloney murine leukemia virus reverse transcriptase (MMLV RT). As a result, the terminal transferase activity of MMLV can add several nontemplate C nucleotides to the 3′ ends of the reverse transcribed products when the reverse transcription reaction reaches the 5′ end of a template transcript during first-strand cDNA generation (Fig. 2b(II)). Then, the poly-cytidine overhangs are used to complete the double-strand cDNA generation, which ensures that the prepared library for scRNA-seq only contains full-length cDNAs. However, the lack of strand-specific identification and inability to read transcripts longer than 4 kb partly limit the application of this method [149]. Compared to Smart-seq, DP-seq shows the advantage of being to amplify RNAs from smaller size samples, as low as 50 pg, or from transcripts longer than 4 kb [61]. DP-seq uses a defined set of heptamer primers, which target regions less likely to form secondary structures and reside upstream of the unique regions on certain transcriptomes, and they amplify the majority of expressed transcripts from a limited number of RNAs [61]. In the original study, preparation of a DP-seq library successfully amplified over 80% of the mouse transcriptome with 44 heptamer primers. Moreover, this approach can also suppresse highly expressed rRNAs in the cDNA library and is able to detect transcripts at relatively low levels [61]. In addition, Quartz-seq is an alternative scRNA-seq method with reduced background noise that utilizes specially designed suppression polymerase chain reaction (PCR) primers to reduce the generation of unwanted side products [62].
Recent studies on scRNA-seq methods preferably focused on total RNA sequencing, which provided rich information on biological systems in addition to the abundance of mRNAs. Thus far, much efforts have been made to develop scRNA-seq techniques with full-length coverage or sensitivity to nonpolyadenylated RNAs. There are several scRNA-seq methods, such as Smart-seq, that can provide full-length coverage of transcripts [60]. Nevertheless, these methods fail to measure nonpolyadenylated transcripts due to oligo (dT) priming [60]. Single-cell universal poly(A)-independent sequencing (SUPeR-seq), which uses random primers with fixed anchor sequences to replace oligo (dT) primers for cDNA synthesis, has been reported for the detection of nonpolyadenylated RNAs, especially circRNAs, in a single cell with robust precision and accuracy (Fig. 2b(III)) [63]. In the original study, researchers discovered 2891 circRNAs and 913 novel linear RNAs in mouse preimplantation embryos using SUPeR-seq and deciphered regulation mechanism of circRNA during early embryonic development [63]. However, SUPeR-seq also exhibits relatively low sensitivity for nonpolyadenylated RNAs [64].
Random displacement amplification sequencing (RamDA-seq) is a full-length total RNA-sequencing method for analyzing single cells, but it has a high sensitivity for nonpolyadenylated RNAs [64]. This approach can measure not only polyadenylated but also nonpolyadenylated RNAs, including nascent RNAs, lncRNAs, circRNAs, and eRNAs, and it can uncover the dynamics of recursive splicing [64]. Furthermore, it can provide full-length coverage for extremely long transcripts (more than 10 kb). RamDA-seq simplifies the experimental procedure to amplify cDNA as early as possible by using a novel RT technology, RT with random displacement amplification (RT-RamDA), which aims to obtain higher capture efficiency of RNAs and global cDNA amplification for further sequencing (Fig. 2b(IV)). Moreover, not-so-random primers (NSRs) are used to enable random priming while preventing the synthesis of cDNA from rRNAs [64]. Analysis of mouse embryonic stem cells undergoing differentiation using RamDA-seq revealed the cell state-dependent expression of known and novel nonpolyadenylated RNAs, including nonpolyadenylated isoforms of the lncRNA Neat1, and the specific genome-wide eRNA expression patterns in single cells [64].

Nascent RNA-seq

RNA-seq is a revolutionary tool for transcriptome profiling that provides information on the dynamic changes of gene expression against different conditions or after exposure to different stimuli [58]. However, the traditional RNA-seq technique is generally performed to determine steady-state RNA levels, and changes in RNA transcription and decay rates cannot be easily distinguished [150]. Moreover, common RNA-seq also fails to provide efficient temporal information on RNA kinetics [150]. To address these issues, new sequencing methods for measuring nascent transcripts, as opposed to total RNAs, have been developed [151].
Nascent RNA-seq can reveal the temporal information of gene expression changes. Metabolic labeling and affinity purification of labeled nascent RNAs followed by RNA-seq is a well-known approach for analyzing nascent RNAs [151]. For example, global run-on sequencing (GRO-seq) labels nascent RNAs with 5Br-UTP, enabling labeled nascent RNAs to be immunoprecipitated with the antibody anti-Br-UTP; the isolated RNAs subsequently undergoes deep sequencing (Fig. 3a) [67]. By sequencing nascent RNAs, GRO-seq can also provide a genome-wide view of the location, orientation, and density of Pol II-engaged transcripts, revealing divergent transcription at active promoters that yield antisense ncRNAs [152]. In recent studies, labeling/purifying RNA analysis has also been used to detect nascent ncRNAs, including nascent circRNAs. Nevertheless, the conventional purification assay in GRO-seq is confounded by contamination due to nonspecific binding, which could possibly result in experimental bias [70].
Recently, innovative enrichment-free methods for nascent RNA detection have been developed, which avoid contamination induced by affinity purification [153]. These methods directly distinguish nascent RNA from total RNA in single-base resolution by marking the mapping reads of nascent RNAs with introduced base mutations. In brief, nascent transcripts are labeled by adding a thiol-labeled nucleoside (s4U or s6G) to cell culture media, and these newly synthesized RNAs can then be isolated and treated with specific chemical reagents, leading to a change in the base-pairing manner of metabolically incorporated nucleosides (Fig. 3b) [153]. For example, SLAM-seq uses nucleophilic substitution chemistry to induce s4U-to-C conversion in an RT-dependent manner [68], and TimeLapse-seq employs s4U-to-C conversion via an oxidative nucleophilic aromatic substitution reaction (Fig. 3b) [69]; however, this oxidation condition caused certain oxidative damage to guanine [69]. A recent study reported an improved method, AMUC-seq, which transformed s4U into a cytidine derivative using acrylonitrile (Fig. 3b) [70]. Compared to other enrichment-free methods for nascent RNA detection, AMUC-seq has been reported to be more efficient and reliable because it has a minimal influence on the base-pairing manner of other nucleosides and can quantitatively analyze RNA at the transcriptome scale [70].

Innovative techniques based on RNA location and interactome for functional ncRNA discovery

As discussed above, the vast majority of the human genome can be transcribed into ncRNAs; thus, it is important to reveal potentially functional ncRNAs that may play a role in certain biological processes, especially in cancer occurrence and development. It has been shown that ncRNAs are commonly folded into highly ordered structures that play a role within their interactome [154, 155]. Therefore, in this section, we will discuss the discovery and identification of functional ncRNAs based on their interaction networks and subcellular location levels, and we will provide some novel techniques that can be used to screen purposefully for functional ncRNAs.

RNA-chromatin interaction

An increasing number of studies have reported that diverse species of ncRNAs show regulatory functions in different layers of and gene expression. Many cnRNAs perform direct actions on chromatin, some of which may mediate genomic interactions predominantly in cis, whereas others are capable of acting extensively in trans [156158]. These findings suggest a common role of specific RNA-chromatin interactions in modulating gene expression. Global RNA interactions with DNA by deep sequencing (GRID-seq) is a method that aims to comprehensively detect and determine the localization of all potential chromatin-interacting RNAs [71]. This approach uses a bivalent linker to ligate RNA to DNA in situ in fixed nuclei (Fig. 4a). Briefly, cells are fixed with disuccinimidyl glutarate (DSG) and formaldehyde first to stabilize RNAs on chromatin. Then, nuclei are extracted, and DNA is digested in situ by the frequent 4-base cutter AluI. A specifically designed bivalent linker labeled by biotin that consists of single-stranded RNA (ssRNA) portions, to ligate RNA, and a double-stranded DNA (dsDNA) portion, to ligate DNA, is used to link RNAs to AluI-digested genomic DNAs. Then, the DNA-RNA complexes are purified, filtered, and sequenced. In the original article, GRID-seq performed in human, mouse, and Drosophila cells revealed a large set of tissue-specific coding and noncoding RNAs that bind to active promoters and enhancers, especially super-enhancers [71]. Interestingly, the study also exhibited a large number of snoRNAs interacting with chromatin, suggesting possibly important roles of snoRNAs at the chromatin level [71].
Other alternative techniques based on the ligation of RNA to DNA have been reported for detecting genome-wide RNA-chromatin interactions, including MARGI and its improved version iMARGI [72, 159], and chromatin-associated RNA sequencing (ChAR-seq) [73]. Analysis of chromatin-associated RNA (caRNA) sequencing by MARGI and iMARGI revealed that caRNAs not only are associated with genomic regions where they are generated (proximal interactions) but also are attached to distal genomic regions (distal interactions) on the same chromosomes or on other chromosomes (interchromosomal interactions) [72, 159]. Interestingly, transcription star sites (TSSs) were identified as the preferred genomic regions targeted by chromatin-associated ncRNAs through distal or interchromosomal interactions. ChAR-seq also uncovered a range of chromatin-associated RNAs, especially chromosome-specific dosage compensation ncRNAs, and genome-wide trans-associated RNAs, which are involved in cotranscriptional RNA processing (Fig. 4b) [73].
In addition to the sequencing methods for identification of global RNA-chromatin interactomes mentioned above, various techniques were developed to detect specific localization on chromatin of target RNAs [160162]. These techniques use hybridization of complementary oligonucleotides to pull down a single target RNA, and then NGS or mass spectrometry is performed to identify its DNA- or protein-binding partners.

RNA-RNA spatial interactions

Structured RNAs such as duplexes represent a feature that is critical for most steps in the gene expression pathway. Numerous characterized ncRNAs function via base pairing with target RNAs to control their biological activities, such as dynamic interactions involving snRNA-snRNA and snRNA-pre-mRNA during the assembly and disassembly of spliceosomes, interactions between snoRNAs and their target RNAs to guide RNA modification, and interactions between ncRNAs and mRNAs that regulate transcript turnover and translation. Thus far, an increasing number of sequencing techniques have been developed for global mapping of RNA-RNA interactions (Fig. 5).
RNA proximity ligation is a set of molecular biological techniques that can be used to analyze the conformation and spatial proximity of RNAs in cells [74]. The typical first steps in these approaches involves cross-linking biological samples with UV light or psoralen, which is followed by partial fragmentation of RNA, RNA-RNA ligation, library preparation, and high-throughput sequencing. UV light and psoralen are two widely used methods for sample preparation prior to proximity ligation: UV light treatment stabilizes and enriches the RNA duplexes that are bound to a specific protein or protein complex; however, psoralen is used to stabilize and enrich RNA-RNA interactions. Studies on RNA conformation have shown different emphases, as some approaches identified pairs of RNAs that are in direct contact or in close proximity with each other, while others recovered pairs of RNAs that are part of the same protein complex or subcellular compartment [163]. Alternative cross-linking methods provide alternative treatments for diverse purposes (Fig. 5). Cross-linking ligation and sequencing of hybrids (CLASH) is a relatively early method that uses UV cross-linking to capture direct RNA-RNA hybridization [74]. Compared to chemically cross-linking methods, which also induce extra protein-protein cross-linking, CLASH has the advantage of avoiding noise from protein intermediate-mediated interactions, and has been used to identify novel snoRNA-rRNA interactions in yeast [74], miRNA-mRNA interactions in human HEK293 cells [164], and piRNAs interactomes [164]. In another method, RNA immunoprecipitation and proximity ligation in tandem (RIPPLiT), sequential pull-down of components of exon junction complexes showed a mapping of mRNA conformations when bound to this complex [75]. Moreover, another approach, mapping the RNA interactome in vivo (MARIO), has identified RNA-RNA interactions in the vicinity of all RNA-binding proteins using a biotin-linked reagent [76].
Methods for identifying RNA-RNA interactions at the transcriptome scale by cross-linking with psoralen have been reported since 2016 [77, 79, 165]. Unlike CLASH, psoralen-based approaches do not depend on the pull-down of RNA-RNA interactions with a specific protein, and in principle, they can yield transcriptome-wide RNA interactomes. The methods using this principle of cross-linking RNAs are combined with different means to enrich cross-linked fragments, such as two-dimensional gel electrophoresis in PARIS (psoralen analysis of RNA interactions and structures) [77], digestion by RNase R in LIGR-seq (ligation of interacting RNA followed by high-throughput sequencing) [78], and biotin-streptavidin enrichment in SPLASH (sequencing of psoralen cross-linked, ligated, and selected hybrids) [79]. The psoralen cross-linking methods uncovered general properties of RNA-RNA interactomes in mammalian cells (Fig. 5). For example, PARIS uncovered alternative base pairing in intramolecular interactions, which suggests substantial structural heterogeneity in cells, and it also elucidated the structure produced by a repeat of adenosines in Xist in vivo [77]. LIGR-seq in HEK293 cells detected novel snRNA-snRNA and snoRNA-rRNA interactions [78]. More importantly, this approach also revealed that SNORD83B can regulate gene expression by binding to target mRNAs, revealing an unexpected function of these snoRNAs [78]. Psoralen cross-linking methods such as PARIS and SPLASH were also applied to detect dense networks of RNA-RNA interactions within viral genomes inside infected cells [166, 167].
A recent study reported a novel method, RNA in situ conformation sequencing (RIC) technology, for the global mapping of intra- and intermolecular RNA-RNA interactions (Fig. 5) [80]. Compared to the RNA ligation induced in vitro in previous methods, RIC-seq performs RNA proximity ligation in situ, and it enriches chimeric reads using a biotinylated cytidine phosphate (pCp-biotin) [80]. Briefly, the cells are cross-linked by formaldehyde, and then, RNA is randomly cut with micrococcal nuclease and dephosphorylated at 3′ overhangs. The 3′ ends are labeled with pCp-biotin and ligated to proximal 5′ overhangs under in situ and nondenaturing conditions. Total RNAs are fragmented in vitro, and RNAs containing C-biotin are enriched followed by conversion into cDNA libraries for sequencing. In the original article, RIC-seq was used to facilitate the generation of 3D RNA interaction maps in human cells, and it revealed global noncoding RNA targets, RNA topological domains, and trans-interacting hubs [80].
In addition to the sequencing methods using RNA proximity ligation, there are some other approaches without ligation that have been developed because of the possible limitation in efficiency of enzymatic ligations affected by short-range distances between RNA ends [81]. RNA proximity sequencing is a method based on massive-throughput RNA barcoding of particles in water-in–oil emulsion droplets [81]. In brief, this approach uniquely barcodes RNA in millions of subnuclear particles in parallel by a rapid vortexing step that combines fragmented nuclear particles with barcoded beads in a water-in-oil emulsion; then, the cDNA is sequenced. The detection of multiple RNAs in proximity to each other by RNA proximity sequencing distinguished RNA-dense and RNA-sparse compartments, and this technique is an alternative approach for studying the spatial organization of transcripts in the nucleus, including ncRNAs and their functional relevance.

RNAs in protein complexes or subcellular structures

The location of ncRNAs in cells is the primary determinant of their molecular functions. NcRNAs, especially lncRNAs, are often considered as chromatin-restricted modulators of gene transcription and chromatin structure [157, 158]. In addition, a rich population of cytoplasmic ncRNAs, such as extra lncRNAs and exonic circRNAs, have been reported to play roles in diverse biological processes, including translational regulation and signal transduction [8, 168]. Elution-based methods promise to detect RNAs at the transcriptome scale associated with all organelles of mammalian cells, and RNA maps of increasing resolution reveal a subcellular world of highly specific localization patterns.
In situ hybridization (ISH) is the most widely used method of RNA localization using labeled complementary oligonucleotide probes to visualize target RNAs [169, 170]. Single-molecule fluorescence ISH (smFISH) uses multiple probes to amplify the fluorescent signal for the detection of target RNAs at low levels, and it is thought of as the gold-standard technique for single-gene studies [169, 171]. In contrast to RNA smFISH, fluorescent in situ RNA sequencing (FISSEQ) offers in situ information at high-throughput levels [82]. In this approach, RNA is reverse transcribed in situ into cDNA in cross-linked cells and tissue samples, which is then analyzed by sequencing (Fig. 6a). However, compared to standard RNA-seq, FISSEQ also comes at the expense of lower read coverage, which reduces sensitivity for lowly expressed RNAs, especially ncRNAs [82]. Another alternative related technique, spatially resolved transcript amplicon readout mapping (STARmap), provided 3D locational information of RNA expression in intact tissue samples [172].
Biochemical cell fractionation is a fractionation-based method that physically isolates subcellular compartments and identifies their RNAs (Fig. 6b). These types of methods can be based on protein immunoprecipitation, intact organelle purification, or partitioning through sucrose gradients [173]. Then, RNA-seq (biochemical cell fractionation combined with RNA-seq, CeFra-seq) was performed to detect specific RNAs at the transcriptome scale [83]. Fractionation-based methods have high sensitivity for low-abundance transcripts due to aggregation across many cells; however, they are restricted by isolation protocols and the purity of resulting isolates, which possibly induce technical noise by contamination across fractions [24, 174, 175].
Recently, innovative techniques have been developed to overcome the deficiencies of conventional methods. A new fractionation-based method, APEX-RIP [84], was developed, and it combines APEX (engineered ascorbate peroxidase)-catalyzed proximity biotinylation [176] and RNA immunoprecipitation (RIP) [177] to map RNAs at vastly improved spatial resolution (Fig. 6c). In brief, APEX-catalyzed proximity biotinylation is targeted by genetic fusion to proteins from various subcellular compartments of interest. This is followed by protein-RNA crosslinking and RIP to pull down the biotinylated subcellular fraction for further high-throughput sequencing. Using this method, thousands of ncRNAs have been mapped to specific compartments without the need for purification of specific organelles, and it offers high specificity and sensitivity in targeting the transcriptome of membrane-bound organelles [84]. Moreover, in a recent study, a transcriptome-wide subcellular RNA atlas was generated by APEX-RIP [178].

NcRNA database

Various sequencing methods have provided systematic expression profiling of ncRNAs in diverse cells, tissues, and organisms, and they have mapped the interaction networks or subcellular localization of ncRNAs, which inform their potential biological functions. Databases provide important references based on theoretical analysis, sequencing data, and even experimental verification, which play a guiding role in the identification and functional investigation of ncRNAs. Here, we will introduce a series of ncRNA databases that emphasize basic ncRNA information, cancer-associated ncRNA expression patterns, or specific ncRNA interaction networks based on experimental techniques followed by high-throughput sequencing.
The correlations between ncRNA expression and cancer progression provide important hints whether a ncRNA could play a role in certain cancers. There are an increasing number of databases providing comprehensive associations between ncRNAs and human cancers, which are supported by sequencing data or even experiments, such as TANRIC [179], Lnc2Cancer 2.0 [180], lnCaNet [181], and LncRNADisease [182] for lncRNAs, CSCD [183], Circ2Traits [184], CircR2Disease [185], and MiOncoCirc [119] for circRNAs, miRCancer [186], SomamiR 2.0 [187], OncomiR [188], miRCancerdb [189], and dbDEMC 2.0 [190] for miRNAs and YM500v3 [191], tRF2Cancer [192], and MINbase v2.0 [193] for other small ncRNAs, as summarized in Table 2. A recently reported MiOncoCirc is the first database that mainly consists of circRNAs directly detected in tumor tissues [119]. It was established by detecting and characterizing circRNAs across more than 2000 cancer samples with an exome capture RNA sequencing protocol. In the article that originally described the process, candidate circRNAs identified from MiOncoCirc were determined to be useful as biomarkers for prostate cancer and were found to be detected in urine, suggesting that MiOncoCirc could be an alternative tool to uncover novel diagnostic biomarkers for clinical translational strategies [119]. Another interesting, recently reported database is SELER, which collects specific super-enhancer-associated lncRNA profiles from different cancers [195]. In addition, some databases document the basic annotation and functional information on ncRNAs, including lncRNA-associated resources LNCipedia [199], LNCediting [200], lncRNAdb v2. 0[201], circRNA-associated ones circAtlas [206], circBase [207], CIRCpedia v 2[208], TSCD [209], miRNA-associated ones starBase v2.0 [210], miRTarBase [211], miRmine [212], EVmiRNA [213], miRGate [214], miRBase [215], and even other small ncRNA-associated ones DASHR 2.0 [217]. A growing number of databases have undoubtedly played important roles in the discovery and investigation of novel functional ncRNAs.
Table 2
Database of ncRNAs
Cancer or basis
Database
Species
Website
Short description
Ref
Cancer
Lnc2Cancer v2.0
lncRNA
An updated database that provides comprehensive experimentally supported associations between lncRNAs and human cancers.
[180]
 
TANRIC
lncRNA
This database characterizes the expression profiles of lncRNAs in large patient cohorts of 20 cancer types, including TCGA and independent datasets (> 8000 samples overall).
[179]
 
lnCaNet
lncRNA
This database provides a comprehensive co-expression data resource which reveals the interactions between lncRNA and non-neighbouring cancer genes.
[181]
 
LncRNADisease 2.0
lncRNA
A database integrating comprehensive experimentally supported and predicted lncRNA-disease associations.
[182]
 
The Cancer LncRNome Atlas
lncRNA
An academic research database to explore the lncRNA alternations across multiple human cancer types.
[194]
 
SELER
lncRNA
A database of super-enhancer-associated lncRNA-directed transcriptional regulation in human cancers.
[195]
 
CSCD
circRNA
A database that focuses on distinguishing cancer-specific circRNAs from noncancerous circRNAs, and reports predicted cellular location, RBP sites, and ORFs.
[183]
 
Circ2Traits
circRNA
Provide cirRNA-disease association based on the interaction of circRNAs with disease-related miRNAs and SNP mapped on circRNA loci.
[184]
 
CircR2Disease
circRNA
Provide a comprehensive resource for circRNA deregulation in various diseases, containing 725 associations between 661 circRNAs and 100 diseases.
[185]
 
CircRNA disease
circRNA
A manually curated database of experimentally supported circRNA-disease associations.
[196]
 
MiOncoCirc
circRNA
circRNA detection in 2093 clinical human cancer samples using exome capture sequencing.
[119]
 
CircRiC
circRNA
A database focusing on lineage-specific circRNAs in 935 cancer cell lines including drug response.
[197]
 
miRCancer
miRNA
A database currently documents more than 9000 relationships between 57,984 miRNAs and 196 human cancers.
[186]
 
SomamiR 2.0
miRNA
A database of cancer somatic mutations in microRNAs (miRNA) and their target sites that potentially alter the interactions between miRNAs and competing endogenous RNAs (ceRNA).
[187]
 
OncomiR
miRNA
An online resource for exploring miRNA dysregulation in cancer.
[188]
 
miRCancerdb
miRNA
An easy-to-use database to investigate the microRNAs-dependent regulation of target genes involved in development of cancer.
[189]
 
miR2Disease
miRNA
A database aiming at providing a comprehensive resource of microRNA deregulation in various human diseases.
[198]
 
YM500v3
small ncRNA
A database which contains more than 8000 small RNA-seq dataseta and focuses on piRNAs, tRFs, snRNAs, snoRNAs, and miRNAs.
[191]
 
tRF2Cancer
small ncRNA
A web server to detect tRFs and their expression in multiple cancers.
[192]
 
MINTbase v2.0
Small ncRNA
A framework for the interactive exploration of mitochondrial and nuclear tRNA fragments.
[193]
Basis
LNCipedia
lncRNA
A public database for lncRNA sequence and annotation.
[199]
 
LNCediting
lncRNA
This database provides a comprehensive resource for the functional prediction of RNA editing in lncRNAs.
[200]
 
lncRNAdb v2.0
lncRNA
This database provides comprehensive annotations of eukaryotic lncRNAs.
[201]
 
LncRNAWiki
lncRNA
This database is a publicly editable and open-content platform for community curation of human lncRNAs.
[202]
 
LncBook
lncRNA
This database is a curated knowledgebase of human lncRNAs.
[203]
 
MONOCLdb
lncRNA
20,728 mouse lncRNA genes.
[204]
 
NONCODE
lncRNA
An interactive database that aims to present the most complete collection and annotation of ncRNAs especially lncRNAs from 17 species.
[205]
 
CircAtlas
circRNA
An integrated resource of one million highly accurate circular RNAs from 1070 vertebrate transcriptomes.
[206]
 
circBase
circRNA
A database containing thousands of recently identified circRNAs in eukaryotic cells.
[207]
 
CIRCpedia v2
circRNA
A database for comprehensive circRNA annotation from over 180 RNA-seq datasets across six different species.
[208]
 
TSCD
circRNA
A tissue-specific circRNA database from RNA-seq datasets and characterized the features of circRNAs in human and mouse.
[209]
 
starBase v2.0
miRNA
A database decoding miRNA-ceRNA, miRNA-ncRNA, and protein–RNA interaction networks from large-scale CLIP-Seq data.
[210]
 
miRTarBase
miRNA
A resource for experimentally validated microRNA-target interactions.
[211]
 
miRmine
miRNA
A database of human miRNA expression profiles.
[212]
 
EVmiRNA
miRNA
A database focusing on miRNA expression profiles in extracellular vesicles.
[213]
 
miRGate
miRNA
A curated database of human, mouse, and rat miRNA–mRNA targets.
[214]
 
miRBase
miRNA
A database containing microRNA sequences from 271 organisms: 38,589 hairpin precursors and 48,860 mature microRNAs.
[215]
 
DIANA-TarBase v8
miRNA
A reference database devoted to the indexing of experimentally supported miRNA targets.
[216]
 
DASHR 2.0
small ncRNA
A database that integrates human small ncRNA gene and mature products derived from all major RNA classes.
[217]
Several specific RNA-seq datasets have revealed the subcellular locations and potential interactomes of ncRNAs, which provide more real information than what is learned from bioinformatics prediction. There are some databases that provide high-quality RNA subcellular location resources in accordance with the results of subcellular compartment sequencing, such as RNALocate [218] and LncATLAS [219]. RNALocate documents more than 37,700 manually curated RNA subcellular location entries with experimental evidence, and it has data on 65 organisms, 42 subcellular locations (such as cytoplasm, nucleus, endoplasmic reticulum), and 9 RNA categories, such as lncRNAs [218]. However, thus far, few interactome database of ncRNAs except miRNA [210, 211, 216], has been established based on experimental techniques and sequencing. NPInter v3.0 is a database of ncRNA-associated interactions based on experimental techniques followed by high-throughput sequencing, such as crosslinking and immunoprecipitation followed by deep sequencing (CLIP-seq) [220], and chromatin isolation by RNA purification followed by high-throughput sequencing (ChIRP-seq) [161, 221]. NPInter v3.0 documented approximately 500,000 interactions in 188 tissues (or cell lines) from 68 kinds of experiments and predicted the functions of lncRNAs in humans on the basis of their interactions in the database [221]. Furthermore, a database of RNA interactomes identified by sequencing at the transcriptome scale is lacking, and it is needed for identification of novel functional ncRNAs.
Due to their highly tissue-specific expression patterns identified by various sequencing techniques and their key roles in regulating biological activity in cancer, ncRNAs, including miRNAs, lncRNAs, and circRNAs, are generally considered to have potential as novel biomarkers for cancer diagnosis [20, 222, 223]. This section aims to present new developments in diagnostic kits for cancer diagnosis by the analysis of cancer-related ncRNAs.
Cancer seriously threatens the human life and gives rise to an enormous burden on society. However, the incidence and mortality of cancer could be decreased effectively by preventative measures, including early detection tests and monitoring of cancer prognosis. Therefore, searching for novel biomarkers that are easy to use, are not invasive, and exhibit high sensitivity, specificity, and stability for cancer diagnosis and prognosis has been a key clinical translational strategy. In addition to the features of specific expression patterns, some types of ncRNAs, such as miRNAs, lncRNAs, and circRNAs, have also been shown to be relatively stable in serum, plasma, saliva, or urine, which can be easier to collect and is less harmful or invasive for patients than other collection methods. In the past few years, seeking novel biomarkers in cancer diagnosis has mainly focused on miRNAs [224]. Recently, growing research has shown that other ncRNAs, especially lncRNAs and circRNAs, could also serve as a hallmark of carcinomas.
MiRNAs, lncRNAs, and circRNAs have been observed to have highly specific expression patterns in diverse types of cancers, and this aberrant expression usually occurs in certain tumor cells or cancer tissues at a specific stage of disease progression [13, 14, 225]. According to patent searches in resources such as the EPO (https://​worldwide.​espacenet.​com), there are growing uses of these three types of ncRNAs in the preparation of diagnostic kits for various cancers, including hepatocellular, cervical, stomach, liver, breast, prostatic, and bladder cancers (Table 3). Generally, detecting cancer-related nucleic acids in patient samples using qRT-PCR with specific primers or probes is the main method for diagnosis based on ncRNAs, which is also the primary approach for diagnosing disease in the recent COVID-19 (CoronaVirusDisease2019) pandemic [226]. For example, a recent patent provided a circRNA hsacirc_0028185 qPCR assay kit for the diagnosis of hepatocellular carcinoma. By detecting expression changes of serum hsacirc_0028185, it is possible to assess the occurrence and development of hepatocellular carcinoma. Another sample is that lncRNA-AC006159.3 in the blood could be used for the diagnostic kit to rapidly speculate the cetuximab-resistant possibility of rectal cancer. Briefly, the lower the expression level of lncRNA-AC006159.3, the higher the possibility of cetuximab resistance. In addition, a patent provided application of miRNA-410 in preparation of a prostatic cancer diagnostic kit.
Table 3
Developments in diagnostic kits for cancer diagnosis (EPO https://​worldwide.​espacenet.​com)
Species
Name
Expression in cancer
Diseases
Application
Patent number
circRNA
hsacirc_0028185
Up
Hepatocellular carcinoma
Cancer auxiliary diagnosis
CN111004850A (2020)
circRNA
hsa_circ_001477
Up
Gastric cancer
Cancer diagnosis
CN110129324A (2019)
circRNA
hsa_circRNA_012515
Up
Non-small cell lung cancer
Cancer diagnosis
CN110592223A (2019)
circRNA
hsa_circRNA_405124 or hsa_circ_0012152
Up
Leukemia
Cancer early diagnosis
CN109593859A (2019)
circRNA
circ_104075
Up
Liver cancer
Cancer diagnosis
CN109161595A (2019)
circRNA
circ3823
Up
Colorectal cancer
Cancer early diagnosis
CN110592220A (2019)
circRNA
hsa_circ_0021977
Up
Breast cancer
Cancer diagnosis
CN109022583A (2018)
circRNA
hsa_circ_0012755
Up
Prostate cancer
Cancer diagnosis
CN108624688A (2018)
circRNA
circ_0047921, circ_0007761 and circ_0056285
Up
Non-small cell lung cancer
Cancer early diagnosis
CN108179190A (2018)
circRNA
hsa-circRPL15-001
Up
Chronic lymphocytic leukemia
Cancer diagnosis
CN109055564A (2018)
circRNA
has_circ_0117909
Up
Acute lymphoblastic leukemia
Cancer diagnosis
CN107937522A (2017)
has_circ_0005720
Down
circRNA
cRNA-ZFR
Up
Bladder cancer
Cancer diagnosis
CN106011139A (2016)
lncRNA
lncRNA-AC006159.3
Down
Colorectal cancer
Cetuximab-resistance diagnosis
CN108949993A
(2018)
lncRNA
lncRNAXLOC_004122, Linc00467 and lncRNAA1049452
Up
Breast cancer
Cancer bone metastasis diagnosis
CN107699619A (2017)
lncRNA
LncRNA GENE NO.9
Up
Bladder cancer
Cancer diagnosis
CN107267636A (2017)
lncRNA
LINC00516
Up
Lung cancer
Cancer or cancer metastasis diagnosis
CN108998528A (2018)
lncRNA
LSAMP-AS1
Up
Gastric cancer
Cancer diagnosis
CN110628915A (2019)
miRNA
miRNA-4692
Down
Hepatocellular carcinoma
Cancer diagnosis
(2018)
miRNA
miRNA-1266
Up
Endometrial carcinoma
Cancer diagnosis
CN105907883A
(2016)
miRNA
miR-320
Down
Cervical cancer
Cancer early diagnosis
CN105506076A
(2016)
miRNA
miRNA-2116
Up
Lung adenocarcinoma
Cancer metastasis diagnosis
CN104774966A
(2015)
miRNA
miRNA-410
Up
Prostate cancer
Cancer diagnosis
CN104651492A
(2015)
miRNA
miRNA-1262
Up
Acute myeloid leukemia
Cancer diagnosis
CN105063052A
(2015)
It is noteworthy that the same RNA may be aberrantly expressed in many types of cancers, which allows the same RNA to be used to diagnose different kinds of cancers. Moreover, the RNA-seq data show that a number of diverse species of ncRNAs are dysregulated in cancer samples compared to normal tissues, suggesting that diagnostic kits can be designed to detect multiple ncRNAs at the same time for more efficient cancer diagnosis.

Conclusion and perspective

The tissue-specific expression patterns, complicated regulatory networks, and emerging roles all suggest that ncRNAs are not simply debris or side products of transcriptional processes or aberrant splicing; rather, they are important regulatory molecules [102]. New technologies have endlessly emerged with different goals in ncRNA identification in multiple areas of research, including detection of ncRNA expression at the transcriptome scale, identification of novel ncRNA categories, searching for potential functional RNA within specific subcellular compartments, or discovering applicable biomarkers for cancer diagnosis. There are also some ncRNA-associated databases that provide multiple ncRNA information to enable further functional RNA investigations. Moreover, with the increasing number of studies on cancer-associated ncRNAs, translational applications of specific ncRNA identification for clinical diagnosis have been developed, such as diagnostic kits.
It is noteworthy that different RNA categories can be generated from the same regions of DNA, and they can share the same sequences. In addition to small ncRNAs derived from snoRNAs, tRNAs, or rRNAs, which can play a role in the miRNA-like pathway, some long nonpolyadenylated transcripts, such as sno-lncRNAs and circRNAs, have also been found to be generated from the genetic sequences of well-known ncRNAs [10, 37, 42, 126, 139]. Sno-lncRNAs have the same classical stem-loop as snoRNAs originating from the same genomic regions. However, both types of ncRNAs have been verified to have individual functions and to be important regulatory molecules in biological processes [42]. Another example is circANRIL, a circRNA formed from the lncRNA ANRIL, which performs functions in apoptosis and proliferation that are the opposite of the functions of ANRIL [227]. Taken together, these results suggest that transcripts derived from canonical DNA regions have functions in addition to their classical ones by interacting with nonclassical binding molecules or by being located in novel components, which indicates that the transcriptome extends far beyond the genome. A larger RNA world is waiting for us to explore.
High-throughput sequencing with purposeful sample preparation not only has uncovered novel species of ncRNAs but also has mapped the interaction networks and subcellular locations of ncRNAs [221]. In accordance with sequencing data of the RNA-associated interactome and RNA subcellular locations that have been determined at the transcriptome scale, a series of noncoding transcripts, especially lncRNAs and circRNAs, exhibit specific distributions in organelles, protein complexes, or subcellular structures [228]. These results further indicate that ncRNAs are functional molecules playing roles in specific compartments, providing a pool of candidates for us to search for specifically functional ncRNAs. Using these sequencing methods, a huge number of snoRNAs have been found to be enriched on chromatin, robustly suggesting that other potential functions of these well-known small ncRNAs are located in a nonclassical compartment [71]. However, the functions of chromatin-associated snoRNAs remain unanswered. In addition, thousands of potential functional ncRNAs with specific interactions or locations have been discovered and await further investigation.
Due to the features of specific expression patterns in cancers and relatively high stability in serum, plasma, saliva, or urine, ncRNAs especially miRNAs, lncRNAs, and circRNAs are generally considered to have potential as non-invasive diagnostic biomarkers for cancers. A growing number of researehes have provided the suitable ncRNA candidates for diagnosis of different cancers and increasing patents about preparation of ncRNA diagnostic kits for cancer diagnosis have been approved. However, most of these candidate ncRNAs are still in the preclinical stages. In addition, the results of some studies evaluating the potential of ncRNAs as biomarkers are conflicting [20]. Thus, more accurate evaluation of RNA expression pattern in larger cohorts of clinical data are needed to reconcile the controversies.

Acknowledgements

Not applicable.
Not applicable for this review.
Not applicable for this article.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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
1.
Zurück zum Zitat Warner, Soeiro R, Birnboim HC, Girard M, Darnell JE. Rapidly labeled HeLa cell nuclear RNA. I. Identification by zone sedimentation of a heterogeneous fraction separate from ribosomal precursor RNA. J Mol Biol. 1966;19:349–61.PubMed Warner, Soeiro R, Birnboim HC, Girard M, Darnell JE. Rapidly labeled HeLa cell nuclear RNA. I. Identification by zone sedimentation of a heterogeneous fraction separate from ribosomal precursor RNA. J Mol Biol. 1966;19:349–61.PubMed
2.
Zurück zum Zitat Weinberg RA, Penman S. Small molecular weight monodisperse nuclear RNA. J Mol Biol. 1968;38:289–304.PubMed Weinberg RA, Penman S. Small molecular weight monodisperse nuclear RNA. J Mol Biol. 1968;38:289–304.PubMed
3.
Zurück zum Zitat Zieve G, Penman S. Small RNA species of the HeLa cell: metabolism and subcellular localization. Cell. 1976;8:19–31.PubMed Zieve G, Penman S. Small RNA species of the HeLa cell: metabolism and subcellular localization. Cell. 1976;8:19–31.PubMed
4.
Zurück zum Zitat Eddy SR. Non-coding RNA genes and the modern RNA world. Nat Rev Genet. 2001;2:919–29.PubMed Eddy SR. Non-coding RNA genes and the modern RNA world. Nat Rev Genet. 2001;2:919–29.PubMed
5.
Zurück zum Zitat Pachnis V, Brannan CI, Tilghman SM. The structure and expression of a novel gene activated in early mouse embryogenesis. EMBO J. 1988;7:673–81.PubMedPubMedCentral Pachnis V, Brannan CI, Tilghman SM. The structure and expression of a novel gene activated in early mouse embryogenesis. EMBO J. 1988;7:673–81.PubMedPubMedCentral
6.
Zurück zum Zitat Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17:47–62.PubMed Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17:47–62.PubMed
7.
Zurück zum Zitat Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–8.PubMed Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–8.PubMed
8.
Zurück zum Zitat Li X, Yang L, Chen LL. The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 2018;71:428–42.PubMed Li X, Yang L, Chen LL. The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 2018;71:428–42.PubMed
9.
Zurück zum Zitat Schimmel P. The emerging complexity of the tRNA world: mammalian tRNAs beyond protein synthesis. Nat Rev Mol Cell Biol. 2018;19:45–58.PubMed Schimmel P. The emerging complexity of the tRNA world: mammalian tRNAs beyond protein synthesis. Nat Rev Mol Cell Biol. 2018;19:45–58.PubMed
10.
Zurück zum Zitat Ender C, Krek A, Friedlander MR, Beitzinger M, Weinmann L, Chen W, et al. A human snoRNA with microRNA-like functions. Mol Cell. 2008;32:519–28.PubMed Ender C, Krek A, Friedlander MR, Beitzinger M, Weinmann L, Chen W, et al. A human snoRNA with microRNA-like functions. Mol Cell. 2008;32:519–28.PubMed
11.
Zurück zum Zitat Lambert M, Benmoussa A, Provost P. Small non-coding RNAs derived from eukaryotic ribosomal RNA. Noncoding RNA. 2019;5:16.PubMedCentral Lambert M, Benmoussa A, Provost P. Small non-coding RNAs derived from eukaryotic ribosomal RNA. Noncoding RNA. 2019;5:16.PubMedCentral
12.
13.
Zurück zum Zitat Huarte M. The emerging role of lncRNAs in cancer. Nat Med. 2015;21:1253–61.PubMed Huarte M. The emerging role of lncRNAs in cancer. Nat Med. 2015;21:1253–61.PubMed
14.
Zurück zum Zitat Dong Y, He D, Peng Z, Peng W, Shi W, Wang J, et al. Circular RNAs in cancer: an emerging key player. J Hematol Oncol. 2017;10:2.PubMedPubMedCentral Dong Y, He D, Peng Z, Peng W, Shi W, Wang J, et al. Circular RNAs in cancer: an emerging key player. J Hematol Oncol. 2017;10:2.PubMedPubMedCentral
15.
Zurück zum Zitat Pekarsky Y, Balatti V, Palamarchuk A, Rizzotto L, Veneziano D, Nigita G, et al. Dysregulation of a family of short noncoding RNAs, tsRNAs, in human cancer. Proc Natl Acad Sci U S A. 2016;113:5071–6.PubMedPubMedCentral Pekarsky Y, Balatti V, Palamarchuk A, Rizzotto L, Veneziano D, Nigita G, et al. Dysregulation of a family of short noncoding RNAs, tsRNAs, in human cancer. Proc Natl Acad Sci U S A. 2016;113:5071–6.PubMedPubMedCentral
16.
Zurück zum Zitat Li Y, Zeng C, Hu J, Pan Y, Shan Y, Liu B, et al. Long non-coding RNA-SNHG7 acts as a target of miR-34a to increase GALNT7 level and regulate PI3K/Akt/mTOR pathway in colorectal cancer progression. J Hematol Oncol. 2018;11:89.PubMedPubMedCentral Li Y, Zeng C, Hu J, Pan Y, Shan Y, Liu B, et al. Long non-coding RNA-SNHG7 acts as a target of miR-34a to increase GALNT7 level and regulate PI3K/Akt/mTOR pathway in colorectal cancer progression. J Hematol Oncol. 2018;11:89.PubMedPubMedCentral
17.
Zurück zum Zitat Fort RS, Matho C, Oliveira-Rizzo C, Garat B, Sotelo-Silveira JR, Duhagon MA. An integrated view of the role of miR-130b/301b miRNA cluster in prostate cancer. Exp Hematol Oncol. 2018;7:10.PubMedPubMedCentral Fort RS, Matho C, Oliveira-Rizzo C, Garat B, Sotelo-Silveira JR, Duhagon MA. An integrated view of the role of miR-130b/301b miRNA cluster in prostate cancer. Exp Hematol Oncol. 2018;7:10.PubMedPubMedCentral
18.
Zurück zum Zitat Basati G, Khaksarian M, Abbaszadeh S, Lashgarian HE, Marzban A. Cancer stem cells and nanotechnological approaches for eradication. Stem Cell Investig. 2019;6:38.PubMedPubMedCentral Basati G, Khaksarian M, Abbaszadeh S, Lashgarian HE, Marzban A. Cancer stem cells and nanotechnological approaches for eradication. Stem Cell Investig. 2019;6:38.PubMedPubMedCentral
19.
Zurück zum Zitat Wang WT, Chen TQ, Zeng ZC, Pan Q, Huang W, Han C, et al. The lncRNA LAMP5-AS1 drives leukemia cell stemness by directly modulating DOT1L methyltransferase activity in MLL leukemia. J Hematol Oncol. 2020;13:78.PubMedPubMedCentral Wang WT, Chen TQ, Zeng ZC, Pan Q, Huang W, Han C, et al. The lncRNA LAMP5-AS1 drives leukemia cell stemness by directly modulating DOT1L methyltransferase activity in MLL leukemia. J Hematol Oncol. 2020;13:78.PubMedPubMedCentral
20.
Zurück zum Zitat Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 2019;12:55.PubMedPubMedCentral Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol. 2019;12:55.PubMedPubMedCentral
21.
22.
Zurück zum Zitat Rastgoo N, Abdi J, Hou J, Chang H. Role of epigenetics-microRNA axis in drug resistance of multiple myeloma. J Hematol Oncol. 2017;10:121.PubMedPubMedCentral Rastgoo N, Abdi J, Hou J, Chang H. Role of epigenetics-microRNA axis in drug resistance of multiple myeloma. J Hematol Oncol. 2017;10:121.PubMedPubMedCentral
23.
Zurück zum Zitat Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, et al. The transcriptional landscape of the mammalian genome. Science. 2005;309:1559–63.PubMed Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, et al. The transcriptional landscape of the mammalian genome. Science. 2005;309:1559–63.PubMed
24.
Zurück zum Zitat Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012;489:101–8.PubMedPubMedCentral Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012;489:101–8.PubMedPubMedCentral
25.
Zurück zum Zitat Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316:1484–8.PubMed Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316:1484–8.PubMed
26.
Zurück zum Zitat Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705.PubMed Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705.PubMed
27.
Zurück zum Zitat Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–37.PubMed Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–37.PubMed
28.
Zurück zum Zitat Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60.PubMedPubMedCentral Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature. 2007;448:553–60.PubMedPubMedCentral
29.
Zurück zum Zitat Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature. 2009;459:108–12.PubMedPubMedCentral Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature. 2009;459:108–12.PubMedPubMedCentral
30.
Zurück zum Zitat Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–7.PubMedPubMedCentral Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–7.PubMedPubMedCentral
31.
Zurück zum Zitat Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A. 2009;106:11667–72.PubMedPubMedCentral Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A. 2009;106:11667–72.PubMedPubMedCentral
32.
Zurück zum Zitat Guttman M, Garber M, Levin JZ, Donaghey J, Robinson J, Adiconis X, et al. Ab initio reconstruction of cell type-specific transcriptomes in mouse reveals the conserved multi-exonic structure of lincRNAs. Nat Biotechnol. 2010;28:503–10.PubMedPubMedCentral Guttman M, Garber M, Levin JZ, Donaghey J, Robinson J, Adiconis X, et al. Ab initio reconstruction of cell type-specific transcriptomes in mouse reveals the conserved multi-exonic structure of lincRNAs. Nat Biotechnol. 2010;28:503–10.PubMedPubMedCentral
33.
Zurück zum Zitat De Santa F, Barozzi I, Mietton F, Ghisletti S, Polletti S, Tusi BK, et al. A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol. 2010;8:e1000384.PubMedPubMedCentral De Santa F, Barozzi I, Mietton F, Ghisletti S, Polletti S, Tusi BK, et al. A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol. 2010;8:e1000384.PubMedPubMedCentral
34.
Zurück zum Zitat Lai F, Orom UA, Cesaroni M, Beringer M, Taatjes DJ, Blobel GA, et al. Activating RNAs associate with mediator to enhance chromatin architecture and transcription. Nature. 2013;494:497–501.PubMedPubMedCentral Lai F, Orom UA, Cesaroni M, Beringer M, Taatjes DJ, Blobel GA, et al. Activating RNAs associate with mediator to enhance chromatin architecture and transcription. Nature. 2013;494:497–501.PubMedPubMedCentral
35.
Zurück zum Zitat Jiao W, Chen Y, Song H, Li D, Mei H, Yang F, et al. HPSE enhancer RNA promotes cancer progression through driving chromatin looping and regulating hnRNPU/p300/EGR1/HPSE axis. Oncogene. 2018;37:2728–45.PubMed Jiao W, Chen Y, Song H, Li D, Mei H, Yang F, et al. HPSE enhancer RNA promotes cancer progression through driving chromatin looping and regulating hnRNPU/p300/EGR1/HPSE axis. Oncogene. 2018;37:2728–45.PubMed
36.
Zurück zum Zitat Li S, Xu Z, Sheng J. tRNA-derived small RNA: a novel regulatory small non-coding RNA. Genes (Basel). 2018, 9:246. Li S, Xu Z, Sheng J. tRNA-derived small RNA: a novel regulatory small non-coding RNA. Genes (Basel). 2018, 9:246.
38.
Zurück zum Zitat Danan M, Schwartz S, Edelheit S, Sorek R. Transcriptome-wide discovery of circular RNAs in archaea. Nucleic Acids Res. 2012;40:3131–42.PubMed Danan M, Schwartz S, Edelheit S, Sorek R. Transcriptome-wide discovery of circular RNAs in archaea. Nucleic Acids Res. 2012;40:3131–42.PubMed
39.
Zurück zum Zitat Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17:205–11.PubMed Chen LL. The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 2016;17:205–11.PubMed
41.
42.
Zurück zum Zitat Yin QF, Yang L, Zhang Y, Xiang JF, Wu YW, Carmichael GG, et al. Long noncoding RNAs with snoRNA ends. Mol Cell. 2012;48:219–30.PubMed Yin QF, Yang L, Zhang Y, Xiang JF, Wu YW, Carmichael GG, et al. Long noncoding RNAs with snoRNA ends. Mol Cell. 2012;48:219–30.PubMed
43.
Zurück zum Zitat Dinger ME, Pang KC, Mercer TR, Mattick JS. Differentiating protein-coding and noncoding RNA: challenges and ambiguities. PLoS Comput Biol. 2008;4:e1000176.PubMedPubMedCentral Dinger ME, Pang KC, Mercer TR, Mattick JS. Differentiating protein-coding and noncoding RNA: challenges and ambiguities. PLoS Comput Biol. 2008;4:e1000176.PubMedPubMedCentral
44.
Zurück zum Zitat Anderson DM, Anderson KM, Chang CL, Makarewich CA, Nelson BR, McAnally JR, et al. A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell. 2015;160:595–606.PubMedPubMedCentral Anderson DM, Anderson KM, Chang CL, Makarewich CA, Nelson BR, McAnally JR, et al. A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell. 2015;160:595–606.PubMedPubMedCentral
45.
Zurück zum Zitat Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017;68:171–84.PubMed Huang JZ, Chen M, Chen D, Gao XC, Zhu S, Huang H, et al. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017;68:171–84.PubMed
46.
Zurück zum Zitat Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, et al. Translation of CircRNAs. Mol Cell. 2017;66:9–21.PubMedPubMedCentral Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, et al. Translation of CircRNAs. Mol Cell. 2017;66:9–21.PubMedPubMedCentral
47.
Zurück zum Zitat Zhang M, Zhao K, Xu X, Yang Y, Yan S, Wei P, et al. A peptide encoded by circular form of LINC-PINT suppresses oncogenic transcriptional elongation in glioblastoma. Nat Commun. 2018;9:4475.PubMedPubMedCentral Zhang M, Zhao K, Xu X, Yang Y, Yan S, Wei P, et al. A peptide encoded by circular form of LINC-PINT suppresses oncogenic transcriptional elongation in glioblastoma. Nat Commun. 2018;9:4475.PubMedPubMedCentral
48.
Zurück zum Zitat Brannan CI, Dees EC, Ingram RS, Tilghman SM. The product of the H19 gene may function as an RNA. Mol Cell Biol. 1990;10:28–36.PubMedPubMedCentral Brannan CI, Dees EC, Ingram RS, Tilghman SM. The product of the H19 gene may function as an RNA. Mol Cell Biol. 1990;10:28–36.PubMedPubMedCentral
49.
Zurück zum Zitat Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–26.PubMed Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–26.PubMed
50.
Zurück zum Zitat Novikova IV, Hennelly SP, Sanbonmatsu KY. Structural architecture of the human long non-coding RNA, steroid receptor RNA activator. Nucleic Acids Res. 2012;40:5034–51.PubMedPubMedCentral Novikova IV, Hennelly SP, Sanbonmatsu KY. Structural architecture of the human long non-coding RNA, steroid receptor RNA activator. Nucleic Acids Res. 2012;40:5034–51.PubMedPubMedCentral
51.
Zurück zum Zitat Duret L, Chureau C, Samain S, Weissenbach J, Avner P. The Xist RNA gene evolved in eutherians by pseudogenization of a protein-coding gene. Science. 2006;312:1653–5.PubMed Duret L, Chureau C, Samain S, Weissenbach J, Avner P. The Xist RNA gene evolved in eutherians by pseudogenization of a protein-coding gene. Science. 2006;312:1653–5.PubMed
52.
Zurück zum Zitat Lin MF, Jungreis I, Kellis M. PhyloCSF: a comparative genomics method to distinguish protein coding and non-coding regions. Bioinformatics. 2011;27:i275–82.PubMedPubMedCentral Lin MF, Jungreis I, Kellis M. PhyloCSF: a comparative genomics method to distinguish protein coding and non-coding regions. Bioinformatics. 2011;27:i275–82.PubMedPubMedCentral
53.
Zurück zum Zitat Wang L, Park HJ, Dasari S, Wang S, Kocher JP, Li W. CPAT: coding-potential assessment tool using an alignment-free logistic regression model. Nucleic Acids Res. 2013;41:e74.PubMedPubMedCentral Wang L, Park HJ, Dasari S, Wang S, Kocher JP, Li W. CPAT: coding-potential assessment tool using an alignment-free logistic regression model. Nucleic Acids Res. 2013;41:e74.PubMedPubMedCentral
54.
Zurück zum Zitat El-Gebali S, Mistry J, Bateman A, Eddy SR, Luciani A, Potter SC, et al. The Pfam protein families database in 2019. Nucleic Acids Res. 2019;47:D427–32.PubMed El-Gebali S, Mistry J, Bateman A, Eddy SR, Luciani A, Potter SC, et al. The Pfam protein families database in 2019. Nucleic Acids Res. 2019;47:D427–32.PubMed
55.
Zurück zum Zitat Malone B, Atanassov I, Aeschimann F, Li X, Grosshans H, Dieterich C. Bayesian prediction of RNA translation from ribosome profiling. Nucleic Acids Res. 2017;45:2960–72.PubMedPubMedCentral Malone B, Atanassov I, Aeschimann F, Li X, Grosshans H, Dieterich C. Bayesian prediction of RNA translation from ribosome profiling. Nucleic Acids Res. 2017;45:2960–72.PubMedPubMedCentral
56.
Zurück zum Zitat Mockler TC, Chan S, Sundaresan A, Chen H, Jacobsen SE, Ecker JR. Applications of DNA tiling arrays for whole-genome analysis. Genomics. 2005;85:1–15.PubMed Mockler TC, Chan S, Sundaresan A, Chen H, Jacobsen SE, Ecker JR. Applications of DNA tiling arrays for whole-genome analysis. Genomics. 2005;85:1–15.PubMed
57.
Zurück zum Zitat Yan B, Wang ZH, Guo JT. The research strategies for probing the function of long noncoding RNAs. Genomics. 2012;99:76–80.PubMed Yan B, Wang ZH, Guo JT. The research strategies for probing the function of long noncoding RNAs. Genomics. 2012;99:76–80.PubMed
58.
59.
Zurück zum Zitat Mercer TR, Gerhardt DJ, Dinger ME, Crawford J, Trapnell C, Jeddeloh JA, et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol. 2011;30:99–104.PubMedPubMedCentral Mercer TR, Gerhardt DJ, Dinger ME, Crawford J, Trapnell C, Jeddeloh JA, et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol. 2011;30:99–104.PubMedPubMedCentral
60.
Zurück zum Zitat Ramskold D, Luo S, Wang YC, Li R, Deng Q, Faridani OR, et al. Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells. Nat Biotechnol. 2012;30:777–82.PubMedPubMedCentral Ramskold D, Luo S, Wang YC, Li R, Deng Q, Faridani OR, et al. Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells. Nat Biotechnol. 2012;30:777–82.PubMedPubMedCentral
61.
Zurück zum Zitat Bhargava V, Ko P, Willems E, Mercola M, Subramaniam S. Quantitative transcriptomics using designed primer-based amplification. Sci Rep. 2013;3:1740.PubMedPubMedCentral Bhargava V, Ko P, Willems E, Mercola M, Subramaniam S. Quantitative transcriptomics using designed primer-based amplification. Sci Rep. 2013;3:1740.PubMedPubMedCentral
62.
Zurück zum Zitat Sasagawa Y, Nikaido I, Hayashi T, Danno H, Uno KD, Imai T, et al. Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity. Genome Biol. 2013;14:R31.PubMedPubMedCentral Sasagawa Y, Nikaido I, Hayashi T, Danno H, Uno KD, Imai T, et al. Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity. Genome Biol. 2013;14:R31.PubMedPubMedCentral
63.
Zurück zum Zitat Fan X, Zhang X, Wu X, Guo H, Hu Y, Tang F, et al. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 2015;16:148.PubMedPubMedCentral Fan X, Zhang X, Wu X, Guo H, Hu Y, Tang F, et al. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 2015;16:148.PubMedPubMedCentral
64.
Zurück zum Zitat Hayashi T, Ozaki H, Sasagawa Y, Umeda M, Danno H, Nikaido I. Single-cell full-length total RNA sequencing uncovers dynamics of recursive splicing and enhancer RNAs. Nat Commun. 2018;9:619.PubMedPubMedCentral Hayashi T, Ozaki H, Sasagawa Y, Umeda M, Danno H, Nikaido I. Single-cell full-length total RNA sequencing uncovers dynamics of recursive splicing and enhancer RNAs. Nat Commun. 2018;9:619.PubMedPubMedCentral
65.
Zurück zum Zitat Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129:1401–14.PubMedPubMedCentral Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129:1401–14.PubMedPubMedCentral
66.
Zurück zum Zitat Hagemann-Jensen M, Abdullayev I, Sandberg R, Faridani OR. Small-seq for single-cell small-RNA sequencing. Nat Protoc. 2018;13:2407–24.PubMed Hagemann-Jensen M, Abdullayev I, Sandberg R, Faridani OR. Small-seq for single-cell small-RNA sequencing. Nat Protoc. 2018;13:2407–24.PubMed
67.
Zurück zum Zitat Core LJ, Waterfall JJ, Lis JT. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science. 2008;322:1845–8.PubMedPubMedCentral Core LJ, Waterfall JJ, Lis JT. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science. 2008;322:1845–8.PubMedPubMedCentral
68.
Zurück zum Zitat Herzog VA, Reichholf B, Neumann T, Rescheneder P, Bhat P, Burkard TR, et al. Thiol-linked alkylation of RNA to assess expression dynamics. Nat Methods. 2017;14:1198–204.PubMedPubMedCentral Herzog VA, Reichholf B, Neumann T, Rescheneder P, Bhat P, Burkard TR, et al. Thiol-linked alkylation of RNA to assess expression dynamics. Nat Methods. 2017;14:1198–204.PubMedPubMedCentral
69.
Zurück zum Zitat Schofield JA, Duffy EE, Kiefer L, Sullivan MC, Simon MD. TimeLapse-seq: adding a temporal dimension to RNA sequencing through nucleoside recoding. Nat Methods. 2018;15:221–5.PubMedPubMedCentral Schofield JA, Duffy EE, Kiefer L, Sullivan MC, Simon MD. TimeLapse-seq: adding a temporal dimension to RNA sequencing through nucleoside recoding. Nat Methods. 2018;15:221–5.PubMedPubMedCentral
70.
Zurück zum Zitat Chen Y, Wu F, Chen Z, He Z, Wei Q, Zeng W, et al. Acrylonitrile-mediated nascent RNA sequencing for transcriptome-wide profiling of cellular RNA dynamics. Adv Sci (Weinh). 2020;7:1900997.PubMedCentral Chen Y, Wu F, Chen Z, He Z, Wei Q, Zeng W, et al. Acrylonitrile-mediated nascent RNA sequencing for transcriptome-wide profiling of cellular RNA dynamics. Adv Sci (Weinh). 2020;7:1900997.PubMedCentral
71.
Zurück zum Zitat Li X, Zhou B, Chen L, Gou LT, Li H, Fu XD. GRID-seq reveals the global RNA-chromatin interactome. Nat Biotechnol. 2017;35:940–50.PubMedPubMedCentral Li X, Zhou B, Chen L, Gou LT, Li H, Fu XD. GRID-seq reveals the global RNA-chromatin interactome. Nat Biotechnol. 2017;35:940–50.PubMedPubMedCentral
72.
Zurück zum Zitat Yan Z, Huang N, Wu W, Chen W, Jiang Y, Chen J, et al. Genome-wide colocalization of RNA-DNA interactions and fusion RNA pairs. Proc Natl Acad Sci U S A. 2019;116:3328–37.PubMedPubMedCentral Yan Z, Huang N, Wu W, Chen W, Jiang Y, Chen J, et al. Genome-wide colocalization of RNA-DNA interactions and fusion RNA pairs. Proc Natl Acad Sci U S A. 2019;116:3328–37.PubMedPubMedCentral
73.
Zurück zum Zitat Bell JC, Jukam D, Teran NA, Risca VI, Smith OK, Johnson WL, et al. Chromatin-associated RNA sequencing (ChAR-seq) maps genome-wide RNA-to-DNA contacts. Elife. 2018;7:e27024.PubMedPubMedCentral Bell JC, Jukam D, Teran NA, Risca VI, Smith OK, Johnson WL, et al. Chromatin-associated RNA sequencing (ChAR-seq) maps genome-wide RNA-to-DNA contacts. Elife. 2018;7:e27024.PubMedPubMedCentral
74.
Zurück zum Zitat Kudla G, Granneman S, Hahn D, Beggs JD, Tollervey D. Cross-linking, ligation, and sequencing of hybrids reveals RNA-RNA interactions in yeast. Proc Natl Acad Sci U S A. 2011;108:10010–5.PubMedPubMedCentral Kudla G, Granneman S, Hahn D, Beggs JD, Tollervey D. Cross-linking, ligation, and sequencing of hybrids reveals RNA-RNA interactions in yeast. Proc Natl Acad Sci U S A. 2011;108:10010–5.PubMedPubMedCentral
75.
Zurück zum Zitat Metkar M, Ozadam H, Lajoie BR, Imakaev M, Mirny LA, Dekker J, et al. Higher-order organization principles of pre-translational mRNPs. Mol Cell. 2018;72:715–26.PubMedPubMedCentral Metkar M, Ozadam H, Lajoie BR, Imakaev M, Mirny LA, Dekker J, et al. Higher-order organization principles of pre-translational mRNPs. Mol Cell. 2018;72:715–26.PubMedPubMedCentral
76.
Zurück zum Zitat Nguyen TC, Cao X, Yu P, Xiao S, Lu J, Biase FH, et al. Mapping RNA-RNA interactome and RNA structure in vivo by MARIO. Nat Commun. 2016;7:12023.PubMedPubMedCentral Nguyen TC, Cao X, Yu P, Xiao S, Lu J, Biase FH, et al. Mapping RNA-RNA interactome and RNA structure in vivo by MARIO. Nat Commun. 2016;7:12023.PubMedPubMedCentral
77.
Zurück zum Zitat Lu Z, Zhang QC, Lee B, Flynn RA, Smith MA, Robinson JT, et al. RNA duplex map in living cells reveals higher-order transcriptome structure. Cell. 2016;165:1267–79.PubMedPubMedCentral Lu Z, Zhang QC, Lee B, Flynn RA, Smith MA, Robinson JT, et al. RNA duplex map in living cells reveals higher-order transcriptome structure. Cell. 2016;165:1267–79.PubMedPubMedCentral
78.
Zurück zum Zitat Sharma E, Sterne-Weiler T, O’Hanlon D, Blencowe BJ. Global mapping of human RNA-RNA interactions. Mol Cell. 2016;62:618–26.PubMed Sharma E, Sterne-Weiler T, O’Hanlon D, Blencowe BJ. Global mapping of human RNA-RNA interactions. Mol Cell. 2016;62:618–26.PubMed
79.
Zurück zum Zitat Aw JG, Shen Y, Wilm A, Sun M, Lim XN, Boon KL, et al. In vivo mapping of eukaryotic RNA interactomes reveals principles of higher-order organization and regulation. Mol Cell. 2016;62:603–17.PubMed Aw JG, Shen Y, Wilm A, Sun M, Lim XN, Boon KL, et al. In vivo mapping of eukaryotic RNA interactomes reveals principles of higher-order organization and regulation. Mol Cell. 2016;62:603–17.PubMed
80.
Zurück zum Zitat Cai Z, Cao C, Ji L, Ye R, Wang D, Xia C, et al. RIC-seq for global in situ profiling of RNA–RNA spatial interactions. Nature. 2020;582:432–7.PubMed Cai Z, Cao C, Ji L, Ye R, Wang D, Xia C, et al. RIC-seq for global in situ profiling of RNA–RNA spatial interactions. Nature. 2020;582:432–7.PubMed
81.
Zurück zum Zitat Morf J, Wingett SW, Farabella I, Cairns J, Furlan-Magaril M, Jimenez-Garcia LF, et al. RNA proximity sequencing reveals the spatial organization of the transcriptome in the nucleus. Nat Biotechnol. 2019;37:793–802.PubMed Morf J, Wingett SW, Farabella I, Cairns J, Furlan-Magaril M, Jimenez-Garcia LF, et al. RNA proximity sequencing reveals the spatial organization of the transcriptome in the nucleus. Nat Biotechnol. 2019;37:793–802.PubMed
82.
Zurück zum Zitat Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Ferrante TC, Terry R, et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat Protoc. 2015;10:442–58.PubMedPubMedCentral Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Ferrante TC, Terry R, et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat Protoc. 2015;10:442–58.PubMedPubMedCentral
84.
Zurück zum Zitat Kaewsapsak P, Shechner DM, Mallard W, Rinn JL, Ting AY. Live-cell mapping of organelle-associated RNAs via proximity biotinylation combined with protein-RNA crosslinking. Elife. 2017;6:e29224.PubMedPubMedCentral Kaewsapsak P, Shechner DM, Mallard W, Rinn JL, Ting AY. Live-cell mapping of organelle-associated RNAs via proximity biotinylation combined with protein-RNA crosslinking. Elife. 2017;6:e29224.PubMedPubMedCentral
85.
Zurück zum Zitat Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, et al. Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 2017;66:22–37.PubMedPubMedCentral Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, et al. Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 2017;66:22–37.PubMedPubMedCentral
86.
Zurück zum Zitat Diallo LH, Tatin F, David F, Godet AC, Zamora A, Prats AC, et al. How are circRNAs translated by non-canonical initiation mechanisms? Biochimie. 2019;164:45–52.PubMed Diallo LH, Tatin F, David F, Godet AC, Zamora A, Prats AC, et al. How are circRNAs translated by non-canonical initiation mechanisms? Biochimie. 2019;164:45–52.PubMed
87.
Zurück zum Zitat Jiao Y, Meyerowitz EM. Cell-type specific analysis of translating RNAs in developing flowers reveals new levels of control. Mol Syst Biol. 2010;6:419.PubMedPubMedCentral Jiao Y, Meyerowitz EM. Cell-type specific analysis of translating RNAs in developing flowers reveals new levels of control. Mol Syst Biol. 2010;6:419.PubMedPubMedCentral
88.
Zurück zum Zitat Ingolia NT, Lareau LF, Weissman JS. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell. 2011;147:789–802.PubMedPubMedCentral Ingolia NT, Lareau LF, Weissman JS. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell. 2011;147:789–802.PubMedPubMedCentral
89.
Zurück zum Zitat Sun YM, Wang WT, Zeng ZC, Chen TQ, Han C, Pan Q, et al. circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood. 2019;134:1533–46.PubMedPubMedCentral Sun YM, Wang WT, Zeng ZC, Chen TQ, Han C, Pan Q, et al. circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood. 2019;134:1533–46.PubMedPubMedCentral
90.
Zurück zum Zitat Struhl K. Transcriptional noise and the fidelity of initiation by RNA polymerase II. Nat Struct Mol Biol. 2007;14:103–5.PubMed Struhl K. Transcriptional noise and the fidelity of initiation by RNA polymerase II. Nat Struct Mol Biol. 2007;14:103–5.PubMed
91.
Zurück zum Zitat Giambruno R, Mihailovich M, Bonaldi T. Mass spectrometry-based proteomics to unveil the non-coding RNA world. Front Mol Biosci. 2018;5:90.PubMedPubMedCentral Giambruno R, Mihailovich M, Bonaldi T. Mass spectrometry-based proteomics to unveil the non-coding RNA world. Front Mol Biosci. 2018;5:90.PubMedPubMedCentral
92.
Zurück zum Zitat Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12:861–74.PubMed Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12:861–74.PubMed
93.
Zurück zum Zitat Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97.PubMed Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97.PubMed
94.
Zurück zum Zitat Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21-and 22-nucleotide RNAs. Genes Dev. 2001;15:188–200.PubMedPubMedCentral Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21-and 22-nucleotide RNAs. Genes Dev. 2001;15:188–200.PubMedPubMedCentral
95.
Zurück zum Zitat Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K, Hannon GJ. Developmentally regulated piRNA clusters implicate MILI in transposon control. Science. 2007;316:744–7.PubMed Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K, Hannon GJ. Developmentally regulated piRNA clusters implicate MILI in transposon control. Science. 2007;316:744–7.PubMed
96.
Zurück zum Zitat Taft RJ, Glazov EA, Cloonan N, Simons C, Stephen S, Faulkner GJ, et al. Tiny RNAs associated with transcription start sites in animals. Nat Genet. 2009;41:572–8.PubMed Taft RJ, Glazov EA, Cloonan N, Simons C, Stephen S, Faulkner GJ, et al. Tiny RNAs associated with transcription start sites in animals. Nat Genet. 2009;41:572–8.PubMed
98.
100.
Zurück zum Zitat Bachellerie JP, Cavaille J, Huttenhofer A. The expanding snoRNA world. Biochimie. 2002;84:775–90.PubMed Bachellerie JP, Cavaille J, Huttenhofer A. The expanding snoRNA world. Biochimie. 2002;84:775–90.PubMed
101.
Zurück zum Zitat Hamm J, Darzynkiewicz E, Tahara SM, Mattaj IW. The trimethylguanosine cap structure of U1 snRNA is a component of a bipartite nuclear targeting signal. Cell. 1990;62:569–77.PubMed Hamm J, Darzynkiewicz E, Tahara SM, Mattaj IW. The trimethylguanosine cap structure of U1 snRNA is a component of a bipartite nuclear targeting signal. Cell. 1990;62:569–77.PubMed
103.
Zurück zum Zitat Guo JU, Agarwal V, Guo H, Bartel DP. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15:409.PubMedPubMedCentral Guo JU, Agarwal V, Guo H, Bartel DP. Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 2014;15:409.PubMedPubMedCentral
104.
Zurück zum Zitat Salzman J, Chen RE, Olsen MN, Wang PL, Brown PO. Cell-type specific features of circular RNA expression. PLoS Genet. 2013;9:e1003777.PubMedPubMedCentral Salzman J, Chen RE, Olsen MN, Wang PL, Brown PO. Cell-type specific features of circular RNA expression. PLoS Genet. 2013;9:e1003777.PubMedPubMedCentral
105.
Zurück zum Zitat Xing YH, Yao RW, Zhang Y, Guo CJ, Jiang S, Xu G, et al. SLERT regulates DDX21 rings associated with pol I transcription. Cell. 2017;169:664–78.PubMed Xing YH, Yao RW, Zhang Y, Guo CJ, Jiang S, Xu G, et al. SLERT regulates DDX21 rings associated with pol I transcription. Cell. 2017;169:664–78.PubMed
106.
Zurück zum Zitat Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, et al. An atlas of active enhancers across human cell types and tissues. Nature. 2014;507:455–61.PubMedPubMedCentral Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, Boyd M, et al. An atlas of active enhancers across human cell types and tissues. Nature. 2014;507:455–61.PubMedPubMedCentral
107.
Zurück zum Zitat Selenko P, Sprangers R, Stier G, Buhler D, Fischer U, Sattler M. SMN tudor domain structure and its interaction with the Sm proteins. Nat Struct Biol. 2001;8:27–31.PubMed Selenko P, Sprangers R, Stier G, Buhler D, Fischer U, Sattler M. SMN tudor domain structure and its interaction with the Sm proteins. Nat Struct Biol. 2001;8:27–31.PubMed
108.
Zurück zum Zitat Dupuis-Sandoval F, Poirier M, Scott MS. The emerging landscape of small nucleolar RNAs in cell biology. Wiley Interdiscip Rev RNA. 2015;6:381–97.PubMedPubMedCentral Dupuis-Sandoval F, Poirier M, Scott MS. The emerging landscape of small nucleolar RNAs in cell biology. Wiley Interdiscip Rev RNA. 2015;6:381–97.PubMedPubMedCentral
109.
Zurück zum Zitat Hofacker IL, Fontana W, Stadler PF, Bonhoeffer LS, Tacker M, Schuster P. Fast folding and comparison of RNA secondary structures. Monatshefte für Chemie/Chemical Monthly. 1994;125:167–88. Hofacker IL, Fontana W, Stadler PF, Bonhoeffer LS, Tacker M, Schuster P. Fast folding and comparison of RNA secondary structures. Monatshefte für Chemie/Chemical Monthly. 1994;125:167–88.
110.
Zurück zum Zitat Xu L, Sun L, Guan G, Huang Q, Lv J, Yan L, et al. The effects of pH and salts on nucleic acid partitioning during phenol extraction. Nucleosides Nucleotides Nucleic Acids. 2019;38:305–20.PubMed Xu L, Sun L, Guan G, Huang Q, Lv J, Yan L, et al. The effects of pH and salts on nucleic acid partitioning during phenol extraction. Nucleosides Nucleotides Nucleic Acids. 2019;38:305–20.PubMed
111.
Zurück zum Zitat Nicosia A, Tagliavia M, Costa S. Regeneration of total RNA purification silica-based columns. Biomed Chromatogr. 2010;24:1263–4.PubMed Nicosia A, Tagliavia M, Costa S. Regeneration of total RNA purification silica-based columns. Biomed Chromatogr. 2010;24:1263–4.PubMed
112.
Zurück zum Zitat Fromm B, Harris PD, Bachmann L. MicroRNA preparations from individual monogenean Gyrodactylus salaris-a comparison of six commercially available totalRNA extraction kits. BMC Res Notes. 2011;4:217.PubMedPubMedCentral Fromm B, Harris PD, Bachmann L. MicroRNA preparations from individual monogenean Gyrodactylus salaris-a comparison of six commercially available totalRNA extraction kits. BMC Res Notes. 2011;4:217.PubMedPubMedCentral
113.
Zurück zum Zitat Chen EA, Souaiaia T, Herstein JS, Evgrafov OV, Spitsyna VN, Rebolini DF, et al. Effect of RNA integrity on uniquely mapped reads in RNA-Seq. BMC Res Notes. 2014;7:753.PubMedPubMedCentral Chen EA, Souaiaia T, Herstein JS, Evgrafov OV, Spitsyna VN, Rebolini DF, et al. Effect of RNA integrity on uniquely mapped reads in RNA-Seq. BMC Res Notes. 2014;7:753.PubMedPubMedCentral
114.
Zurück zum Zitat Veneziano D, Di Bella S, Nigita G, Lagana A, Ferro A, Croce CM. Noncoding RNA: current deep sequencing data analysis approaches and challenges. Hum Mutat. 2016;37:1283–98.PubMed Veneziano D, Di Bella S, Nigita G, Lagana A, Ferro A, Croce CM. Noncoding RNA: current deep sequencing data analysis approaches and challenges. Hum Mutat. 2016;37:1283–98.PubMed
115.
Zurück zum Zitat Modrek B, Lee C. A genomic view of alternative splicing. Nat Genet. 2002;30:13–9.PubMed Modrek B, Lee C. A genomic view of alternative splicing. Nat Genet. 2002;30:13–9.PubMed
116.
Zurück zum Zitat Zhang XO, Dong R, Zhang Y, Zhang JL, Luo Z, Zhang J, et al. Diverse alternative back-splicing and alternative splicing landscape of circular RNAs. Genome Res. 2016;26:1277–87.PubMedPubMedCentral Zhang XO, Dong R, Zhang Y, Zhang JL, Luo Z, Zhang J, et al. Diverse alternative back-splicing and alternative splicing landscape of circular RNAs. Genome Res. 2016;26:1277–87.PubMedPubMedCentral
117.
Zurück zum Zitat Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL, Yang L. Complementary sequence-mediated exon circularization. Cell. 2014;159:134–47.PubMed Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL, Yang L. Complementary sequence-mediated exon circularization. Cell. 2014;159:134–47.PubMed
118.
Zurück zum Zitat Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22:256–64.PubMed Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22:256–64.PubMed
119.
Zurück zum Zitat Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, et al. The landscape of circular RNA in cancer. Cell. 2019;176:869–81.PubMedPubMedCentral Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, et al. The landscape of circular RNA in cancer. Cell. 2019;176:869–81.PubMedPubMedCentral
120.
Zurück zum Zitat Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, et al. Oncogenic role of fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 2016;166:1055–6.PubMed Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, et al. Oncogenic role of fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 2016;166:1055–6.PubMed
121.
Zurück zum Zitat Filipowicz W, Jaskiewicz L, Kolb FA, Pillai RS. Post-transcriptional gene silencing by siRNAs and miRNAs. Curr Opin Struct Biol. 2005;15:331–41.PubMed Filipowicz W, Jaskiewicz L, Kolb FA, Pillai RS. Post-transcriptional gene silencing by siRNAs and miRNAs. Curr Opin Struct Biol. 2005;15:331–41.PubMed
122.
Zurück zum Zitat Seto AG, Kingston RE, Lau NC. The coming of age for Piwi proteins. Mol Cell. 2007;26:603–9.PubMed Seto AG, Kingston RE, Lau NC. The coming of age for Piwi proteins. Mol Cell. 2007;26:603–9.PubMed
123.
Zurück zum Zitat Kedersha NL, Rome LH. Isolation and characterization of a novel ribonucleoprotein particle: large structures contain a single species of small RNA. J Cell Biol. 1986;103:699–709.PubMed Kedersha NL, Rome LH. Isolation and characterization of a novel ribonucleoprotein particle: large structures contain a single species of small RNA. J Cell Biol. 1986;103:699–709.PubMed
124.
Zurück zum Zitat van Zon A, Mossink MH, Schoester M, Scheffer GL, Scheper RJ, Sonneveld P, et al. Multiple human vault RNAs. Expression and association with the vault complex. J Biol Chem. 2001;276:37715–21.PubMed van Zon A, Mossink MH, Schoester M, Scheffer GL, Scheper RJ, Sonneveld P, et al. Multiple human vault RNAs. Expression and association with the vault complex. J Biol Chem. 2001;276:37715–21.PubMed
125.
Zurück zum Zitat Kickhoefer VA, Searles RP, Kedersha NL, Garber ME, Johnson DL, Rome LH. Vault ribonucleoprotein particles from rat and bullfrog contain a related small RNA that is transcribed by RNA polymerase III. J Biol Chem. 1993;268:7868–73.PubMed Kickhoefer VA, Searles RP, Kedersha NL, Garber ME, Johnson DL, Rome LH. Vault ribonucleoprotein particles from rat and bullfrog contain a related small RNA that is transcribed by RNA polymerase III. J Biol Chem. 1993;268:7868–73.PubMed
126.
Zurück zum Zitat Kiss-Laszlo Z, Henry Y, Bachellerie JP, Caizergues-Ferrer M, Kiss T. Site-specific ribose methylation of preribosomal RNA: a novel function for small nucleolar RNAs. Cell. 1996;85:1077–88.PubMed Kiss-Laszlo Z, Henry Y, Bachellerie JP, Caizergues-Ferrer M, Kiss T. Site-specific ribose methylation of preribosomal RNA: a novel function for small nucleolar RNAs. Cell. 1996;85:1077–88.PubMed
127.
Zurück zum Zitat Mondal T, Rasmussen M, Pandey GK, Isaksson A, Kanduri C. Characterization of the RNA content of chromatin. Genome Res. 2010;20:899–907.PubMedPubMedCentral Mondal T, Rasmussen M, Pandey GK, Isaksson A, Kanduri C. Characterization of the RNA content of chromatin. Genome Res. 2010;20:899–907.PubMedPubMedCentral
128.
Zurück zum Zitat Schubert T, Pusch MC, Diermeier S, Benes V, Kremmer E, Imhof A, et al. Df31 protein and snoRNAs maintain accessible higher-order structures of chromatin. Mol Cell. 2012;48:434–44.PubMed Schubert T, Pusch MC, Diermeier S, Benes V, Kremmer E, Imhof A, et al. Df31 protein and snoRNAs maintain accessible higher-order structures of chromatin. Mol Cell. 2012;48:434–44.PubMed
129.
Zurück zum Zitat Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311–23.PubMedPubMedCentral Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311–23.PubMedPubMedCentral
130.
Zurück zum Zitat Yazaki J, Gregory BD, Ecker JR. Mapping the genome landscape using tiling array technology. Curr Opin Plant Biol. 2007;10:534–42.PubMedPubMedCentral Yazaki J, Gregory BD, Ecker JR. Mapping the genome landscape using tiling array technology. Curr Opin Plant Biol. 2007;10:534–42.PubMedPubMedCentral
131.
Zurück zum Zitat Quinn EM, Cormican P, Kenny EM, Hill M, Anney R, Gill M, et al. Development of strategies for SNP detection in RNA-seq data: application to lymphoblastoid cell lines and evaluation using 1000 genomes data. PLoS One. 2013;8:e58815.PubMedPubMedCentral Quinn EM, Cormican P, Kenny EM, Hill M, Anney R, Gill M, et al. Development of strategies for SNP detection in RNA-seq data: application to lymphoblastoid cell lines and evaluation using 1000 genomes data. PLoS One. 2013;8:e58815.PubMedPubMedCentral
132.
Zurück zum Zitat Sultan M, Schulz MH, Richard H, Magen A, Klingenhoff A, Scherf M, et al. A global view of gene activity and alternative splicing by deep sequencing of the human transcriptome. Science. 2008;321:956–60.PubMed Sultan M, Schulz MH, Richard H, Magen A, Klingenhoff A, Scherf M, et al. A global view of gene activity and alternative splicing by deep sequencing of the human transcriptome. Science. 2008;321:956–60.PubMed
133.
Zurück zum Zitat Edgren H, Murumagi A, Kangaspeska S, Nicorici D, Hongisto V, Kleivi K, et al. Identification of fusion genes in breast cancer by paired-end RNA-sequencing. Genome Biol. 2011;12:R6.PubMedPubMedCentral Edgren H, Murumagi A, Kangaspeska S, Nicorici D, Hongisto V, Kleivi K, et al. Identification of fusion genes in breast cancer by paired-end RNA-sequencing. Genome Biol. 2011;12:R6.PubMedPubMedCentral
134.
Zurück zum Zitat Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105–11.PubMedPubMedCentral Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105–11.PubMedPubMedCentral
135.
Zurück zum Zitat Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–27.PubMedPubMedCentral Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–27.PubMedPubMedCentral
136.
Zurück zum Zitat Muller S, Rycak L, Winter P, Kahl G, Koch I, Rotter B. omiRas: a web server for differential expression analysis of miRNAs derived from small RNA-Seq data. Bioinformatics. 2013;29:2651–2.PubMed Muller S, Rycak L, Winter P, Kahl G, Koch I, Rotter B. omiRas: a web server for differential expression analysis of miRNAs derived from small RNA-Seq data. Bioinformatics. 2013;29:2651–2.PubMed
137.
Zurück zum Zitat Han BW, Wang W, Zamore PD, Weng Z. piPipes: a set of pipelines for piRNA and transposon analysis via small RNA-seq, RNA-seq, degradome- and CAGE-seq, ChIP-seq and genomic DNA sequencing. Bioinformatics. 2015;31:593–5.PubMed Han BW, Wang W, Zamore PD, Weng Z. piPipes: a set of pipelines for piRNA and transposon analysis via small RNA-seq, RNA-seq, degradome- and CAGE-seq, ChIP-seq and genomic DNA sequencing. Bioinformatics. 2015;31:593–5.PubMed
138.
Zurück zum Zitat Hoogstrate Y, Jenster G, Martens-Uzunova ES. FlaiMapper: computational annotation of small ncRNA-derived fragments using RNA-seq high-throughput data. Bioinformatics. 2015;31:665–73.PubMed Hoogstrate Y, Jenster G, Martens-Uzunova ES. FlaiMapper: computational annotation of small ncRNA-derived fragments using RNA-seq high-throughput data. Bioinformatics. 2015;31:665–73.PubMed
139.
140.
Zurück zum Zitat Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentral Conesa A, Madrigal P, Tarazona S, Gomez-Cabrero D, Cervera A, McPherson A, et al. A survey of best practices for RNA-seq data analysis. Genome Biol. 2016;17:13.PubMedPubMedCentral
141.
Zurück zum Zitat Baker M. MicroRNA profiling: separating signal from noise. Nat Methods. 2010;7:687–92.PubMed Baker M. MicroRNA profiling: separating signal from noise. Nat Methods. 2010;7:687–92.PubMed
142.
Zurück zum Zitat Langenberger D, Bermudez-Santana C, Hertel J, Hoffmann S, Khaitovich P, Stadler PF. Evidence for human microRNA-offset RNAs in small RNA sequencing data. Bioinformatics. 2009;25:2298–301.PubMed Langenberger D, Bermudez-Santana C, Hertel J, Hoffmann S, Khaitovich P, Stadler PF. Evidence for human microRNA-offset RNAs in small RNA sequencing data. Bioinformatics. 2009;25:2298–301.PubMed
143.
Zurück zum Zitat Li Z, Ender C, Meister G, Moore PS, Chang Y, John B. Extensive terminal and asymmetric processing of small RNAs from rRNAs, snoRNAs, snRNAs, and tRNAs. Nucleic Acids Res. 2012;40:6787–99.PubMedPubMedCentral Li Z, Ender C, Meister G, Moore PS, Chang Y, John B. Extensive terminal and asymmetric processing of small RNAs from rRNAs, snoRNAs, snRNAs, and tRNAs. Nucleic Acids Res. 2012;40:6787–99.PubMedPubMedCentral
144.
Zurück zum Zitat Giraldez MD, Spengler RM, Etheridge A, Godoy PM, Barczak AJ, Srinivasan S, et al. Comprehensive multi-center assessment of small RNA-seq methods for quantitative miRNA profiling. Nat Biotechnol. 2018;36:746–57.PubMedPubMedCentral Giraldez MD, Spengler RM, Etheridge A, Godoy PM, Barczak AJ, Srinivasan S, et al. Comprehensive multi-center assessment of small RNA-seq methods for quantitative miRNA profiling. Nat Biotechnol. 2018;36:746–57.PubMedPubMedCentral
145.
Zurück zum Zitat Wright C, Rajpurohit A, Burke EE, Williams C, Collado-Torres L, Kimos M, et al. Comprehensive assessment of multiple biases in small RNA sequencing reveals significant differences in the performance of widely used methods. BMC Genomics. 2019;20:513.PubMedPubMedCentral Wright C, Rajpurohit A, Burke EE, Williams C, Collado-Torres L, Kimos M, et al. Comprehensive assessment of multiple biases in small RNA sequencing reveals significant differences in the performance of widely used methods. BMC Genomics. 2019;20:513.PubMedPubMedCentral
146.
Zurück zum Zitat Potter SS. Single-cell RNA sequencing for the study of development, physiology and disease. Nat Rev Nephrol. 2018;14:479–92.PubMedPubMedCentral Potter SS. Single-cell RNA sequencing for the study of development, physiology and disease. Nat Rev Nephrol. 2018;14:479–92.PubMedPubMedCentral
147.
Zurück zum Zitat Warren L, Bryder D, Weissman IL, Quake SR. Transcription factor profiling in individual hematopoietic progenitors by digital RT-PCR. Proc Natl Acad Sci U S A. 2006;103:17807–12.PubMedPubMedCentral Warren L, Bryder D, Weissman IL, Quake SR. Transcription factor profiling in individual hematopoietic progenitors by digital RT-PCR. Proc Natl Acad Sci U S A. 2006;103:17807–12.PubMedPubMedCentral
148.
Zurück zum Zitat Serra L, Chang DZ, Macchietto M, Williams K, Murad R, Lu D, et al. Adapting the smart-seq2 protocol for robust single worm RNA-seq. Bio Protoc. 2018;8:e2729.PubMedPubMedCentral Serra L, Chang DZ, Macchietto M, Williams K, Murad R, Lu D, et al. Adapting the smart-seq2 protocol for robust single worm RNA-seq. Bio Protoc. 2018;8:e2729.PubMedPubMedCentral
149.
Zurück zum Zitat Bhargava V, Head SR, Ordoukhanian P, Mercola M, Subramaniam S. Technical variations in low-input RNA-seq methodologies. Sci Rep. 2014;4:3678.PubMedPubMedCentral Bhargava V, Head SR, Ordoukhanian P, Mercola M, Subramaniam S. Technical variations in low-input RNA-seq methodologies. Sci Rep. 2014;4:3678.PubMedPubMedCentral
150.
Zurück zum Zitat Friedel CC, Dolken L. Metabolic tagging and purification of nascent RNA: implications for transcriptomics. Mol BioSyst. 2009;5:1271–8.PubMed Friedel CC, Dolken L. Metabolic tagging and purification of nascent RNA: implications for transcriptomics. Mol BioSyst. 2009;5:1271–8.PubMed
151.
Zurück zum Zitat Wissink EM, Vihervaara A, Tippens ND, Lis JT. Nascent RNA analyses: tracking transcription and its regulation. Nat Rev Genet. 2019;20:705–23.PubMedPubMedCentral Wissink EM, Vihervaara A, Tippens ND, Lis JT. Nascent RNA analyses: tracking transcription and its regulation. Nat Rev Genet. 2019;20:705–23.PubMedPubMedCentral
152.
Zurück zum Zitat Min IM, Waterfall JJ, Core LJ, Munroe RJ, Schimenti J, Lis JT. Regulating RNA polymerase pausing and transcription elongation in embryonic stem cells. Genes Dev. 2011;25:742–54.PubMedPubMedCentral Min IM, Waterfall JJ, Core LJ, Munroe RJ, Schimenti J, Lis JT. Regulating RNA polymerase pausing and transcription elongation in embryonic stem cells. Genes Dev. 2011;25:742–54.PubMedPubMedCentral
153.
Zurück zum Zitat Baptista MAP, Dolken L. RNA dynamics revealed by metabolic RNA labeling and biochemical nucleoside conversions. Nat Methods. 2018;15:171–2.PubMed Baptista MAP, Dolken L. RNA dynamics revealed by metabolic RNA labeling and biochemical nucleoside conversions. Nat Methods. 2018;15:171–2.PubMed
154.
Zurück zum Zitat Washietl S, Hofacker IL, Lukasser M, Huttenhofer A, Stadler PF. Mapping of conserved RNA secondary structures predicts thousands of functional noncoding RNAs in the human genome. Nat Biotechnol. 2005;23:1383–90.PubMed Washietl S, Hofacker IL, Lukasser M, Huttenhofer A, Stadler PF. Mapping of conserved RNA secondary structures predicts thousands of functional noncoding RNAs in the human genome. Nat Biotechnol. 2005;23:1383–90.PubMed
155.
Zurück zum Zitat Fabbri M, Girnita L, Varani G, Calin GA. Decrypting noncoding RNA interactions, structures, and functional networks. Genome Res. 2019;29:1377–88.PubMedPubMedCentral Fabbri M, Girnita L, Varani G, Calin GA. Decrypting noncoding RNA interactions, structures, and functional networks. Genome Res. 2019;29:1377–88.PubMedPubMedCentral
156.
Zurück zum Zitat Fu XD. Non-coding RNA: a new frontier in regulatory biology. Natl Sci Rev. 2014;1:190–204.PubMed Fu XD. Non-coding RNA: a new frontier in regulatory biology. Natl Sci Rev. 2014;1:190–204.PubMed
157.
Zurück zum Zitat Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81:145–66.PubMed Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81:145–66.PubMed
158.
Zurück zum Zitat West JA, Davis CP, Sunwoo H, Simon MD, Sadreyev RI, Wang PI, et al. The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol Cell. 2014;55:791–802.PubMedPubMedCentral West JA, Davis CP, Sunwoo H, Simon MD, Sadreyev RI, Wang PI, et al. The long noncoding RNAs NEAT1 and MALAT1 bind active chromatin sites. Mol Cell. 2014;55:791–802.PubMedPubMedCentral
159.
Zurück zum Zitat Sridhar B, Rivas-Astroza M, Nguyen TC, Chen W, Yan Z, Cao X, et al. Systematic mapping of RNA-chromatin interactions in vivo. Curr Biol. 2017;27:610–2.PubMed Sridhar B, Rivas-Astroza M, Nguyen TC, Chen W, Yan Z, Cao X, et al. Systematic mapping of RNA-chromatin interactions in vivo. Curr Biol. 2017;27:610–2.PubMed
160.
Zurück zum Zitat Simon MD, Wang CI, Kharchenko PV, West JA, Chapman BA, Alekseyenko AA, et al. The genomic binding sites of a noncoding RNA. Proc Natl Acad Sci U S A. 2011;108:20497–502.PubMedPubMedCentral Simon MD, Wang CI, Kharchenko PV, West JA, Chapman BA, Alekseyenko AA, et al. The genomic binding sites of a noncoding RNA. Proc Natl Acad Sci U S A. 2011;108:20497–502.PubMedPubMedCentral
161.
Zurück zum Zitat Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol Cell. 2011;44:667–78.PubMedPubMedCentral Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol Cell. 2011;44:667–78.PubMedPubMedCentral
162.
Zurück zum Zitat Engreitz JM, Pandya-Jones A, McDonel P, Shishkin A, Sirokman K, Surka C, et al. The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science. 2013;341:1237973.PubMedPubMedCentral Engreitz JM, Pandya-Jones A, McDonel P, Shishkin A, Sirokman K, Surka C, et al. The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science. 2013;341:1237973.PubMedPubMedCentral
163.
Zurück zum Zitat Kudla G, Wan Y, Helwak A. RNA conformation capture by proximity ligation. Annu Rev Genomics Hum Genet. 2020;21(1):14. Kudla G, Wan Y, Helwak A. RNA conformation capture by proximity ligation. Annu Rev Genomics Hum Genet. 2020;21(1):14.
164.
Zurück zum Zitat Helwak A, Kudla G, Dudnakova T, Tollervey D. Mapping the human miRNA interactome by CLASH reveals frequent noncanonical binding. Cell. 2013;153:654–65.PubMedPubMedCentral Helwak A, Kudla G, Dudnakova T, Tollervey D. Mapping the human miRNA interactome by CLASH reveals frequent noncanonical binding. Cell. 2013;153:654–65.PubMedPubMedCentral
165.
Zurück zum Zitat Hearst JE. Psoralen photochemistry and nucleic acid structure. J Invest Dermatol. 1981;77:39–44.PubMed Hearst JE. Psoralen photochemistry and nucleic acid structure. J Invest Dermatol. 1981;77:39–44.PubMed
166.
Zurück zum Zitat Huber RG, Lim XN, Ng WC, Sim AYL, Poh HX, Shen Y, et al. Structure mapping of dengue and Zika viruses reveals functional long-range interactions. Nat Commun. 2019;10:1408.PubMedPubMedCentral Huber RG, Lim XN, Ng WC, Sim AYL, Poh HX, Shen Y, et al. Structure mapping of dengue and Zika viruses reveals functional long-range interactions. Nat Commun. 2019;10:1408.PubMedPubMedCentral
167.
Zurück zum Zitat Li P, Wei Y, Mei M, Tang L, Sun L, Huang W, et al. Integrative analysis of Zika virus genome RNA structure reveals critical determinants of viral infectivity. Cell Host Microbe. 2018;24:875–86.PubMed Li P, Wei Y, Mei M, Tang L, Sun L, Huang W, et al. Integrative analysis of Zika virus genome RNA structure reveals critical determinants of viral infectivity. Cell Host Microbe. 2018;24:875–86.PubMed
168.
Zurück zum Zitat Sahu A, Singhal U, Chinnaiyan AM. Long noncoding RNAs in cancer: from function to translation. Trends Cancer. 2015;1:93–109.PubMedPubMedCentral Sahu A, Singhal U, Chinnaiyan AM. Long noncoding RNAs in cancer: from function to translation. Trends Cancer. 2015;1:93–109.PubMedPubMedCentral
169.
Zurück zum Zitat Cabili MN, Dunagin MC, McClanahan PD, Biaesch A, Padovan-Merhar O, Regev A, et al. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 2015;16:20.PubMedPubMedCentral Cabili MN, Dunagin MC, McClanahan PD, Biaesch A, Padovan-Merhar O, Regev A, et al. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 2015;16:20.PubMedPubMedCentral
170.
Zurück zum Zitat Hougaard DM, Hansen H, Larsson LI. Non-radioactive in situ hybridization for mRNA with emphasis on the use of oligodeoxynucleotide probes. Histochem Cell Biol. 1997;108:335–44.PubMed Hougaard DM, Hansen H, Larsson LI. Non-radioactive in situ hybridization for mRNA with emphasis on the use of oligodeoxynucleotide probes. Histochem Cell Biol. 1997;108:335–44.PubMed
171.
Zurück zum Zitat Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat Methods. 2008;5:877–9.PubMedPubMedCentral Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat Methods. 2008;5:877–9.PubMedPubMedCentral
172.
173.
Zurück zum Zitat Lee YH, Tan HT, Chung MC. Subcellular fractionation methods and strategies for proteomics. Proteomics. 2010;10:3935–56.PubMed Lee YH, Tan HT, Chung MC. Subcellular fractionation methods and strategies for proteomics. Proteomics. 2010;10:3935–56.PubMed
174.
Zurück zum Zitat Sultan M, Amstislavskiy V, Risch T, Schuette M, Dokel S, Ralser M, et al. Influence of RNA extraction methods and library selection schemes on RNA-seq data. BMC Genomics. 2014;15:675.PubMedPubMedCentral Sultan M, Amstislavskiy V, Risch T, Schuette M, Dokel S, Ralser M, et al. Influence of RNA extraction methods and library selection schemes on RNA-seq data. BMC Genomics. 2014;15:675.PubMedPubMedCentral
176.
Zurück zum Zitat Rhee HW, Zou P, Udeshi ND, Martell JD, Mootha VK, Carr SA, et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science. 2013;339:1328–31.PubMedPubMedCentral Rhee HW, Zou P, Udeshi ND, Martell JD, Mootha VK, Carr SA, et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science. 2013;339:1328–31.PubMedPubMedCentral
177.
Zurück zum Zitat Gilbert C, Kristjuhan A, Winkler GS, Svejstrup JQ. Elongator interactions with nascent mRNA revealed by RNA immunoprecipitation. Mol Cell. 2004;14:457–64.PubMed Gilbert C, Kristjuhan A, Winkler GS, Svejstrup JQ. Elongator interactions with nascent mRNA revealed by RNA immunoprecipitation. Mol Cell. 2004;14:457–64.PubMed
178.
Zurück zum Zitat Fazal FM, Han S, Parker KR, Kaewsapsak P, Xu J, Boettiger AN, et al. Atlas of subcellular RNA localization revealed by APEX-Seq. Cell. 2019;178:473–90.PubMedPubMedCentral Fazal FM, Han S, Parker KR, Kaewsapsak P, Xu J, Boettiger AN, et al. Atlas of subcellular RNA localization revealed by APEX-Seq. Cell. 2019;178:473–90.PubMedPubMedCentral
179.
Zurück zum Zitat Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, et al. TANRIC: an interactive open platform to explore the function of lncRNAs in cancer. Cancer Res. 2015;75:3728–37.PubMedPubMedCentral Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, et al. TANRIC: an interactive open platform to explore the function of lncRNAs in cancer. Cancer Res. 2015;75:3728–37.PubMedPubMedCentral
180.
Zurück zum Zitat Gao Y, Wang P, Wang Y, Ma X, Zhi H, Zhou D, et al. Lnc2Cancer v2.0: updated database of experimentally supported long non-coding RNAs in human cancers. Nucleic Acids Res. 2019;47:D1028–33.PubMed Gao Y, Wang P, Wang Y, Ma X, Zhi H, Zhou D, et al. Lnc2Cancer v2.0: updated database of experimentally supported long non-coding RNAs in human cancers. Nucleic Acids Res. 2019;47:D1028–33.PubMed
181.
Zurück zum Zitat Liu Y, Zhao M. lnCaNet: pan-cancer co-expression network for human lncRNA and cancer genes. Bioinformatics. 2016;32:1595–7.PubMed Liu Y, Zhao M. lnCaNet: pan-cancer co-expression network for human lncRNA and cancer genes. Bioinformatics. 2016;32:1595–7.PubMed
182.
Zurück zum Zitat Bao Z, Yang Z, Huang Z, Zhou Y, Cui Q, Dong D. LncRNADisease 2.0: an updated database of long non-coding RNA-associated diseases. Nucleic Acids Res. 2019;47:D1034–7.PubMed Bao Z, Yang Z, Huang Z, Zhou Y, Cui Q, Dong D. LncRNADisease 2.0: an updated database of long non-coding RNA-associated diseases. Nucleic Acids Res. 2019;47:D1034–7.PubMed
183.
Zurück zum Zitat Xia S, Feng J, Chen K, Ma Y, Gong J, Cai F, et al. CSCD: a database for cancer-specific circular RNAs. Nucleic Acids Res. 2018;46:D925–9.PubMed Xia S, Feng J, Chen K, Ma Y, Gong J, Cai F, et al. CSCD: a database for cancer-specific circular RNAs. Nucleic Acids Res. 2018;46:D925–9.PubMed
184.
Zurück zum Zitat Ghosal S, Das S, Sen R, Basak P, Chakrabarti J. Circ2Traits: a comprehensive database for circular RNA potentially associated with disease and traits. Front Genet. 2013;4:283.PubMedPubMedCentral Ghosal S, Das S, Sen R, Basak P, Chakrabarti J. Circ2Traits: a comprehensive database for circular RNA potentially associated with disease and traits. Front Genet. 2013;4:283.PubMedPubMedCentral
186.
Zurück zum Zitat Xie B, Ding Q, Han H, Wu D. miRCancer: a microRNA-cancer association database constructed by text mining on literature. Bioinformatics. 2013;29:638–44.PubMed Xie B, Ding Q, Han H, Wu D. miRCancer: a microRNA-cancer association database constructed by text mining on literature. Bioinformatics. 2013;29:638–44.PubMed
187.
Zurück zum Zitat Bhattacharya A, Cui Y. SomamiR 2.0: a database of cancer somatic mutations altering microRNA-ceRNA interactions. Nucleic Acids Res. 2016;44:D1005–10.PubMed Bhattacharya A, Cui Y. SomamiR 2.0: a database of cancer somatic mutations altering microRNA-ceRNA interactions. Nucleic Acids Res. 2016;44:D1005–10.PubMed
188.
Zurück zum Zitat Wong NW, Chen Y, Chen S, Wang X. OncomiR: an online resource for exploring pan-cancer microRNA dysregulation. Bioinformatics. 2018;34:713–5.PubMed Wong NW, Chen Y, Chen S, Wang X. OncomiR: an online resource for exploring pan-cancer microRNA dysregulation. Bioinformatics. 2018;34:713–5.PubMed
189.
Zurück zum Zitat Ahmed M, Nguyen H, Lai T, Kim DR. miRCancerdb: a database for correlation analysis between microRNA and gene expression in cancer. BMC Res Notes. 2018;11:103.PubMedPubMedCentral Ahmed M, Nguyen H, Lai T, Kim DR. miRCancerdb: a database for correlation analysis between microRNA and gene expression in cancer. BMC Res Notes. 2018;11:103.PubMedPubMedCentral
190.
Zurück zum Zitat Yang Z, Wu L, Wang A, Tang W, Zhao Y, Zhao H, et al. dbDEMC 2.0: updated database of differentially expressed miRNAs in human cancers. Nucleic Acids Res. 2017;45:D812–8.PubMed Yang Z, Wu L, Wang A, Tang W, Zhao Y, Zhao H, et al. dbDEMC 2.0: updated database of differentially expressed miRNAs in human cancers. Nucleic Acids Res. 2017;45:D812–8.PubMed
191.
Zurück zum Zitat Chung IF, Chang SJ, Chen CY, Liu SH, Li CY, Chan CH, et al. YM500v3: a database for small RNA sequencing in human cancer research. Nucleic Acids Res. 2017;45:D925–31.PubMed Chung IF, Chang SJ, Chen CY, Liu SH, Li CY, Chan CH, et al. YM500v3: a database for small RNA sequencing in human cancer research. Nucleic Acids Res. 2017;45:D925–31.PubMed
192.
Zurück zum Zitat Zheng LL, Xu WL, Liu S, Sun WJ, Li JH, Wu J, et al. tRF2Cancer: a web server to detect tRNA-derived small RNA fragments (tRFs) and their expression in multiple cancers. Nucleic Acids Res. 2016;44:W185–93.PubMedPubMedCentral Zheng LL, Xu WL, Liu S, Sun WJ, Li JH, Wu J, et al. tRF2Cancer: a web server to detect tRNA-derived small RNA fragments (tRFs) and their expression in multiple cancers. Nucleic Acids Res. 2016;44:W185–93.PubMedPubMedCentral
193.
Zurück zum Zitat Pliatsika V, Loher P, Magee R, Telonis AG, Londin E, Shigematsu M, et al. MINTbase v2.0: a comprehensive database for tRNA-derived fragments that includes nuclear and mitochondrial fragments from all the cancer genome atlas projects. Nucleic Acids Res. 2018;46:D152–9.PubMed Pliatsika V, Loher P, Magee R, Telonis AG, Londin E, Shigematsu M, et al. MINTbase v2.0: a comprehensive database for tRNA-derived fragments that includes nuclear and mitochondrial fragments from all the cancer genome atlas projects. Nucleic Acids Res. 2018;46:D152–9.PubMed
194.
Zurück zum Zitat Yan X, Hu Z, Feng Y, Hu X, Yuan J, Zhao SD, et al. Comprehensive genomic characterization of long non-coding RNAs across human cancers. Cancer Cell. 2015;28:529–40.PubMedPubMedCentral Yan X, Hu Z, Feng Y, Hu X, Yuan J, Zhao SD, et al. Comprehensive genomic characterization of long non-coding RNAs across human cancers. Cancer Cell. 2015;28:529–40.PubMedPubMedCentral
196.
Zurück zum Zitat Zhao Z, Wang K, Wu F, Wang W, Zhang K, Hu H, et al. circRNA disease: a manually curated database of experimentally supported circRNA-disease associations. Cell Death Dis. 2018;9:475.PubMedPubMedCentral Zhao Z, Wang K, Wu F, Wang W, Zhang K, Hu H, et al. circRNA disease: a manually curated database of experimentally supported circRNA-disease associations. Cell Death Dis. 2018;9:475.PubMedPubMedCentral
197.
Zurück zum Zitat Ruan H, Xiang Y, Ko J, Li S, Jing Y, Zhu X, et al. Comprehensive characterization of circular RNAs in ~1000 human cancer cell lines. Genome Med. 2019;11:55.PubMedPubMedCentral Ruan H, Xiang Y, Ko J, Li S, Jing Y, Zhu X, et al. Comprehensive characterization of circular RNAs in ~1000 human cancer cell lines. Genome Med. 2019;11:55.PubMedPubMedCentral
198.
Zurück zum Zitat Jiang Q, Wang Y, Hao Y, Juan L, Teng M, Zhang X, et al. miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 2009;37:D98–104.PubMed Jiang Q, Wang Y, Hao Y, Juan L, Teng M, Zhang X, et al. miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 2009;37:D98–104.PubMed
199.
Zurück zum Zitat Volders PJ, Anckaert J, Verheggen K, Nuytens J, Martens L, Mestdagh P, et al. LNCipedia 5: towards a reference set of human long non-coding RNAs. Nucleic Acids Res. 2019;47:D135–9.PubMed Volders PJ, Anckaert J, Verheggen K, Nuytens J, Martens L, Mestdagh P, et al. LNCipedia 5: towards a reference set of human long non-coding RNAs. Nucleic Acids Res. 2019;47:D135–9.PubMed
200.
Zurück zum Zitat Gong J, Liu C, Liu W, Xiang Y, Diao L, Guo AY, et al. LNCediting: a database for functional effects of RNA editing in lncRNAs. Nucleic Acids Res. 2017;45:D79–84.PubMed Gong J, Liu C, Liu W, Xiang Y, Diao L, Guo AY, et al. LNCediting: a database for functional effects of RNA editing in lncRNAs. Nucleic Acids Res. 2017;45:D79–84.PubMed
201.
Zurück zum Zitat Quek XC, Thomson DW, Maag JL, Bartonicek N, Signal B, Clark MB, et al. lncRNAdb v2.0: expanding the reference database for functional long noncoding RNAs. Nucleic Acids Res. 2015;43:D168–73.PubMed Quek XC, Thomson DW, Maag JL, Bartonicek N, Signal B, Clark MB, et al. lncRNAdb v2.0: expanding the reference database for functional long noncoding RNAs. Nucleic Acids Res. 2015;43:D168–73.PubMed
202.
Zurück zum Zitat Ma L, Li A, Zou D, Xu X, Xia L, Yu J, et al. LncRNAWiki: harnessing community knowledge in collaborative curation of human long non-coding RNAs. Nucleic Acids Res. 2015;43:D187–92.PubMed Ma L, Li A, Zou D, Xu X, Xia L, Yu J, et al. LncRNAWiki: harnessing community knowledge in collaborative curation of human long non-coding RNAs. Nucleic Acids Res. 2015;43:D187–92.PubMed
203.
Zurück zum Zitat Ma L, Cao J, Liu L, Du Q, Li Z, Zou D, et al. LncBook: a curated knowledgebase of human long non-coding RNAs. Nucleic Acids Res. 2019;47:D128–34.PubMed Ma L, Cao J, Liu L, Du Q, Li Z, Zou D, et al. LncBook: a curated knowledgebase of human long non-coding RNAs. Nucleic Acids Res. 2019;47:D128–34.PubMed
204.
Zurück zum Zitat Josset L, Tchitchek N, Gralinski LE, Ferris MT, Eisfeld AJ, Green RR, et al. Annotation of long non-coding RNAs expressed in collaborative cross founder mice in response to respiratory virus infection reveals a new class of interferon-stimulated transcripts. RNA Biol. 2014;11:875–90.PubMedPubMedCentral Josset L, Tchitchek N, Gralinski LE, Ferris MT, Eisfeld AJ, Green RR, et al. Annotation of long non-coding RNAs expressed in collaborative cross founder mice in response to respiratory virus infection reveals a new class of interferon-stimulated transcripts. RNA Biol. 2014;11:875–90.PubMedPubMedCentral
205.
Zurück zum Zitat Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, et al. NONCODE 2016: an informative and valuable data source of long non-coding RNAs. Nucleic Acids Res. 2016;44:D203–8.PubMed Zhao Y, Li H, Fang S, Kang Y, Wu W, Hao Y, et al. NONCODE 2016: an informative and valuable data source of long non-coding RNAs. Nucleic Acids Res. 2016;44:D203–8.PubMed
206.
Zurück zum Zitat Wu W, Ji P, Zhao F. CircAtlas: an integrated resource of one million highly accurate circular RNAs from 1070 vertebrate transcriptomes. Genome Biol. 2020;21:101.PubMedPubMedCentral Wu W, Ji P, Zhao F. CircAtlas: an integrated resource of one million highly accurate circular RNAs from 1070 vertebrate transcriptomes. Genome Biol. 2020;21:101.PubMedPubMedCentral
207.
208.
Zurück zum Zitat Dong R, Ma XK, Li GW, Yang L. CIRCpedia v2: an updated database for comprehensive circular RNA annotation and expression comparison. Genomics Proteomics Bioinformatics. 2018;16:226–33.PubMedPubMedCentral Dong R, Ma XK, Li GW, Yang L. CIRCpedia v2: an updated database for comprehensive circular RNA annotation and expression comparison. Genomics Proteomics Bioinformatics. 2018;16:226–33.PubMedPubMedCentral
209.
Zurück zum Zitat Xia S, Feng J, Lei L, Hu J, Xia L, Wang J, et al. Comprehensive characterization of tissue-specific circular RNAs in the human and mouse genomes. Brief Bioinform. 2017;18:984–92.PubMed Xia S, Feng J, Lei L, Hu J, Xia L, Wang J, et al. Comprehensive characterization of tissue-specific circular RNAs in the human and mouse genomes. Brief Bioinform. 2017;18:984–92.PubMed
210.
Zurück zum Zitat Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42:D92–7.PubMed Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42:D92–7.PubMed
211.
Zurück zum Zitat Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, et al. miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018;46:D296–302.PubMed Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, et al. miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018;46:D296–302.PubMed
212.
Zurück zum Zitat Panwar B, Omenn GS, Guan Y. miRmine: a database of human miRNA expression profiles. Bioinformatics. 2017;33:1554–60.PubMedPubMedCentral Panwar B, Omenn GS, Guan Y. miRmine: a database of human miRNA expression profiles. Bioinformatics. 2017;33:1554–60.PubMedPubMedCentral
213.
Zurück zum Zitat Liu T, Zhang Q, Zhang J, Li C, Miao YR, Lei Q, et al. EVmiRNA: a database of miRNA profiling in extracellular vesicles. Nucleic Acids Res. 2019;47:D89–93.PubMed Liu T, Zhang Q, Zhang J, Li C, Miao YR, Lei Q, et al. EVmiRNA: a database of miRNA profiling in extracellular vesicles. Nucleic Acids Res. 2019;47:D89–93.PubMed
215.
Zurück zum Zitat Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2019;47:D155–62.PubMed Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2019;47:D155–62.PubMed
216.
Zurück zum Zitat Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018;46:D239–45.PubMed Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018;46:D239–45.PubMed
217.
Zurück zum Zitat Kuksa PP, Amlie-Wolf A, Katanic Z, Valladares O, Wang LS, Leung YY. DASHR 2.0: integrated database of human small non-coding RNA genes and mature products. Bioinformatics. 2019;35:1033–9.PubMed Kuksa PP, Amlie-Wolf A, Katanic Z, Valladares O, Wang LS, Leung YY. DASHR 2.0: integrated database of human small non-coding RNA genes and mature products. Bioinformatics. 2019;35:1033–9.PubMed
218.
Zurück zum Zitat Zhang T, Tan P, Wang L, Jin N, Li Y, Zhang L, et al. RNALocate: a resource for RNA subcellular localizations. Nucleic Acids Res. 2017;45:D135–8.PubMed Zhang T, Tan P, Wang L, Jin N, Li Y, Zhang L, et al. RNALocate: a resource for RNA subcellular localizations. Nucleic Acids Res. 2017;45:D135–8.PubMed
219.
Zurück zum Zitat Mas-Ponte D, Carlevaro-Fita J, Palumbo E, Hermoso Pulido T, Guigo R, Johnson R. LncATLAS database for subcellular localization of long noncoding RNAs. RNA. 2017;23:1080–7.PubMedPubMedCentral Mas-Ponte D, Carlevaro-Fita J, Palumbo E, Hermoso Pulido T, Guigo R, Johnson R. LncATLAS database for subcellular localization of long noncoding RNAs. RNA. 2017;23:1080–7.PubMedPubMedCentral
220.
Zurück zum Zitat Wang Z, Tollervey J, Briese M, Turner D, Ule J. CLIP: construction of cDNA libraries for high-throughput sequencing from RNAs cross-linked to proteins in vivo. Methods. 2009;48:287–93.PubMed Wang Z, Tollervey J, Briese M, Turner D, Ule J. CLIP: construction of cDNA libraries for high-throughput sequencing from RNAs cross-linked to proteins in vivo. Methods. 2009;48:287–93.PubMed
222.
Zurück zum Zitat Wong NK, Huang CL, Islam R, Yip SP. Long non-coding RNAs in hematological malignancies: translating basic techniques into diagnostic and therapeutic strategies. J Hematol Oncol. 2018;11:131.PubMedPubMedCentral Wong NK, Huang CL, Islam R, Yip SP. Long non-coding RNAs in hematological malignancies: translating basic techniques into diagnostic and therapeutic strategies. J Hematol Oncol. 2018;11:131.PubMedPubMedCentral
223.
Zurück zum Zitat Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, et al. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol. 2018;11:63.PubMedPubMedCentral Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, et al. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol. 2018;11:63.PubMedPubMedCentral
224.
Zurück zum Zitat Tie Y, Liu B, Fu H, Zheng X. Circulating miRNA and cancer diagnosis. Sci China C Life Sci. 2009;52:1117–22.PubMed Tie Y, Liu B, Fu H, Zheng X. Circulating miRNA and cancer diagnosis. Sci China C Life Sci. 2009;52:1117–22.PubMed
225.
Zurück zum Zitat Rupaimoole R, Calin GA, Lopez-Berestein G, Sood AK. miRNA deregulation in cancer cells and the tumor microenvironment. Cancer Discov. 2016;6:235–46.PubMedPubMedCentral Rupaimoole R, Calin GA, Lopez-Berestein G, Sood AK. miRNA deregulation in cancer cells and the tumor microenvironment. Cancer Discov. 2016;6:235–46.PubMedPubMedCentral
226.
Zurück zum Zitat Tahamtan A, Ardebili A. Real-time RT-PCR in COVID-19 detection: issues affecting the results. Expert Rev Mol Diagn. 2020;20:453–4.PubMed Tahamtan A, Ardebili A. Real-time RT-PCR in COVID-19 detection: issues affecting the results. Expert Rev Mol Diagn. 2020;20:453–4.PubMed
227.
Zurück zum Zitat Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 2016;7:12429.PubMedPubMedCentral Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 2016;7:12429.PubMedPubMedCentral
228.
Zurück zum Zitat Carlevaro-Fita J, Johnson R. Global positioning system: understanding long noncoding RNAs through subcellular localization. Mol Cell. 2019;73:869–83.PubMed Carlevaro-Fita J, Johnson R. Global positioning system: understanding long noncoding RNAs through subcellular localization. Mol Cell. 2019;73:869–83.PubMed
Metadaten
Titel
Principles and innovative technologies for decrypting noncoding RNAs: from discovery and functional prediction to clinical application
verfasst von
Yu-Meng Sun
Yue-Qin Chen
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00945-8

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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