Next Article in Journal
Function and Clinical Implications of Long Non-Coding RNAs in Melanoma
Previous Article in Journal
Reply to the Letter to the Editor by D. D’Arcangelo et al.: “Ion Channels in Brain Metastasis”—Ion Channels in Cancer Set up and Metastatic Progression Ion Channels in Brain Metastasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

It Is Imperative to Establish a Pellucid Definition of Chimeric RNA and to Clear Up a Lot of Confusion in the Relevant Research

1
Department of Biochemistry, China Three Gorges University, Yichang 443002, China
2
Hormel Institute, University of Minnesota, Austin, MN 55912, USA
3
Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, China
4
Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical University, Guiyang 550004, China
5
School of Clinical Laboratory Science, Guizhou Medical University, Guiyang 550004, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(4), 714; https://doi.org/10.3390/ijms18040714
Submission received: 7 February 2017 / Revised: 15 March 2017 / Accepted: 17 March 2017 / Published: 28 March 2017
(This article belongs to the Section Biochemistry)

Abstract

:
There have been tens of thousands of RNAs deposited in different databases that contain sequences of two genes and are coined chimeric RNAs, or chimeras. However, “chimeric RNA” has never been lucidly defined, partly because “gene” itself is still ill-defined and because the means of production for many RNAs is unclear. Since the number of putative chimeras is soaring, it is imperative to establish a pellucid definition for it, in order to differentiate chimeras from regular RNAs. Otherwise, not only will chimeric RNA studies be misled but also characterization of fusion genes and unannotated genes will be hindered. We propose that only those RNAs that are formed by joining two RNA transcripts together without a fusion gene as a genomic basis should be regarded as authentic chimeras, whereas those RNAs transcribed as, and cis-spliced from, single transcripts should not be deemed as chimeras. Many RNAs containing sequences of two neighboring genes may be transcribed via a readthrough mechanism, and thus are actually RNAs of unannotated genes or RNA variants of known genes, but not chimeras. In today’s chimeric RNA research, there are still several key flaws, technical constraints and understudied tasks, which are also described in this perspective essay.

1. Introduction

Classically, mature RNAs in eukaryotic cells are categorized as ribosomal RNAs (rRNAs), messenger RNAs (mRNAs) and transfer RNAs (tRNAs), which are synthesized by RNA polymerases I, II and III, respectively [1]. One of us has recently proposed that noncoding RNAs, which can be processed from RNAs transcribed by either one of the three RNA polymerases, should be considered as the fourth category [1]. However, RNAs can also be classified by other criteria. For instance, while each RNA in any of the abovementioned four categories is derived from a single gene on a chromosome, there are so-called “chimeric RNAs” (herein “chimeras”), which defines those RNAs containing sequences of two genes. Actually, it remains possible that there are trimeric or even tetrameric RNAs, each of which contains sequences from three or four genes, respectively, although until now their existence is only suggested by many expression sequence tags (ESTs), but not by conclusive experimental evidence [2]. As described by some of us before [3,4,5], there hitherto have been tens of thousands of chimeric RNAs of human origin deposited in different databases, with a few examples in the references cited herein [2,6,7,8,9,10], while the entire human genome contains only slightly over 20,000 genes [1,11,12,13,14]. Therefore, mature RNAs can also be dichotomized based on whether they are derived from a single gene or from more than one gene. However, information from chimeric RNA research greatly baffles us in several aspects, largely because there still is a lack of a pellucid definition of chimeric RNAs, which in turn is partly because there is no clear answer to the very basic question “what is a gene?” [15,16,17]. In this essay, we describe our rumination over chimeric RNA, propose a new criterion for defining it to lucidly distinguish chimeras from other RNAs, and expound why we consider that most of the chimeric RNAs reported are inauthentic or should not be esteemed as chimeras. We also describe some common problems, flaws, technical constraints, and understudied tasks in today’s chimeric RNA research.

2. Currently, Chimeric RNAs Are Thought to Be Derived from Three Mechanisms

A mature RNA that contains sequences from two genes can be derived from three different mechanisms known hitherto [2,3,4]. The first and well-studied mechanism involves various chromosomal alterations, including DNA insertion, amplification [18], deletion and different types of rearrangement such as translocation. For instance, chromosomal translocation can cause fusion of chromosomes or chromosomal parts, resulting in one or two fusion sites and thus one or two fusion genes, as each fusion site may form a fusion gene. The first and also the best example of the consequence of translocation is the formation of the so-called Philadelphia chromosome observed in 1959 [19,20,21,22,23], which involves chromosomes 9 and 22, i.e., t(9;22)(q34;q11), and results in different BCL-ABL fusion genes [24,25]. There have been about 1,000 fusion genes identified hitherto [26], although ten times more are thought to exist, mainly in cancer [27]. In very rare situations, fusion genes can occur in normal human individuals as well, as exemplified by the TFG-GPR128 [28], POTE-actin [29,30,31], and PIPSL [32,33] genes. However, this type of fusion gene may actually be regarded as evolutionarily new genes, but not fusion ones [3]. Transcription of fusion genes can be initiated from alternative sites and/or terminated at alternative sites to yield different RNA transcripts, and each of these transcripts can undergo alternative cis-splicing to produce different mature RNAs as well. This means that fusion genes, once formed, are expressed and regulated just like other genes in eukaryotic cells with little difference, and therefore it makes more sense to regard their RNAs as regular ones, but not as chimeras.
The second mechanism is trans-splicing. While cis-splicing is a biochemical reaction with one RNA molecule as the substrate and one new RNA molecule as the product (scenarios A and B in Figure 1), trans-splicing is a biochemical reaction with two RNA molecules as the substrates but only one RNA molecule as the product (scenario C in Figure 1), which is a chimeric RNA [34]. Obviously, chimeric RNAs derived from trans-splicing have no genomic DNA basis [3,4]. Trans-splicing occurs often in unicellular organisms [35]. Some mitochondria and chloroplasts of low eukaryotes and plants also use trans-splicing to remove discontinuous group II introns [36,37]. Chimeric RNAs have been reported to occur in human cells and in cells of other animals in a physiological situation [5,38,39,40,41,42,43], with examples such as the KLK4 [44], Acy1-CoA and ACAT1 [45,46] RNAs in humans, the Dmr [47] and Msh4 mRNA variants [48] in mice, as well as some mdg4 mRNA variants in drosophila [49,50]. The IGH-BCL2 RNA chimera has been observed in normal spleen [51], whereas IGH-myc chimeric RNA has been found in mouse B lymphocytes [52] and in the Peyer’s patch follicles [53]. During the human developmental stage, normal uterine endometrium shows a chimeric RNA formed between the transcript of the JAZF1 gene on chromosome 7p15 and the transcript of the JJAZ1 gene at 17q11 [38,40]. Many of these chimeric RNAs in normal cells are thought to be derived from trans-splicing events [7,46,50,54,55], which, however, has hardly received unimpeachable experimental evidence, due largely to technical constraints, as described later. In our opinion, trans-splicing occurs only as rare events in cells of evolutionarily high animals, and in the physiological situation in humans, the events are probably as scarce as hen’s teeth. However, in carcinogenesis, which is an atavistic process [56,57,58], it may occur more often.
The third mechanism is for those chimeras formed by two neighboring genes on the same chromosome, which is the largest category of chimeric RNAs reported so far but, in our opinion, is also the most debatable one. The ENCODE (the Encyclopedia Of DNA Elements) project once estimated that transcripts from 65% (about two-thirds) of the human genes might be involved in forming chimeric RNAs, but the vast majority of these chimeras involve two neighboring genes on the same chromosome [34,59]. The ENCODE project did not provide mechanistic detail about how the two-neighboring-genes-containing chimeras were formed. However, some studies suggest that at least some of them may be derived from a mechanism of transcriptional readthough [60], i.e., transcription from the upstream gene to the downstream one [3,59], which is illustrated herein as scenario B in Figure 1, such as the SLC45A3-ELK4 RNA that is present in normal prostate and in prostate cancer [61]. In the database of the National Center for Biotechnology Information (NCBI) of the United States, such RNAs are indicated by using a hyphen to connect the two neighboring genes, as exemplified by the eight noncoding RNAs that are alternatively-splicing products of the TSNAX-DISC1 transcript (Figure 2 in [62]).

3. Most RNAs from Two Neighboring Genes Should Not Be Deemed as Chimeras

An RNA transcribed via the aforementioned readthrough mechanism can actually be regarded in three different ways: (1) it can be considered as an RNA variant of the upstream gene, dubbed herein as gene A, transcription of which does not stop at the annotated site but, instead, reads into and stops at the downstream gene, designated herein as gene B; (2) it can be considered as an RNA variant of gene B, transcription of which is initiated from an alternative site upstream of the annotated one; and (3) it can be deemed as an RNA transcript of an unannotated gene harbored at this genomic locus, herein referred to as gene C. We favor the third scenario, since it has been a well-accepted notion that “one gene may contain one or more other genes” with many examples in the NCBI database (some example illustrations are copied from the NCBI and shown in [1,62]). However, we have no objection to the first two scenarios that are actually the situations of alternative initiation and alternative termination, respectively, of transcription, mechanisms of which have all been well articulated in the literature for many genes. In any of these three scenarios, RNA transcripts may undergo regular cis-splicing, showing no any obvious difference from transcripts from already-annotated genes [63,64]. We favor the third scenario because these unannotated, i.e., newly discovered genes have no difference from, and should be studied like, already-annotated genes at all levels, including the levels of transcription, post-transcription, translation, post-translation, and protein transportation, although some of these genes may be noncoding and are not regulated at some of these levels, such as the TSNAX-DISC1 gene (Figure 2 in [62]). We strongly argue against the currently dominant concept in the chimeric RNA research that esteems the cis-splicing-derived mature RNAs from these unannotated genes as chimeras. This is because considering these RNAs as chimeras implies that these genes are different from the vast majority of already-annotated ones, thus misleading and encumbering their characterization at some of these levels, although it sounds more novel and more easily brings us grant supports and publications. On the other hand, considering most of these with two neighboring genes as individual unannotated ones also implies that are many more human genes than just approximately 20,000 genes as we currently thought [11,12,13,14], thus making more sense.
Theoretically, two neighboring genes may be transcribed separately to produce their own RNA transcripts in the regular way, but the two transcripts are then trans-spliced to an RNA, which is an unadulterated chimera (scenario C in Figure 1). The problem is that, although there have been many mature RNAs known to contain sequences of two adjacent genes, few, if any, studies provide cogent mechanistic detail about whether these RNAs are formed via cis-splicing of a single RNA transcribed via a readthrough mechanism or are formed via a trans-splicing of two RNA transcripts. In some studies that claim the occurrence of a trans-splicing event, little mechanistic detail and experimental evidence about such event per se have actually been given. Therefore, there is no way of knowing which of these reported two-neighboring genes-containing RNAs are authentic chimeras and which others are just cis-splicing-derived regular RNAs of unannotated genes, mainly because there is no feasible technique to determine RNA transcripts before splicing. Splicing occurs immediately after transcription is initiated [65], and is finished almost at the same time of transcription termination [66], leaving us only a small window of time to sift out the product RNAs from the substrate RNAs during a splicing procedure, especially for many genes in a high throughput manner.

4. There Are Different Ways to Catalog Chimeric RNAs

Chimeric RNAs can be sorted to different subgroups by different criteria, as recounted before [2,3,4]. Besides the criteria delineated above, a further criterion is the relationship between the two gene elements, herein referred to as two partners, of a chimera [2,8]. Based on this relationship, chimeras can be sorted to three groups, as illustrated in Figure 2. One group contains those in which the two partner sequences are reversely complementary to each other at a region called “short homologous sequence (SHS)”. Another group includes those in which the 5′ and 3′ partner sequences are directly connected. The third group includes those in which there is a sequence inserted between the two partners that cannot be matched to any genomic region and is thus called a “gap”. Actually, for the putative trimeric or tetrameric RNAs, the relationship between any two neighboring genes’ sequences should also be one of the three situations [2].

5. RT or PCR Creates Many Artifacts that Fabricate “Trans-Splicing”

Most known DNA polymerases require a DNA or RNA oligo as a primer to initiate the DNA synthesis, although some others use a protein to prime [67,68]. DNA polymerases are widely used in nucleic acid research. For instance, reverse transcriptases (RTases), such as those derived from the Avian myeloblastosis virus (AMV) [69] and the Moloney murine leukemia virus (MMLV) [70,71], as one type of DNA polymerase use tRNA as the primer in the parental retroviruses but can also use DNA primers in reverse transcription (RT) in vitro. On the other hand, Taq DNA polymerase is often used in polymerase chain reactions (PCR) with DNA primers, which often follows RT in molecular cloning procedure.
Lerat et al. [72] and Tuiskunen et al. [73] have once surmised that artifacts may occur in the following situations during an RT-PCR procedure: (1) RTase may be carried over from the RT system to the PCR tube wherein it allows RT to continue with the PCR primers as the primers and with the first strand of cDNA as the template; (2) when RNAs are carried over from the RT system to the PCR tube, Taq may have an RT activity and convert RNA to cDNA; and (3) the 3′ end of the first cDNA strand can loop back to form a hairpin structure and prime the synthesis of the second cDNA strand during the PCR. The first scenario would actually be consecutive RT reactions, as one of us phrased previously [3], for one of two basic reasons: First, if the first cDNA strand has its last several nucleotides (Nts) reversely complementary to the end of another RNA or cDNA, these Nts can anneal to this RNA or cDNA and use it as the template for the RT to proceed (scenario 1 in Figure 3). Since all DNA or RNA sequences are formed by only four bases, a several-Nt homolog (in a reversely complementary manner) should be common in the 3′ ends of numerous DNA or RNA shreds in a reaction tube. This is why usually in vitro RT does not really require the presence of random hexamers or even a poly-dT primer, because the RNA sample contains numerous RNA or DNA shards as endogenous random primers [3,69,70,74,75,76,77,78,79,80]. Second, most, if not all, DNA polymerases can append one or several Nts at the end of the newly synthesized DNA in a non-template manner [81,82,83,84,85,86], as one of us summarized before [3]. MMLV RTase as the most commonly used enzyme in RT more often appends GGG or CCC at the cDNA end [87,88,89,90,91,92], which can anneal to any RNA or DNA sequence ending with CCC or GGG to allow the RT to elongate (scenario 2 in Figure 3). If it is an RNA that anneals to the cDNA, an RNA–cDNA chimera is created as the first strand but then the second strand is a DNA chimera, synthesized in the first cycle of the ensuing PCR either by Taq or by the carried-over RTase [3].
Artifacts can also occur in PCR with production of many single-stranded DNA oligos or double-stranded DNA fragments that are shorter than the anticipated amplicon, due to aborted DNA polymerization. The last several Nts of these shorter DNA oligos or fragments may anneal to another gene’s cDNA and serve as the template for the DNA synthesis to continue, yielding a chimeric DNA fragment [93]. This first-strand of chimeric DNA may be elongated to a much longer sequence in the subsequent cycles of PCR.
16S rRNA, which is commonly used in molecular surveys of bacterial and archaeal diversity [94,95,96,97,98], has been well known to engender many chimeric cDNAs during cDNA library construction and the ensuing deep sequencing [93,99,100,101,102,103,104,105,106,107,108], which serve as good examples of fakes. A few 16S-rRNA-containing chimeras have also been reported in human and mouse cells [109,110,111,112,113,114,115,116,117], which are repeatedly detected and well-studied but in our opinion are technical artifacts [4]. They are frequently detected partly because in human and mouse cells the 16S rRNA is encoded by the mitochondrial genome that not only has hundreds or even thousands of copies in a single cell but also contains many reversely complementary regions [4]. These regions allow an RNA to loop back to form a hairpin structure, which allows the RT to elongate with the 5′ sequence as the template, creating a cDNA that looks like a product of trans-splicing of the sense and antisense transcripts (Figure 4). This sequence property of many 16S-rRNA-containing chimeras suggests to us that, if in a chimera one of the two partners has its sequence at the junction reversely complementary to a 5′ region of its parental RNA, it should alert us about the possibility of the self-priming derived spuriousness, as elucidated in more detail previously [4]. Transcription of 16S rRNA has been reported to be terminated at many sites [118,119], and these many ends of the RNAs create many opportunities for the scenarios illustrated in Figure 3 and Figure 4 to occur [4].
The above described self-priming by looping back often occurs in retroviruses [120], retroplasmids [121] and some bacteria [122]. The stem part of the hairpin structure formed via looping-back, which serves as the primer, should be short, because the reversely complementary region at the 3′ end of an RNA or DNA is usually short. A question is thus raised as to how short an RNA or DNA oligo can be a functional primer to initiate DNA synthesis. Unfortunately, this question has received much less attention so far and has not yet had a clear answer, but it may vary among different situations and depend on the DNA polymerase. RTases of the AMV and MMLV origins have been reported to use 15–18 Nts of a tRNA as a primer, and RTases from other retroviruses, such as the human immunodeficiency virus (HIV), also use similar numbers of Nts in tRNA as primers [123,124,125,126,127]. However, it is possible that the HIV RTase may only need a primer of 9 Nts [128], and it has also been reported that AMV RTase only requires a 3-Nt or 4-Nt DNA primer to prime DNA synthesis [129]. It has been reported that Taq requires a primer of, or longer than, 9-Nts, and for this reason some peers consider that Taq should not be able to act as an RTase in PCR even if some random hexamers are carried over from the RT system to the PCR tube [130]. One of us recently showed that a 4-Nt homolog might be able to prime DNA synthesis with an MMLV RTase and a Taq in a common RT-PCR procedure [4]. It is clear that random hexamers are sufficient to prime RT in vitro, as it is a routine lab practice. Of course, the efficiency of a primer depends not only on the length of the oligo but also on which bases that are in the primer and on the temperature provided for the annealing, which usually varies among different PCR procedures. Moreover, it is worth mentioning that unlike DNA polymerases, some RNA polymerases can initiate de novo RNA synthesis in a primer-independent manner, besides the back-priming like DNA polymerases [131], but whether this property may also create artifacts remains unknown.

6. There Are Other Artifacts with Unknown Mechanisms

While those chimeric RNAs in which one partner sequence is directly linked to the other without an SHS or a gap, i.e., those type 2 chimeras in Figure 2, may be artifacts created by a ligase during construction of cDNA libraries, the reason for the appearance of many cDNAs that contain a gap, as seen in many ESTs [2], is completely unknown. Trans-splicing should not be able to create such gap sequences that are completely unmatchable to the genomic DNA of any known organism deposited in the NCBI database [2]. Actually, there are many cDNA sequences, especially seen in RNA deep sequencing results, which cannot be matched to any organism’s genome in the NCBI database either. It is possible that these unmatchable sequences occur as artifacts in RT or PCR and may contribute to formation of artificial chimeric RNAs, although this hypothetic thinking still lacks convincing evidence. The gap sequences, which can be over one kilo-base pairs in many chimeric ESTs [2], may be good examples for the possible existence of unknown mechanisms for creating artifacts.

7. Currently, cDNA Protection Assay Is the Best Approach for Verification of Chimeric RNAs

Most of the tens of thousands of chimeric RNAs deposited in different databases have not yet been verified with any bench technique. Nevertheless, a small portion of reported chimeras have been verified using RT-PCR with or without confirmation by sequencing the cDNA, while an even smaller number of them have been authenticated using RNA protection assay, Northern blotting and/or in-situ hybridization. However, although these latter techniques have their merits and are more reliable than RT-PCR, none of them are authoritative enough to corroborate the true existence of a chimeric RNA. RNA protection assay does not involve RT or PCR amplification and thus will not create those RT- or PCR-related artifacts narrated above. However, because it does not involve amplification, its sensitivity is low and it can only detect those chimeras that are highly expressed. Moreover, the sequence of the probe used needs to be carefully designed or otherwise it can lead to data misinterpretation. The biggest weakness of RNA protection assay is that it does not provide sequence information of the protected RNA and thus cannot confirm that both partner sequences of a chimera have been protected. Actually, it is highly possible that only one of the two partner sequences is protected (Figure 5). Northern blotting has the same weaknesses as RNA protection assay and is less sensitive. In-situ hybridization assay has the same weakness as well, especially because it lacks sequence data to prove that the hybrid contains both, but not just one of the two, partner sequences (Figure 5).
Attempting to correct the weaknesses of the abovementioned methods, we have modified the RNA protection assay to a cDNA protection assay in which it is the cDNA, but not the parental RNA, that is protected [79]. In this assay, an aliquot of RNA is reversely transcribed to the first strand of cDNA. If an MMLV RTase, but not its mutant (MMLV-M), is used, inactivation of its RNase H activity ensues. The cDNA is then used as the probe to hybridize with another aliquot of the original RNA to form a RNA–cDNA hybrid. With its RNAase H activity inactivated, the carried-over RTase should not be able to digest the RNAs. After an S1 endonuclease is used to digest away irrelevant RNAs and all the excessive single-stranded cDNA probe, the RNA–cDNA hybrid is amplified using PCR with Taq, followed by DNA sequencing to confirm the identity of the protected cDNA. Compared with its parental RNA protection assay, this cDNA protection assay has two advantages, i.e., offering PCR amplification and DNA sequencing. Although this method involves RT that will likely create some bogus cDNA chimeras, the fakes will not be protected by unadulterated chimeric RNAs and will thus be chopped up by the S1.

8. We Propose New Criteria to Classify Chimeric RNAs

We propose to use “fusion RNA” to describe those RNAs transcribed from fusion genes, and to use “chimeric RNA” to describe those RNAs that are authentic chimeras derived from amalgamation of two RNA transcripts, via trans-splicing of two RNA molecules or via other currently unknown mechanisms, such as the hypothetical “transcriptional jump” and “chromosomal interaction” [132]. For those RNAs containing sequences of two adjacent genes on the same chromosome but sans a knowledge of how they are formed, we can temporarily put them under the umbrella of “readthrough RNA”. Of these RNAs, those that are later confirmed to be transcribed from unannotated genes via a readthrough mechanism should be excluded from this “readthrough RNA” category and deemed as RNAs of newly discovered regular genes that await annotation (scenario 2 in Figure 1). Since readthrough genes in the NCBI are indicated with a hyphen to join two neighboring genes together, as exemplified by the TSNAX-DISC1 (Figure 2 in [62]), the simplest way to annotate them is to follow this way of the NCBI. Those that are later confirmed to be products of trans-splicing of two individual transcripts (scenario 3 in Figure 1), or are formed via a currently unknown mechanism that joins two RNAs together, should also be removed from the “readthrough RNA” category but put into the authentic chimera family. In a nutshell, only those RNAs that lack a fusion gene as a genomic basis and are derived from two individual RNA molecules should be considered veritable chimeras, whereas fusion genes and unannotated genes are actually expressed and regulated in the same way as classically annotated genes, and thus there is no reason to consider them special.

9. There Are Other Flaws, Constraints and Understudied Tasks in Chimeric RNA Research

Most reported chimeras are “identified” using high-throughput sequencing or a bioinformatic analysis of ESTs, or coalescence of the two, and are dubbed by us as “putative chimeras” because they have not yet been verified. Besides those already described in the above sections, in our meditation today’s chimeric RNA research still has several other major flaws, technical constraints and understudied tasks:
  • There are still thousands of putative chimeric RNAs that contain sequences of two genes on two different chromosomes [27]. For most of them, it is still unclear whether they have a fusion gene as the genomic basis, and thus it is still unclear whether they are genuine chimeric RNAs by our definition.
  • Many RNAs are claimed to be derived from a readthrough mechanism but actually lack concrete experimental evidence proving the readthrough, since detection of a long pre-RNA transcript is one thing but causally-linking it to the mature RNA that contains two-gene sequences is another thing. Therefore, the possibility for them to be derived from a trans-splicing mechanism and thus to be genuine chimeric RNAs still exists, which needs to be ruled in or out.
  • Some chimeric RNAs have been reported to be recurrent, usually in cancer. However, often it is not clear whether the “recurrence” means that it is exactly the same chimeric sequence that appears repeatedly, or it means that the same two genes produce chimeric RNAs that are highly similar but still differ slightly in sequence. This question is raised because we sometimes have inquired of peers about the “recurrent” chimeric RNAs they observed and knew that the repeatedly detected chimeras differed slightly among each other in sequence. The reason for the small difference in sequence should be determined.
  • Technical detail is insufficiently discussed in many published studies of chimeric RNAs, especially about how possible it is that the chimeras are spurious. In our opinion, when describing a new chimeric RNA, more-than-usual technical detail needs to be furnished and, moreover, it needs to address: (1) whether it has a fusion gene as a genomic basis; and (2) whether it contains an SHS or a gap sequence that highly indicates an artifact.
  • Some chimeras encode proteins that share part of the sequence with the proteins produced from one or both of the parental genes. Determining whether these chimeras have protein products is technically difficult in most cases, due to the lack of fusion-protein specific primary antibodies, which in turn is due to a technical constraint in raising such antibodies. Antibodies raised via a traditional approach will likely recognize proteins from one or both parental genes. Theoretically, antibodies for the proteins from one of the two parental genes may be available and be used in western blotting to distinguish fusion-proteins from the proteins of the parental genes by their difference in molecular weight. However, since most genes produce multiple protein isoforms [62], as one of us has shown for protein products of many genes [133,134,135,136,137,138], in practice it is difficult to use these antibodies to corroborate the true existence of protein product(s) of a chimeric RNA, especially with an immunohistochemical staining approach that does not allow us to distinguish one protein from the others by their molecular weights.

10. Concluding Remarks

The overarching theme of this perspective essay is that chimeric RNA is still ill-defined and many of those chimeras deposited in various databases are likely to be technical artifacts or should not be regarded as chimeras, which may sound provocative to some peers. We propose that only those mature RNAs formed by joining two RNA molecules together via trans-splicing or some currently-unknown mechanisms without a fusion gene as a genomic basis should be authentic. Many of those containing two adjacent genes’ sequences are probably mature RNAs of newly discovered genes awaiting annotation, or are RNA variants of one of the two parental genes, but are not chimeric RNAs. Since there have already been tens of thousands of putative chimeric RNAs reported and the number is still soaring in the literature, it is imperative to clearly define chimeric RNAs and vigorously verify them. Otherwise, such a larger number of putative ones will not only mislead the chimeric RNA fraternity but will also hamper cloning and characterization of those fusion genes and those newly discovered genes. Actually, most fusion genes have not yet been fully characterized to such a detail as for the BCL-ABL fusion genes on the Philadelphia chromosome, while most two-neighboring-genes-containing RNAs have not yet been fully studied at the transcription and other steps. On the other hand, at present, trans-splicing is the only known mechanism for formation of authentic chimeras, but it may not be commonly used in human cells, meaning that either genuine chimeras in human cells are many fewer than many peers think or other unknown but commonly used mechanisms exist and await unearthing.

Acknowledgments

We would like to thank Fred Bogott at Austin Medical Center, Austin of Minnesota, for his excellent English editing of this manuscript. This work was supported by Chinese Natural Science Foundation grants to Hai Huang (grant No. 81460364) and D. Joshua Liao (grant No. 81660501).

Author Contributions

Chengfu Yuan, Wenxiu Yang, Wenfeng Yu, Zhizhong Guan, Hai Huang, and Dezhong Joshua Liao conceived the concepts presented; Chengfu Yuan, Hai Huang and Joshua Liao drafted and finalized the manuscript; and Yaping Han and Lucas Zellmer prepared or modified the figures and provided valuable comments on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jia, Y.; Chen, L.; Ma, Y.; Zhang, J.; Xu, N.; Liao, D.J. To Know How a Gene Works, We Need to Redefine It First but then, More Importantly, to Let the Cell Itself Decide How to Transcribe and Process Its RNAs. Int. J. Biol. Sci. 2015, 11, 1413–1423. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, W.; Wu, J.M.; Bi, A.D.; Ou-Yang, Y.C.; Shen, H.H.; Chirn, G.W.; Zhou, J.H.; Weiss, E.; Holman, E.P.; Liao, D.J. Possible Formation of Mitochondrial-RNA Containing Chimeric or Trimeric RNA Implies a Post-Transcriptional and Post-Splicing Mechanism for RNA Fusion. PLoS ONE 2013, 8, e77016. [Google Scholar] [CrossRef] [PubMed]
  3. Peng, Z.; Yuan, C.; Zellmer, L.; Liu, S.; Xu, N.; Liao, D.J. Hypothesis: Artifacts, Including Spurious Chimeric RNAs with a Short Homologous Sequence, Caused by Consecutive Reverse Transcriptions and Endogenous Random Primers. J. Cancer 2015, 6, 555–567. [Google Scholar] [CrossRef] [PubMed]
  4. Xie, B.; Yang, W.; Ouyang, Y.; Chen, L.; Jiang, H.; Liao, Y.; Liao, D.J. Two RNAs or DNAs May Artificially Fuse Together at a Short Homologous Sequence (SHS) during Reverse Transcription or Polymerase Chain Reactions, and Thus Reporting an SHS-Containing Chimeric RNA Requires Extra Caution. PLoS ONE 2016, 11, e0154855. [Google Scholar] [CrossRef] [PubMed]
  5. Zhou, J.; Liao, J.; Zheng, X.; Shen, H. Chimeric RNAs as potential biomarkers for tumor diagnosis. BMB Rep. 2012, 45, 133–140. [Google Scholar] [CrossRef] [PubMed]
  6. Ma, L.; Yang, S.; Zhao, W.; Tang, Z.; Zhang, T.; Li, K. Identification and analysis of pig chimeric mRNAs using RNA sequencing data. BMC Genom. 2012, 13, 429. [Google Scholar] [CrossRef] [PubMed]
  7. Lei, Q.; Li, C.; Zuo, Z.; Huang, C.; Cheng, H.; Zhou, R. Evolutionary Insights into RNA trans-Splicing in Vertebrates. Genome Biol. Evol. 2016, 8, 562–577. [Google Scholar] [CrossRef] [PubMed]
  8. Li, X.; Zhao, L.; Jiang, H.; Wang, W. Short homologous sequences are strongly associated with the generation of chimeric RNAs in eukaryotes. J. Mol. Evol. 2009, 68, 56–65. [Google Scholar] [CrossRef] [PubMed]
  9. Maher, C.A.; Kumar-Sinha, C.; Cao, X.; Kalyana-Sundaram, S.; Han, B.; Jing, X.; Sam, L.; Barrette, T.; Palanisamy, N.; Chinnaiyan, A.M. Transcriptome sequencing to detect gene fusions in cancer. Nature 2009, 458, 97–101. [Google Scholar] [CrossRef] [PubMed]
  10. Maher, C.A.; Palanisamy, N.; Brenner, J.C.; Cao, X.; Kalyana-Sundaram, S.; Luo, S.; Khrebtukova, I.; Barrette, T.R.; Grasso, C.; Yu, J.; et al. Chimeric transcript discovery by paired-end transcriptome sequencing. Proc. Natl. Acad. Sci. USA 2009, 106, 12353–12358. [Google Scholar] [CrossRef] [PubMed]
  11. Bamshad, M.J.; Ng, S.B.; Bigham, A.W.; Tabor, H.K.; Emond, M.J.; Nickerson, D.A.; Shendure, J. Exome sequencing as a tool for Mendelian disease gene discovery. Nat. Rev. Genet. 2011, 12, 745–755. [Google Scholar] [CrossRef] [PubMed]
  12. Belizario, J.E. The humankind genome: From genetic diversity to the origin of human diseases. Genome 2013, 56, 705–716. [Google Scholar] [CrossRef] [PubMed]
  13. Pennisi, E. Genomics. ENCODE project writes eulogy for junk DNA. Science 2012, 337, 1159–1161. [Google Scholar] [CrossRef] [PubMed]
  14. Skipper, M.; Dhand, R.; Campbell, P. Presenting ENCODE. Nature 2012, 489, 45. [Google Scholar] [CrossRef] [PubMed]
  15. Finta, C.; Warner, S.C.; Zaphiropoulos, P.G. Intergenic mRNAs. Minor gene products or tools of diversity? Histol. Histopathol. 2002, 17, 677–682. [Google Scholar] [PubMed]
  16. Gerstein, M.B.; Bruce, C.; Rozowsky, J.S.; Zheng, D.; Du, J.; Korbel, J.O.; Emanuelsson, O.; Zhang, Z.D.; Weissman, S.; Snyder, M. What is a gene, post-ENCODE? History and updated definition. Genome Res. 2007, 17, 669–681. [Google Scholar] [CrossRef] [PubMed]
  17. Portin, P. The elusive concept of the gene. Hereditas 2009, 146, 112–117. [Google Scholar] [CrossRef] [PubMed]
  18. Jia, Y.; Chen, L.; Jia, Q.; Dou, X.; Xu, N.; Liao, D.J. The well-accepted notion that gene amplification contributes to increased expression still remains, after all these years, a reasonable but unproven assumption. J. Carcinog. 2016, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  19. Nowell, P.C.; Hungerford, D.A. Chromosome studies on normal and leukemic human leukocytes. J. Natl. Cancer Inst. 1960, 25, 85–109. [Google Scholar] [PubMed]
  20. Hungerford, D.A. The philadelphia chromosome and some others. Ann. Intern. Med. 1964, 61, 789–793. [Google Scholar] [CrossRef] [PubMed]
  21. Nowell, P.C. The minute chromosome (Phl) in chronic granulocytic leukemia. Blut 1962, 8, 65–66. [Google Scholar] [CrossRef] [PubMed]
  22. Koretzky, G.A. The legacy of the Philadelphia chromosome. J. Clin. Investig. 2007, 117, 2030–2032. [Google Scholar] [CrossRef] [PubMed]
  23. Nowell, P.H.D. A minute chromosome in human chronic granulocytic leukemia. Science 1960, 132, 1497. [Google Scholar]
  24. Bennour, A.; Ouahchi, I.; Moez, M.; Elloumi, M.; Khelif, A.; Saad, A.; Sennana, H. Comprehensive analysis of BCR/ABL variants in chronic myeloid leukemia patients using multiplex RT-PCR. Clin. Lab. 2012, 58, 433–439. [Google Scholar] [PubMed]
  25. Ma, W.; Kantarjian, H.; Yeh, C.H.; Zhang, Z.J.; Cortes, J.; Albitar, M. BCR-ABL truncation due to premature translation termination as a mechanism of resistance to kinase inhibitors. Acta Haematol. 2009, 121, 27–31. [Google Scholar] [CrossRef] [PubMed]
  26. Mertens, F.; Tayebwa, J. Evolving techniques for gene fusion detection in soft tissue tumours. Histopathology 2014, 64, 151–162. [Google Scholar] [CrossRef] [PubMed]
  27. Mertens, F.; Johansson, B.; Fioretos, T.; Mitelman, F. The emerging complexity of gene fusions in cancer. Nat. Rev. Cancer 2015, 15, 371–381. [Google Scholar] [CrossRef] [PubMed]
  28. Chase, A.; Ernst, T.; Fiebig, A.; Collins, A.; Grand, F.; Erben, P.; Reiter, A.; Schreiber, S.; Cross, N.C. TFG, a target of chromosome translocations in lymphoma and soft tissue tumors, fuses to GPR128 in healthy individuals. Haematologica 2010, 95, 20–26. [Google Scholar] [CrossRef] [PubMed]
  29. Lee, Y.; Ise, T.; Ha, D.; Saint Fleur, A.; Hahn, Y.; Liu, X.F.; Nagata, S.; Lee, B.; Bera, T.K.; Pastan, I. Evolution and expression of chimeric POTE-actin genes in the human genome. Proc. Natl. Acad. Sci. USA 2006, 103, 17885–17890. [Google Scholar] [CrossRef] [PubMed]
  30. Liu, X.F.; Bera, T.K.; Liu, L.J.; Pastan, I. A primate-specific POTE-actin fusion protein plays a role in apoptosis. Apoptosis 2009, 14, 1237–1244. [Google Scholar] [CrossRef] [PubMed]
  31. Ise, T.; Das, S.; Nagata, S.; Maeda, H.; Lee, Y.; Onda, M.; Anver, M.R.; Bera, T.K.; Pastan, I. Expression of POTE protein in human testis detected by novel monoclonal antibodies. Biochem. Biophys. Res. Commun. 2008, 365, 603–608. [Google Scholar] [CrossRef] [PubMed]
  32. Babushok, D.V.; Ohshima, K.; Ostertag, E.M.; Chen, X.; Wang, Y.; Mandal, P.K.; Okada, N.; Abrams, C.S.; Kazazian, H.H., Jr. A novel testis ubiquitin-binding protein gene arose by exon shuffling in hominoids. Genome Res. 2007, 17, 1129–1138. [Google Scholar] [CrossRef] [PubMed]
  33. Ohshima, K.; Igarashi, K. Inference for the initial stage of domain shuffling: Tracing the evolutionary fate of the PIPSL retrogene in hominoids. Mol. Biol. Evol. 2010, 27, 2522–2533. [Google Scholar] [CrossRef] [PubMed]
  34. Gingeras, T.R. Implications of chimaeric non-co-linear transcripts. Nature 2009, 461, 206–211. [Google Scholar] [CrossRef] [PubMed]
  35. Lasda, E.L.; Blumenthal, T. Trans-splicing. Wiley Interdiscip. Rev. RNA 2011, 2, 417–434. [Google Scholar] [CrossRef] [PubMed]
  36. Glanz, S.; Kuck, U. Trans-splicing of organelle introns—A detour to continuous RNAs. Bioessays 2009, 31, 921–934. [Google Scholar] [CrossRef] [PubMed]
  37. Jacobs, J.; Glanz, S.; Bunse-Grassmann, A.; Kruse, O.; Kuck, U. RNA trans-splicing: Identification of components of a putative chloroplast spliceosome. Eur. J. Cell Biol. 2010, 89, 932–939. [Google Scholar] [CrossRef]
  38. Rowley, J.D.; Blumenthal, T. Medicine. The cart before the horse. Science 2008, 321, 1302–1304. [Google Scholar] [CrossRef] [PubMed]
  39. Yuan, H.; Qin, F.; Movassagh, M.; Park, H.; Golden, W.; Xie, Z.; Zhang, P.; Sklar, J.; Li, H. A chimeric RNA characteristic of rhabdomyosarcoma in normal myogenesis process. Cancer Discov. 2013, 3, 1394–1403. [Google Scholar] [CrossRef] [PubMed]
  40. Li, H.; Wang, J.; Mor, G.; Sklar, J. A neoplastic gene fusion mimics trans-splicing of RNAs in normal human cells. Science 2008, 321, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  41. Li, H.; Wang, J.; Ma, X.; Sklar, J. Gene fusions and RNA trans-splicing in normal and neoplastic human cells. Cell Cycle 2009, 8, 218–222. [Google Scholar] [CrossRef] [PubMed]
  42. Babiceanu, M.; Qin, F.; Xie, Z.; Jia, Y.; Lopez, K.; Janus, N.; Facemire, L.; Kumar, S.; Pang, Y.; Qi, Y.; et al. Recurrent chimeric fusion RNAs in non-cancer tissues and cells. Nucleic Acids Res. 2016, 44, 2859–2872. [Google Scholar] [CrossRef] [PubMed]
  43. Fang, W.; Wei, Y.; Kang, Y.; Landweber, L.F. Detection of a common chimeric transcript between human chromosomes 7 and 16. Biol. Direct 2012, 7, 49. [Google Scholar] [CrossRef] [PubMed]
  44. Lai, J.; Lehman, M.L.; Dinger, M.E.; Hendy, S.C.; Mercer, T.R.; Seim, I.; Lawrence, M.G.; Mattick, J.S.; Clements, J.A.; Nelson, C.C. A variant of the KLK4 gene is expressed as a cis sense-antisense chimeric transcript in prostate cancer cells. RNA 2010, 16, 1156–1166. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, J.; Zhao, X.N.; Yang, L.; Hu, G.J.; Lu, M.; Xiong, Y.; Yang, X.Y.; Chang, C.C.; Song, B.L.; Chang, T.Y.; et al. RNA secondary structures located in the interchromosomal region of human ACAT1 chimeric mRNA are required to produce the 56-kDa isoform. Cell Res. 2008, 18, 921–936. [Google Scholar] [CrossRef] [PubMed]
  46. Hu, G.J.; Chen, J.; Zhao, X.N.; Xu, J.J.; Guo, D.Q.; Lu, M.; Zhu, M.; Xiong, Y.; Li, Q.; Chang, C.C.; et al. Production of ACAT1 56-kDa isoform in human cells via trans-splicing involving the ampicillin resistance gene. Cell Res. 2013, 23, 1007–1024. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, L.; Lu, H.; Xin, D.; Cheng, H.; Zhou, R. A novel ncRNA gene from mouse chromosome 5 trans-splices with Dmrt1 on chromosome 19. Biochem. Biophys. Res. Commun. 2010, 400, 696–700. [Google Scholar] [CrossRef] [PubMed]
  48. Hirano, M.; Noda, T. Genomic organization of the mouse Msh4 gene producing bicistronic, chimeric and antisense mRNA. Gene 2004, 342, 165–177. [Google Scholar] [CrossRef] [PubMed]
  49. Gabler, M.; Volkmar, M.; Weinlich, S.; Herbst, A.; Dobberthien, P.; Sklarss, S.; Fanti, L.; Pimpinelli, S.; Kress, H.; Reuter, G.; et al. Trans-splicing of the mod(mdg4) complex locus is conserved between the distantly related species Drosophila melanogaster and D. virilis. Genetics 2005, 169, 723–736. [Google Scholar] [CrossRef] [PubMed]
  50. Labrador, M.; Mongelard, F.; Plata-Rengifo, P.; Baxter, E.M.; Corces, V.G.; Gerasimova, T.I. Protein encoding by both DNA strands. Nature 2001, 409, 1000. [Google Scholar] [CrossRef] [PubMed]
  51. Janz, S.; Potter, M.; Rabkin, C.S. Lymphoma- and leukemia-associated chromosomal translocations in healthy individuals. Genes Chromosom. Cancer 2003, 36, 211–223. [Google Scholar] [CrossRef] [PubMed]
  52. Osborne, C.S.; Chakalova, L.; Mitchell, J.A.; Horton, A.; Wood, A.L.; Bolland, D.J.; Corcoran, A.E.; Fraser, P. Myc dynamically and preferentially relocates to a transcription factory occupied by Igh. PLoS Biol. 2007, 5, e192. [Google Scholar] [CrossRef] [PubMed]
  53. Muller, J.R.; Mushinski, E.B.; Williams, J.A.; Hausner, P.F. Immunoglobulin/Myc recombinations in murine Peyer’s patch follicles. Genes Chromosom. Cancer 1997, 20, 1–8. [Google Scholar] [CrossRef]
  54. Horiuchi, T.; Aigaki, T. Alternative trans-splicing: A novel mode of pre-mRNA processing. Biol. Cell 2006, 98, 135–140. [Google Scholar] [CrossRef] [PubMed]
  55. Zaphiropoulos, P.G. Trans-splicing in Higher Eukaryotes: Implications for Cancer Development? Front. Genet. 2011, 2, 92. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, G.; Chen, L.; Yu, B.; Zellmer, L.; Xu, N.; Liao, D.J. Learning about the Importance of Mutation Prevention from Curable Cancers and Benign Tumors. J. Cancer 2016, 7, 436–445. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, J.; Lou, XM.; Jin, LY.; Zhou, R.; Liu, S.; Xu, N.; Liao, D.J. Necrosis, and then stress induced necrosis-like cell death, but not apoptosis, should be the preferred cell death mode for chemotherapy: Clearance of a few misconceptions. Oncoscience 2014, 1, 407–422. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, J.; Lou, X.; Zellmer, L.; Liu, S.; Xu, N.; Liao, D.J. Just like the rest of evolution in Mother Nature, the evolution of cancers may be driven by natural selection, and not by haphazard mutations. Oncoscience 2014, 1, 580–590. [Google Scholar] [CrossRef] [PubMed]
  59. Birney, E.; Stamatoyannopoulos, J.A.; Dutta, A.; Guigo, R.; Gingeras, T.R.; Margulies, E.H.; Weng, Z.; Snyder, M.; Dermitzakis, E.T.; Thurman, R.E.; et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007, 447, 799–816. [Google Scholar] [CrossRef] [PubMed]
  60. Akiva, P.; Toporik, A.; Edelheit, S.; Peretz, Y.; Diber, A.; Shemesh, R.; Novik, A.; Sorek, R. Transcription-mediated gene fusion in the human genome. Genome Res. 2006, 16, 30–36. [Google Scholar] [CrossRef] [PubMed]
  61. Rickman, D.S.; Pflueger, D.; Moss, B.; van Doren, V.E.; Chen, C.X.; Kuefer, R.; Tewari, A.K.; Setlur, S.R.; Demichelis, F.; et al. SLC45A3-ELK4 is a novel and frequent erythroblast transformation-specific fusion transcript in prostate cancer. Cancer Res. 2009, 69, 2734–2738. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, X.; Wang, Y.; Yang, W.; Guan, Z.; Yu, W.; Liao, D.J. Protein multiplicity can lead to misconduct in western blotting and misinterpretation of immunohistochemical staining results, creating much conflicting data. Prog. Histochem. Cytochem. 2016, 51, 51–58. [Google Scholar] [CrossRef] [PubMed]
  63. Lai, J.; An, J.; Seim, I.; Walpole, C.; Hoffman, A.; Moya, L.; Srinivasan, S.; Perry-Keene, J.L.; Australian Prostate Cancer Bioresource; Wang, C.; et al. Erratum to: Fusion transcript loci share many genomic features with non-fusion loci. BMC Genom. 2016, 17, 424. [Google Scholar] [CrossRef] [PubMed]
  64. Lai, J.; An, J.; Seim, I.; Walpole, C.; Hoffman, A.; Moya, L.; Srinivasan, S.; Perry-Keene, J.L.; Australian Prostate Cancer Bioresource; Wang, C.; et al. Fusion transcript loci share many genomic features with non-fusion loci. BMC Genom. 2015, 16, 1021. [Google Scholar] [CrossRef] [PubMed]
  65. Georgomanolis, T.; Sofiadis, K.; Papantonis, A. Cutting a Long Intron Short: Recursive Splicing and Its Implications. Front. Physiol. 2016, 7, 598. [Google Scholar] [CrossRef] [PubMed]
  66. Yang, M.; Wu, J.; Wu, S.H.; Bi, A.D.; Liao, D.J. Splicing of mouse p53 pre-mRNA does not always follow the “first come, first served” principle and may be influenced by cisplatin treatment and serum starvation. Mol. Biol. Rep. 2012, 39, 9247–9256. [Google Scholar] [CrossRef] [PubMed]
  67. Salas, M.; de Vega, M. Protein-Primed Replication of Bacteriophage Phi29 DNA. Enzymes 2016, 39, 137–167. [Google Scholar] [PubMed]
  68. Salas, M.; Holguera, I.; Redrejo-Rodriguez, M.; de Vega, M. DNA-Binding Proteins Essential for Protein-Primed Bacteriophage Phi29 DNA Replication. Front. Mol. Biosci. 2016, 3, 37. [Google Scholar] [CrossRef] [PubMed]
  69. Harada, F.; Sawyer, R.C.; Dahlberg, J.E. A primer ribonucleic acid for initiation of in vitro Rous sarcarcoma virus deoxyribonucleic acid synthesis. J. Biol. Chem. 1975, 250, 3487–3497. [Google Scholar] [PubMed]
  70. Harada, F.; Peters, G.G.; Dahlberg, J.E. The primer tRNA for Moloney murine leukemia virus DNA synthesis. Nucleotide sequence and aminoacylation of tRNAPro. J. Biol. Chem. 1979, 254, 10979–10985. [Google Scholar] [PubMed]
  71. Peters, G.; Harada, F.; Dahlberg, J.E.; Panet, A.; Haseltine, W.A.; Baltimore, D. Low-molecular-weight RNAs of Moloney murine leukemia virus: Identification of the primer for RNA-directed DNA synthesis. J. Virol. 1977, 21, 1031–1041. [Google Scholar] [PubMed]
  72. Lerat, H.; Berby, F.; Trabaud, M.A.; Vidalin, O.; Major, M.; Trepo, C.; Inchauspe, G. Specific detection of hepatitis C virus minus strand RNA in hematopoietic cells. J. Clin. Investig. 1996, 97, 845–851. [Google Scholar] [CrossRef] [PubMed]
  73. Tuiskunen, A.; Leparc-Goffart, I.; Boubis, L.; Monteil, V.; Klingstrom, J.; Tolou, H.J.; Lundkvist, A.; Plumet, S. Self-priming of reverse transcriptase impairs strand-specific detection of dengue virus RNA. J. Gen. Virol. 2010, 91, 1019–1027. [Google Scholar] [CrossRef] [PubMed]
  74. Adrover, M.F.; Munoz, M.J.; Baez, M.V.; Thomas, J.; Kornblihtt, A.R.; Epstein, A.L.; Jerusalinsky, D.A. Characterization of specific cDNA background synthesis introduced by reverse transcription in RT-PCR assays. Biochimie 2010, 92, 1839–1846. [Google Scholar] [CrossRef] [PubMed]
  75. Frech, B.; Peterhans, E. RT-PCR: ‘background priming’ during reverse transcription. Nucleic Acids Res. 1994, 22, 4342–4343. [Google Scholar] [CrossRef] [PubMed]
  76. Haddad, F.; Qin, A.X.; Bodell, P.W.; Zhang, L.Y.; Guo, H.; Giger, J.M.; Baldwin, K.M. Regulation of antisense RNA expression during cardiac MHC gene switching in response to pressure overload. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H2351–H2361. [Google Scholar] [CrossRef] [PubMed]
  77. Moison, C.; Arimondo, P.B.; Guieysse-Peugeot, A.L. Commercial reverse transcriptase as source of false-positive strand-specific RNA detection in human cells. Biochimie 2011, 93, 1731–1737. [Google Scholar] [CrossRef] [PubMed]
  78. Stahlberg, A.; Hakansson, J.; Xian, X.; Semb, H.; Kubista, M. Properties of the reverse transcription reaction in mRNA quantification. Clin. Chem. 2004, 50, 509–515. [Google Scholar] [CrossRef] [PubMed]
  79. Yuan, C.; Liu, Y.; Yang, M.; Liao, D.J. New methods as alternative or corrective measures for the pitfalls and artifacts of reverse transcription and polymerase chain reactions (RT-PCR) in cloning chimeric or antisense-accompanied RNA. RNA Biol. 2013, 10, 958–967. [Google Scholar] [CrossRef] [PubMed]
  80. Gubler, U. Second-strand cDNA synthesis: mRNA fragments as primers. Methods Enzymol. 1987, 152, 330–335. [Google Scholar] [PubMed]
  81. Hanaki, K.; Odawara, T.; Muramatsu, T.; Kuchino, Y.; Masuda, M.; Yamamoto, K.; Nozaki, C.; Mizuno, K.; Yoshikura, H. Primer/template-independent synthesis of poly d(A-T) by Taq polymerase. Biochem. Biophys. Res. Commun. 1997, 238, 113–118. [Google Scholar] [CrossRef] [PubMed]
  82. Hanaki, K.; Odawara, T.; Nakajima, N.; Shimizu, Y.K.; Nozaki, C.; Mizuno, K.; Muramatsu, T.; Kuchino, Y.; Yoshikura, H. Two different reactions involved in the primer/template-independent polymerization of dATP and dTTP by Taq DNA polymerase. Biochem. Biophys. Res. Commun. 1998, 244, 210–219. [Google Scholar] [CrossRef] [PubMed]
  83. Hanaki, K.; Nakatake, H.; Yamamoto, K.; Odawara, T.; Yoshikura, H. DNase I activity retained after heat inactivation in standard buffer. Biotechniques 2000, 29, 38–40, 42. [Google Scholar] [PubMed]
  84. Hanaki, K.; Nishihara, T.; Odawara, T.; Nakajima, N.; Yamamoto, K.; Yoshikura, H. RNAse A treatment of Taq and Tth DNA polymerases eliminates primer/template-independent poly(dA-dT) synthesis. Biotechniques 2001, 31, 734, 736, 738. [Google Scholar] [PubMed]
  85. Nakajima, N.; Hanaki, K.; Shimizu, Y.K.; Ohnishi, S.; Gunji, T.; Nakajima, A.; Nozaki, C.; Mizuno, K.; Odawara, T.; Yoshikura, H. Hybridization-AT-tailing (HybrAT) method for sensitive and strand-specific detection of DNA and RNA. Biochem. Biophys. Res. Commun. 1998, 248, 613–620. [Google Scholar] [CrossRef] [PubMed]
  86. Zhou, M.Y.; Gomez-Sanchez, C.E. Universal TA cloning. Curr. Issues Mol. Biol. 2000, 2, 1–7. [Google Scholar] [PubMed]
  87. Alldred, M.J.; Che, S.; Ginsberg, S.D. Terminal continuation (TC) RNA amplification without second strand synthesis. J. Neurosci. Methods 2009, 177, 381–385. [Google Scholar] [CrossRef] [PubMed]
  88. Che, S.; Ginsberg, S.D. Amplification of RNA transcripts using terminal continuation. Lab. Investig. 2004, 84, 131–137. [Google Scholar] [CrossRef] [PubMed]
  89. Ginsberg, S.D.; Che, S. RNA amplification in brain tissues. Neurochem. Res. 2002, 27, 981–992. [Google Scholar] [CrossRef] [PubMed]
  90. Wellenreuther, R.; Schupp, I.; Poustka, A.; Wiemann, S. SMART amplification combined with cDNA size fractionation in order to obtain large full-length clones. BMC Genom. 2004, 5, 36. [Google Scholar] [CrossRef] [PubMed]
  91. Gubler, U. Second-strand cDNA synthesis: Classical method. Methods Enzymol. 1987, 152, 325–329. [Google Scholar] [PubMed]
  92. Wang, E.; Miller, L.D.; Ohnmacht, G.A.; Liu, E.T.; Marincola, F.M. High-fidelity mRNA amplification for gene profiling. Nat. Biotechnol. 2000, 18, 457–459. [Google Scholar] [PubMed]
  93. Haas, B.J.; Gevers, D.; Earl, A.M.; Feldgarden, M.; Ward, D.V.; Giannoukos, G.; Ciulla, D.; Tabbaa, D.; Highlander, S.K.; Sodergren, E.; et al. Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res. 2011, 21, 494–504. [Google Scholar] [CrossRef] [PubMed]
  94. Galan, M.; Razzauti, M.; Bard, E.; Bernard, M.; Brouat, C.; Charbonnel, N.; Dehne-Garcia, A.; Loiseau, A.; Tatard, C.; Tamisier, L.; et al. 16S rRNA Amplicon Sequencing for Epidemiological Surveys of Bacteria in Wildlife. mSystems 2016, 1, e00032-16. [Google Scholar] [CrossRef] [PubMed]
  95. Garrett, R.A. A backward view from 16S rRNA to archaea to the universal tree of life to progenotes: Reminiscences of Carl Woese. RNA Biol. 2014, 11, 232–235. [Google Scholar] [CrossRef] [PubMed]
  96. Ellegaard, K.M.; Engel, P. Beyond 16S rRNA Community Profiling: Intra-Species Diversity in the Gut Microbiota. Front. Microbiol. 2016, 7, 1475. [Google Scholar] [CrossRef] [PubMed]
  97. Logares, R.; Sunagawa, S.; Salazar, G.; Cornejo-Castillo, F.M.; Ferrera, I.; Sarmento, H.; Sarmento, H.; Hingamp, P.; Ogata, H.; de Vargas, C.; et al. Metagenomic 16S rDNA Illumina tags are a powerful alternative to amplicon sequencing to explore diversity and structure of microbial communities. Environ. Microbiol. 2014, 16, 2659–2671. [Google Scholar] [CrossRef] [PubMed]
  98. Vos, M.; Quince, C.; Pijl, A.S.; de Hollander, M.; Kowalchuk, G.A. A comparison of rpoB and 16S rRNA as markers in pyrosequencing studies of bacterial diversity. PLoS ONE 2012, 7, e30600. [Google Scholar] [CrossRef] [PubMed]
  99. Acinas, S.G.; Sarma-Rupavtarm, R.; Klepac-Ceraj, V.; Polz, M.F. PCR-induced sequence artifacts and bias: Insights from comparison of two 16S rRNA clone libraries constructed from the same sample. Appl. Environ. Microbiol. 2005, 71, 8966–8969. [Google Scholar] [CrossRef] [PubMed]
  100. Ashelford, K.E.; Chuzhanova, N.A.; Fry, J.C.; Jones, A.J.; Weightman, A.J. At least 1 in 20 16S rRNA sequence records currently held in public repositories is estimated to contain substantial anomalies. Appl. Environ. Microbiol. 2005, 71, 7724–7736. [Google Scholar] [CrossRef] [PubMed]
  101. Ashelford, K.E.; Chuzhanova, N.A.; Fry, J.C.; Jones, A.J.; Weightman, A.J. New screening software shows that most recent large 16S rRNA gene clone libraries contain chimeras. Appl. Environ. Microbiol. 2006, 72, 5734–5741. [Google Scholar] [CrossRef] [PubMed]
  102. De Santis, T.Z.; Hugenholtz, P.; Larsen, N.; Rojas, M.; Brodie, E.L.; Keller, K.; Huber, T.; Dalevi, D.; Hu, P.; Andersen, G.L. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 2006, 72, 5069–5072. [Google Scholar] [CrossRef] [PubMed]
  103. Huber, T.; Faulkner, G.; Hugenholtz, P. Bellerophon: A program to detect chimeric sequences in multiple sequence alignments. Bioinformatics 2004, 20, 2317–2319. [Google Scholar] [CrossRef] [PubMed]
  104. Hugenholtz, P.; Huber, T. Chimeric 16S rDNA sequences of diverse origin are accumulating in the public databases. Int. J. Syst. Evol. Microbiol. 2003, 53, 289–293. [Google Scholar] [CrossRef] [PubMed]
  105. Lahr, D.J.; Katz, L.A. Reducing the impact of PCR-mediated recombination in molecular evolution and environmental studies using a new-generation high-fidelity DNA polymerase. Biotechniques 2009, 47, 857–866. [Google Scholar] [PubMed]
  106. Quince, C.; Lanzen, A.; Curtis, T.P.; Davenport, R.J.; Hall, N.; Head, I.M.; Read, L.F.; Sloan, W.T. Accurate determination of microbial diversity from 454 pyrosequencing data. Nat. Methods 2009, 6, 639–641. [Google Scholar] [CrossRef] [PubMed]
  107. Thompson, J.R.; Marcelino, L.A.; Polz, M.F. Heteroduplexes in mixed-template amplifications: Formation, consequence and elimination by “reconditioning PCR”. Nucleic Acids Res. 2002, 30, 2083–2088. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, G.C.; Wang, Y. Frequency of formation of chimeric molecules as a consequence of PCR coamplification of 16S rRNA genes from mixed bacterial genomes. Appl. Environ. Microbiol. 1997, 63, 4645–4650. [Google Scholar] [PubMed]
  109. Burzio, V.A.; Villota, C.; Villegas, J.; Landerer, E.; Boccardo, E.; Villa, L.L.; Martínez, R.; Lopez, C.; Gaete, F.; Toro, V.; et al. Expression of a family of noncoding mitochondrial RNAs distinguishes normal from cancer cells. Proc. Natl. Acad. Sci. USA 2009, 106, 9430–9434. [Google Scholar] [CrossRef] [PubMed]
  110. Landerer, E.; Villegas, J.; Burzio, V.A.; Oliveira, L.; Villota, C.; Lopez, C.; Restovic, F.; Martinez, R.; Castillo, O.; Burzio, L.O. Nuclear localization of the mitochondrial ncRNAs in normal and cancer cells. Cell Oncol. 2011, 34, 297–305. [Google Scholar] [CrossRef] [PubMed]
  111. Rivas, A.; Burzio, V.; Landerer, E.; Borgna, V.; Gatica, S.; Avila, R.; Lopez, C.; Villota, C.; de la Fuente, R.; Echenique, J.; et al. Determination of the differential expression of mitochondrial long non-coding RNAs as a noninvasive diagnosis of bladder cancer. BMC Urol. 2012, 12, 37. [Google Scholar] [CrossRef] [PubMed]
  112. Vidaurre, S.; Fitzpatrick, C.; Burzio, V.A.; Briones, M.; Villota, C.; Villegas, J.; Echenique, J.; Oliveira-Cruz, L.; Araya, M.; Borgna, V.; et al. Down-regulation of the antisense mitochondrial non-coding RNAs (ncRNAs) is a unique vulnerability of cancer cells and a potential target for cancer therapy. J. Biol. Chem. 2014, 289, 27182–27198. [Google Scholar] [CrossRef] [PubMed]
  113. Villegas, J.; Zarraga, A.M.; Muller, I.; Montecinos, L.; Werner, E.; Brito, M.; Meneses, A.M.; Burzio, L.O. A novel chimeric mitochondrial RNA localized in the nucleus of mouse sperm. DNA Cell Biol. 2000, 19, 579–588. [Google Scholar] [CrossRef] [PubMed]
  114. Villegas, J.; Araya, P.; Bustos-Obregon, E.; Burzio, L.O. Localization of the 16S mitochondrial rRNA in the nucleus of mammalian spermatogenic cells. Mol. Hum. Reprod. 2002, 8, 977–983. [Google Scholar] [CrossRef] [PubMed]
  115. Villegas, J.; Burzio, V.; Villota, C.; Landerer, E.; Martinez, R.; Santander, M.; Martinez, R.; Pinto, R.; Vera, M.I.; Boccardo, E.; et al. Expression of a novel non-coding mitochondrial RNA in human proliferating cells. Nucleic Acids Res. 2007, 35, 7336–7347. [Google Scholar] [CrossRef] [PubMed]
  116. Villota, C.; Campos, A.; Vidaurre, S.; Oliveira-Cruz, L.; Boccardo, E.; Burzio, V.A.; Varas, M.; Villegas, J.; Villa, L.L.; Valenzuela, P.D.; et al. Expression of mitochondrial non-coding RNAs (ncRNAs) is modulated by high risk human papillomavirus (HPV) oncogenes. J. Biol. Chem. 2012, 287, 21303–21315. [Google Scholar] [CrossRef] [PubMed]
  117. Lobos-Gonzalez, L.; Silva, V.; Araya, M.; Restovic, F.; Echenique, J.; Oliveira-Cruz, L.; Fitzpatrick, C.; Briones, M.; Villegas, J.; Villota, C.; et al. Targeting antisense mitochondrial ncRNAs inhibits murine melanoma tumor growth and metastasis through reduction in survival and invasion factors. Oncotarget 2016, 7, 58331–58350. [Google Scholar] [CrossRef] [PubMed]
  118. Shepard, P.J.; Choi, E.A.; Lu, J.; Flanagan, L.A.; Hertel, K.J.; Shi, Y. Complex and dynamic landscape of RNA polyadenylation revealed by PAS-Seq. RNA 2011, 17, 761–772. [Google Scholar] [CrossRef] [PubMed]
  119. Slomovic, S.; Laufer, D.; Geiger, D.; Schuster, G. Polyadenylation and degradation of human mitochondrial RNA: The prokaryotic past leaves its mark. Mol. Cell. Biol. 2005, 25, 6427–6435. [Google Scholar] [CrossRef] [PubMed]
  120. Le Grice, S.F. “In the beginning”: Initiation of minus strand DNA synthesis in retroviruses and LTR-containing retrotransposons. Biochemistry 2003, 42, 14349–14355. [Google Scholar] [CrossRef] [PubMed]
  121. Simpson, E.B.; Ross, S.L.; Marchetti, S.E.; Kennell, J.C. Relaxed primer specificity associated with reverse transcriptases encoded by the pFOXC retroplasmids of Fusarium oxysporum. Eukaryot. Cell 2004, 3, 1589–1600. [Google Scholar] [CrossRef] [PubMed]
  122. Yoo, W.; Lim, D.; Kim, S. Compiling Multicopy Single-Stranded DNA Sequences from Bacterial Genome Sequences. Genom. Inform. 2016, 14, 29–33. [Google Scholar] [CrossRef] [PubMed]
  123. Lanchy, J.M.; Isel, C.; Keith, G.; Le Grice, S.F.; Ehresmann, C.; Ehresmann, B.; Marquet, R. Dynamics of the HIV-1 reverse transcription complex during initiation of DNA synthesis. J. Biol. Chem. 2000, 275, 12306–12312. [Google Scholar] [CrossRef] [PubMed]
  124. Isel, C.; Lanchy, J.M.; Le Grice, S.F.; Ehresmann, C.; Ehresmann, B.; Marquet, R. Specific initiation and switch to elongation of human immunodeficiency virus type 1 reverse transcription require the post-transcriptional modifications of primer tRNA3Lys. EMBO J. 1996, 15, 917–924. [Google Scholar] [PubMed]
  125. Marquet, R.; Isel, C.; Ehresmann, C.; Ehresmann, B. tRNAs as primer of reverse transcriptases. Biochimie 1995, 77, 113–124. [Google Scholar] [CrossRef]
  126. Mak, J.; Kleiman, L. Primer tRNAs for reverse transcription. J. Virol. 1997, 71, 8087–8095. [Google Scholar] [PubMed]
  127. Arts, E.J.; Le Grice, S.F. Interaction of retroviral reverse transcriptase with template-primer duplexes during replication. Prog. Nucleic Acid. Res. Mol. Biol. 1998, 58, 339–393. [Google Scholar] [PubMed]
  128. Wakefield, J.K.; Wolf, A.G.; Morrow, C.D. Human immunodeficiency virus type 1 can use different tRNAs as primers for reverse transcription but selectively maintains a primer binding site complementary to tRNA(3Lys). J. Virol. 1995, 69, 6021–6029. [Google Scholar] [PubMed]
  129. Falvey, A.K.; Weiss, G.B.; Krueger, L.J.; Kantor, J.A.; Anderson, W.F. Transcription of single base oligonucleotides by ribonucleic acid-directed deoxyribonucleic acid polymerase. Nucleic Acids Res. 1976, 3, 79–88. [Google Scholar] [CrossRef] [PubMed]
  130. Williams, J.G.; Kubelik, A.R.; Livak, K.J.; Rafalski, J.A.; Tingey, S.V. DNA polymorphisms amplified by arbitrary primers are useful as genetic markers. Nucleic Acids Res. 1990, 18, 6531–6535. [Google Scholar] [CrossRef] [PubMed]
  131. Laurila, M.R.; Salgado, P.S.; Stuart, D.I.; Grimes, J.M.; Bamford, D.H. Back-priming mode of phi6 RNA-dependent RNA polymerase. J. Gen. Virol. 2005, 86, 521–526. [Google Scholar] [CrossRef] [PubMed]
  132. Unneberg, P.; Claverie, J.M. Tentative mapping of transcription-induced interchromosomal interaction using chimeric EST and mRNA data. PLoS ONE 2007, 2, e254. [Google Scholar] [CrossRef] [PubMed]
  133. Bollig-Fischer, A.; Thakur, A.; Sun, Y.; Wu, J.-S.; Liao, D.J. The predominant proteins that react to the MC-20 estrogen receptor alpha antibody differ in molecular weight between the mammary gland and uterus in the mouse and rat. Int. J. Biomed. Sci. 2012, 8, 51–63. [Google Scholar] [PubMed]
  134. Liao, D.J.; Natarajan, G.; Deming, S.L.; Jamerson, M.H.; Johnson, M.; Chepko, G.; Dickson, R.B. Cell cycle basis for the onset and progression of c-Myc-induced, TGFα-enhanced mouse mammary gland carcinogenesis. Oncogene 2000, 19, 1307–1317. [Google Scholar] [CrossRef] [PubMed]
  135. Liao, D.Z.; Pantazis, C.G.; Hou, X.; Li, S.A. Promotion of estrogen-induced mammary gland carcinogenesis by androgen in the male Noble rat: Probable mediation by steroid receptors. Carcinogenesis 1998, 19, 2173–2180. [Google Scholar] [CrossRef] [PubMed]
  136. Sun, Y.; Cao, S.; Yang, M.; Wu, S.; Wang, Z.; Lin, X.; Song, X.; Liao, D.J. Basic anatomy and tumor biology of the RPS6KA6 gene that encodes the p90 ribosomal S6 kinase-4. Oncogene 2013, 32, 1794–1810. [Google Scholar] [CrossRef] [PubMed]
  137. Sun, Y.; Lou, X.; Yang, M.; Yuan, C.; Ma, L.; Xie, B.K.; Wu, J.M.; Yang, W.; Shen, S.X.; Xu, N.; et al. Cyclin-dependent kinase 4 may be expressed as multiple proteins and have functions that are independent of binding to CCND and RB and occur at the S and G 2/M phases of the cell cycle. Cell Cycle 2013, 12, 3512–3525. [Google Scholar] [CrossRef] [PubMed]
  138. Yang, M.; Sun, Y.; Ma, L.; Wang, C.; Wu, J.M.; Bi, A.; Liao, D.J. Complex alternative splicing of the SMARCA2 gene suggests the importance of SMARCA2-B variants. J. Cancer 2011, 2, 386–400. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Depiction of how we define different mature RNAs derived from two neighboring genes within the same genomic locus. (A) Two genes (genes A and B) are transcribed individually to their own RNA transcripts, each being spliced to a mature RNA. This is a cis-splicing procedure; (B) Transcription of the upstream gene (gene A) sometimes may not stop at the annotated site but, instead, may go not only into the possible intergenic region (black box) but also into the downstream gene (gene B) to produce a much longer RNA transcript that is then cis-spliced to a mature RNA, which is considered by us as an RNA of a new, previously unannotated gene (gene C) harbored at this genomic locus; (C) Genes A and B are transcribed to their own transcripts that are trans-spliced to a single mature RNA, which in our opinion is an unadulterated chimera. (A box with an E and a number inside stands for an exon).
Figure 1. Depiction of how we define different mature RNAs derived from two neighboring genes within the same genomic locus. (A) Two genes (genes A and B) are transcribed individually to their own RNA transcripts, each being spliced to a mature RNA. This is a cis-splicing procedure; (B) Transcription of the upstream gene (gene A) sometimes may not stop at the annotated site but, instead, may go not only into the possible intergenic region (black box) but also into the downstream gene (gene B) to produce a much longer RNA transcript that is then cis-spliced to a mature RNA, which is considered by us as an RNA of a new, previously unannotated gene (gene C) harbored at this genomic locus; (C) Genes A and B are transcribed to their own transcripts that are trans-spliced to a single mature RNA, which in our opinion is an unadulterated chimera. (A box with an E and a number inside stands for an exon).
Ijms 18 00714 g001
Figure 2. Three different types of chimeric RNAs classified based on the relationship between the two partner sequences. SHS, short homologous sequence; GAP, an unmatchable sequence.
Figure 2. Three different types of chimeric RNAs classified based on the relationship between the two partner sequences. SHS, short homologous sequence; GAP, an unmatchable sequence.
Ijms 18 00714 g002
Figure 3. Depiction of artificial chimeric cDNA created by hypothetical “consecutive RTs”. In RT (1st RT), an RNA is converted to the first strand of cDNA with its last several Nts coined as “NNNN”. An RNA of another gene may have its first several Nts reversely complementary to these NNNN and thus can anneal to the cDNA end, which allows RT to continue (2nd RT), resulting in an artificial chimeric cDNA (scenario 1). Most RTases append additional Nt or Nts in a non-template manner. For instance, MMLV RTase usually appends CCC or GGG, allowing any RNA with the first Nts being GGG or CCC to anneal to the cDNA end to create a chimeric RNA in the 2nd RT (scenario 2). For detail, see Reference [3]. Arrows point to the 5′-to-3′ direction.
Figure 3. Depiction of artificial chimeric cDNA created by hypothetical “consecutive RTs”. In RT (1st RT), an RNA is converted to the first strand of cDNA with its last several Nts coined as “NNNN”. An RNA of another gene may have its first several Nts reversely complementary to these NNNN and thus can anneal to the cDNA end, which allows RT to continue (2nd RT), resulting in an artificial chimeric cDNA (scenario 1). Most RTases append additional Nt or Nts in a non-template manner. For instance, MMLV RTase usually appends CCC or GGG, allowing any RNA with the first Nts being GGG or CCC to anneal to the cDNA end to create a chimeric RNA in the 2nd RT (scenario 2). For detail, see Reference [3]. Arrows point to the 5′-to-3′ direction.
Ijms 18 00714 g003
Figure 4. Possible formation of spurious “trans-splicing” of sense and antisense transcripts. If an RNA or DNA end has several Nts (say, TGCATG) reversely complementary to a 5′ region (say, CATGCA), it may loop back to anneal to this region and allow RT to continue with the 5′ region as the template. As a result, the cDNA contains not only the sense sequence but also the antisense (in dash arrow) sequence, thus becoming a spurious chimera. Examples can be found in Reference [4].
Figure 4. Possible formation of spurious “trans-splicing” of sense and antisense transcripts. If an RNA or DNA end has several Nts (say, TGCATG) reversely complementary to a 5′ region (say, CATGCA), it may loop back to anneal to this region and allow RT to continue with the 5′ region as the template. As a result, the cDNA contains not only the sense sequence but also the antisense (in dash arrow) sequence, thus becoming a spurious chimera. Examples can be found in Reference [4].
Ijms 18 00714 g004
Figure 5. Illustration of how RNA protection assay, in situ hybridization or Northern blotting may produce a false signal by detecting only one of the two partner sequences. One chimeric RNA contains two-partner-genes’ sequences indicated by a striped bar and a black bar, respectively. In an RNA protection assay, in situ hybridization or Northern blotting, an RNA probe that contains sequences reversely complementary to the two partner-genes’ sequences is used to hybridize with the target chimeric RNA (scenario 1), followed by use of an enzyme to digest away single-stranded excessive probe and irrelevant RNAs. However, in the cells one or both of the two partner genes may also be expressed to RNAs that can also hybridize to part of the probe (scenarios 2 or 3) to form an unwanted hybrid, which is a noise. Since these techniques do not provide sequence information of the hybrid to confirm its identity, there is no way of knowing whether the resultant hybrids belong to the scenario 1, 2 or 3 or to some combination of the three.
Figure 5. Illustration of how RNA protection assay, in situ hybridization or Northern blotting may produce a false signal by detecting only one of the two partner sequences. One chimeric RNA contains two-partner-genes’ sequences indicated by a striped bar and a black bar, respectively. In an RNA protection assay, in situ hybridization or Northern blotting, an RNA probe that contains sequences reversely complementary to the two partner-genes’ sequences is used to hybridize with the target chimeric RNA (scenario 1), followed by use of an enzyme to digest away single-stranded excessive probe and irrelevant RNAs. However, in the cells one or both of the two partner genes may also be expressed to RNAs that can also hybridize to part of the probe (scenarios 2 or 3) to form an unwanted hybrid, which is a noise. Since these techniques do not provide sequence information of the hybrid to confirm its identity, there is no way of knowing whether the resultant hybrids belong to the scenario 1, 2 or 3 or to some combination of the three.
Ijms 18 00714 g005

Share and Cite

MDPI and ACS Style

Yuan, C.; Han, Y.; Zellmer, L.; Yang, W.; Guan, Z.; Yu, W.; Huang, H.; Liao, D.J. It Is Imperative to Establish a Pellucid Definition of Chimeric RNA and to Clear Up a Lot of Confusion in the Relevant Research. Int. J. Mol. Sci. 2017, 18, 714. https://doi.org/10.3390/ijms18040714

AMA Style

Yuan C, Han Y, Zellmer L, Yang W, Guan Z, Yu W, Huang H, Liao DJ. It Is Imperative to Establish a Pellucid Definition of Chimeric RNA and to Clear Up a Lot of Confusion in the Relevant Research. International Journal of Molecular Sciences. 2017; 18(4):714. https://doi.org/10.3390/ijms18040714

Chicago/Turabian Style

Yuan, Chengfu, Yaping Han, Lucas Zellmer, Wenxiu Yang, Zhizhong Guan, Wenfeng Yu, Hai Huang, and D. Joshua Liao. 2017. "It Is Imperative to Establish a Pellucid Definition of Chimeric RNA and to Clear Up a Lot of Confusion in the Relevant Research" International Journal of Molecular Sciences 18, no. 4: 714. https://doi.org/10.3390/ijms18040714

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop