RNA editing

Last updated

RNA editing (also RNA modification) is a molecular process through which some cells can make discrete changes to specific nucleotide sequences within an RNA molecule after it has been generated by RNA polymerase. It occurs in all living organisms and is one of the most evolutionarily conserved properties of RNAs. [1] [2] [3] RNA editing may include the insertion, deletion, and base substitution of nucleotides within the RNA molecule. RNA editing is relatively rare, with common forms of RNA processing (e.g. splicing, 5'-capping, and 3'-polyadenylation) not usually considered as editing. It can affect the activity, localization as well as stability of RNAs, and has been linked with human diseases. [1] [2] [3] [4]

Contents

RNA editing has been observed in some tRNA, rRNA, mRNA, or miRNA molecules of eukaryotes and their viruses, archaea, and prokaryotes. [5] RNA editing occurs in the cell nucleus, as well as within mitochondria and plastids. In vertebrates, editing is rare and usually consists of a small number of changes to the sequence of the affected molecules. In other organisms, such as squids, [6] extensive editing (pan-editing) can occur; in some cases the majority of nucleotides in an mRNA sequence may result from editing. More than 160 types of RNA modifications have been described so far. [7]

RNA-editing processes show great molecular diversity, and some appear to be evolutionarily recent acquisitions that arose independently. The diversity of RNA editing phenomena includes nucleobase modifications such as cytidine (C) to uridine (U) and adenosine (A) to inosine (I) deaminations, as well as non-template nucleotide additions and insertions. RNA editing in mRNAs effectively alters the amino acid sequence of the encoded protein so that it differs from that predicted by the genomic DNA sequence. [8]

The Editosome Complex Editosome.PNG
The Editosome Complex

Detection of RNA editing

Next generation sequencing

To identify diverse post-transcriptional modifications of RNA molecules and determine the transcriptome-wide landscape of RNA modifications by means of next generation RNA sequencing, recently many studies have developed conventional [9] or specialised sequencing methods. [1] [2] [3] Examples of specialised methods are MeRIP-seq, [10] m6A-seq, [11] PA-m5C-seq [12] , methylation-iCLIP, [13] m6A-CLIP, [14] Pseudo-seq, [15] Ψ-seq, [16] CeU-seq, [17] Aza-IP [18] and RiboMeth-seq [19] ). Many of these methods are based on specific capture of the RNA species containing the specific modification, for example through antibody binding coupled with sequencing of the captured reads. After the sequencing these reads are mapped against the whole transcriptome to see where they originate from. [20] Generally with this kind of approach it is possible to see the location of the modifications together with possible identification of some consensus sequences that might help identification and mapping further on. One example of the specialize methods is PA-m5C-seq. This method was further developed from PA-m6A-seq method to identify m5C modifications on mRNA instead of the original target N6-methyladenosine. The easy switch between different modifications as target is made possible with a simple change of the capturing antibody form m6A specific to m5C specific. [12] Application of these methods have identified various modifications (e.g. pseudouridine, m6A, m5C, 2′-O-Me) within coding genes and non-coding genes (e.g. tRNA, lncRNAs, microRNAs) at single nucleotide or very high resolution. [4]

Mass Spectrometry

Mass spectrometry is a way to quantify RNA modifications. [21] More often than not, modifications cause an increase in mass for a given nucleoside. This gives a characteristic readout for the nucleoside and the modified counterpart. [21] Moreover, mass spectrometry allows the investigation of modification dynamics by labelling RNA molecules with stable (non-radioactive) heavy isotopes in vivo. Due to the defined mass increase of heavy isotope labeled nucleosides they can be distinguished from their respective unlabelled isotopomeres by mass spectrometry. This method, called NAIL-MS (nucleic acid isotope labelling coupled mass spectrometry), enables a variety of approaches to investigate RNA modification dynamics. [22] [23] [24]

Types of RNA

Messenger RNA modification

Recently, functional experiments have revealed many novel functional roles of RNA modifications. Most of the RNA modifications are found on transfer-RNA and ribosomal-RNA, but also eukaryotic mRNA has been shown to be modified with multiple different modifications. 17 naturally occurring modifications on mRNA have been identified, from which the N6-methyladenosine is the most abundant and studied. [25] mRNA modifications are linked to many functions in the cell. They ensure the correct maturation and function of the mRNA, but also at the same time act as part of cell's immune system. [26] Certain modifications like 2’O-methylated nucleotides has been associated with cells ability to distinguish own mRNA from foreign RNA. [27] For example, m6A has been predicted to affect protein translation and localization, [1] [2] [3] mRNA stability, [28] alternative polyA choice [14] and stem cell pluripotency. [29] Pseudouridylation of nonsense codons suppresses translation termination both in vitro and in vivo, suggesting that RNA modification may provide a new way to expand the genetic code. [30] 5-methylcytosine on the other hand has been associated with mRNA transport from the nucleus to the cytoplasm and enhancement of translation. These functions of m5C are not fully known and proven but one strong argument towards these functions in the cell is the observed localization of m5C to translation initiation site. [31] Importantly, many modification enzymes are dysregulated and genetically mutated in many disease types. [1] For example, genetic mutations in pseudouridine synthases cause mitochondrial myopathy, sideroblastic anemia (MLASA) [32] and dyskeratosis congenital. [33]

Compared to the modifications identified from other RNA species like tRNA and rRNA, the amount of identified modifications on mRNA is very small. One of the biggest reasons why mRNA modifications are not so well known is missing research techniques. In addition to the lack of identified modifications, the knowledge of associated proteins is also behind other RNA species. Modifications are results of specific enzyme interactions with the RNA molecule. [25] Considering mRNA modifications most of the known related enzymes are the writer enzymes that add the modification on the mRNA. The additional groups of enzymes readers and erasers are for most of the modifications either poorly known of not known at all. [34]  For these reasons there has been during the past decade huge interest in studying these modifications and their function. [20]

Transfer RNA modifications

Transfer RNA or tRNA is the most abundantly modified type of RNA. [35] Modifications in tRNA play crucial roles in maintaining translation efficiency through supporting structure, anticodon-codon interactions, and interactions with enzymes. [36]

Anticodon modifications are important for proper decoding of mRNA. Since the genetic code is degenerate, anticodon modifications are necessary to properly decode mRNA. Particularly, the wobble position of the anticodon determines how the codons are read. For example, in eukaryotes an adenosine at position 34 of the anticodon can be converted to inosine. Inosine is a modification that is able to base-pair with cytosine, adenine, and uridine. [37]

Another commonly modified base in tRNA is the position adjacent to the anticodon. Position 37 is often hypermodified with bulky chemical modifications. These modifications prevent frameshifting and increase anticodon-codon binding stability through stacking interactions. [37]

Ribosomal RNA modification

Ribosomal RNA (rRNA) is essential to the makeup of ribosomes and peptide transfer during translation processes. [38] Ribosomal RNA modifications are made throughout ribosome synthesis, and often occur during and/or after translation. Modifications primarily play a role in the structure of the rRNA in order to protect translational efficiency. [38] Chemical modification in rRNA consists of methylation of ribose sugars, isomerization of uridines, and methylation and acetylation of individual bases. [39]

Methylation

Methylation of rRNA upholds structural rigidity by blocking base pair stacking and surrounds the 2’-OH group to block hydrolysis. It occurs at specific parts of eukaryotic rRNA. The template for methylation consists of 10-21 nucleotides. [38] 2'-O-methylation of the ribose sugar is one of the most common rRNA modifications. [40] Methylation is primarily introduced by small nucleolar RNA's, referred to as snoRNPs. There are two classes of snoRNPs that target methylation sites, and they are referred to box C/D and box H/ACA. [39] [40] One type of methylation, 2′-O-methylation, contributes to helical stabilization. [38]

Isomerization

The isomerization of uridine to pseudoridine is the second most common rRNA modification. These pseudoridines are also introduced by the same classes of snoRNPs that participate in methylation. Psuedouridine synthases are the major participating enzymes in the reaction. [41] The H/ACA box snoRNPs introduce guide sequences that are about 14-15 nucleotides long. [39] Pseudouridylation is triggered in numerous places of rRNAs at once to preserve the thermal stability of RNA. [39] Pseudouridine allows for increased hydrogen bonding and alters translation in rRNA and tRNA. [40] [41] It alters translation by increasing the affinity of the ribosome subunit to specific mRNAs. [38]

Base Editing:

Base editing is the third major class of rRNA modification, specifically in eukaryotes. There are 8 categories of base edits that can occur at the gap between the small and large ribosomal subunits. [38] RNA methyltransferases are the enzymes that introduce base methylation. [38] Acetyltransferases are the enzymes responsible for acetylation of cytosine in rRNA. Base methylation plays a role in translation. These base modifications all work in conjunction with the two other main classes of modification to contribute to RNA structural stability. An example of this occurs in N7-methylation, which increases the nucleotide's charge to increase ionic interactions of proteins attaching to the RNA before translation.

Editing by insertion or deletion

The effect of uracil insertion in pre-mRNA transcripts Insertion.PNG
The effect of uracil insertion in pre-mRNA transcripts

RNA editing through the addition and deletion of uracil has been found in kinetoplasts from the mitochondria of Trypanosoma brucei. [42] Because this may involve a large fraction of the sites in a gene, it is sometimes called "pan-editing" to distinguish it from topical editing of one or a few sites.

Pan-editing starts with the base-pairing of the unedited primary transcript with a guide RNA (gRNA), which contains complementary sequences to the regions around the insertion/deletion points. The newly formed double-stranded region is then enveloped by an editosome, a large multi-protein complex that catalyzes the editing. [43] [44] The editosome opens the transcript at the first mismatched nucleotide and starts inserting uridines. The inserted uridines will base-pair with the guide RNA, and insertion will continue as long as A or G is present in the guide RNA and will stop when a C or U is encountered. [45] [46] The inserted nucleotides cause a frameshift, and result in a translated protein that differs from its gene.

The mechanism of the editosome involves an endonucleolytic cut at the mismatch point between the guide RNA and the unedited transcript. The next step is catalyzed by one of the enzymes in the complex, a terminal U-transferase, which adds Us from UTP at the 3' end of the mRNA. [47] The opened ends are held in place by other proteins in the complex. Another enzyme, a U-specific exoribonuclease, removes the unpaired Us. After editing has made mRNA complementary to gRNA, an RNA ligase rejoins the ends of the edited mRNA transcript. [48] [49] As a consequence, the editosome can edit only in a 3' to 5' direction along the primary RNA transcript. The complex can act on only a single guide RNA at a time. Therefore, a RNA transcript requiring extensive editing will need more than one guide RNA and editosome complex.

Editing by deamination

C-to-U editing

The effect of C-to-U RNA editing on the human ApoB gene Apobgene.PNG
The effect of C-to-U RNA editing on the human ApoB gene

The editing involves cytidine deaminase that deaminates a cytidine base into a uridine base. An example of C-to-U editing is with the apolipoprotein B gene in humans. Apo B100 is expressed in the liver and apo B48 is expressed in the intestines. In the intestines, the mRNA has a CAA sequence edited to be UAA, a stop codon, thus producing the shorter B48 form. C-to-U editing often occurs in the mitochondrial RNA of flowering plants. Different plants have different degrees of C-to-U editing; for example, eight editing events occur in mitochondria of the moss Funaria hygrometrica , whereas over 1,700 editing events occur in the lycophytes Isoetes engelmanii . [50] C-to-U editing is performed by members of the pentatricopeptide repeat (PPR) protein family. Angiosperms have large PPR families, acting as trans -factors for cis -elements lacking a consensus sequence; Arabidopsis has around 450 members in its PPR family. There have been a number of discoveries of PPR proteins in both plastids and mitochondria. [51]

A-to-I editing

Adenosine-to-inosine (A-to-I) modifications contribute to nearly 90% of all editing events in RNA. The deamination of adenosine is catalyzed by the double-stranded RNA-specific adenosine deaminase (ADAR), which typically acts on pre-mRNAs. The deamination of adenosine to inosine disrupts and destabilizes the dsRNA base pairing, therefore rendering that particular dsRNA less able to produce siRNA, which interferes with the RNAi pathway.

The wobble base pairing causes deaminated RNA to have a unique but different structure, which may be related to the inhibition of the initiation step of RNA translation. Studies have shown that I-RNA (RNA with many repeats of the I-U base pair) recruits methylases that are involved in the formation of heterochromatin and that this chemical modification heavily interferes with miRNA target sites. [52] There is active research into the importance of A-to-I modifications and their purpose in the novel concept of epitranscriptomics, in which modifications are made to RNA that alter their function. [53] [54] A long established consequence of A-to-I in mRNA is the interpretation of I as a G, therefore leading to functional A-to-G substitution, e.g. in the interpretation of the genetic code by ribosomes. Newer studies, however, have weakened this correlation by showing that inosines can also be decoded by the ribosome (although in a lesser extent) as adenosines or uracils. Furthermore, it was shown that I's lead to the stalling of ribosomes on the I-rich mRNA. [55]

The development of high-throughput sequencing in recent years has allowed for the development of extensive databases for different modifications and edits of RNA. RADAR (Rigorously Annotated Database of A-to-I RNA editing) was developed in 2013 to catalog the vast variety of A-to-I sites and tissue-specific levels present in humans, mice, and flies. The addition of novel sites and overall edits to the database are ongoing. [56] The level of editing for specific editing sites, e.g. in the filamin A transcript, is tissue-specific. [57] The efficiency of mRNA-splicing is a major factor controlling the level of A-to-I RNA editing. [58] [59] Interestingly, ADAR1 and ADAR2 also affect alternative splicing via both A-to-I editing ability and dsRNA binding ability. [60] [61]

Alternative mRNA editing

Alternative U-to-C mRNA editing was first reported in WT1 (Wilms Tumor-1) transcripts, [62] and non-classic G-A mRNA changes were first observed in HNRNPK (heterogeneous nuclear ribonucleoprotein K) transcripts in both malignant and normal colorectal samples. [63] The latter changes were also later seen alongside non-classic U-to-C alterations in brain cell TPH2 (tryptophan hydroxylase 2) transcripts. [64] Although the reverse amination might be the simplest explanation for U-to-C changes, transamination and transglycosylation mechanisms have been proposed for plant U-to-C editing events in mitochondrial transcripts. [65] A recent study reported novel G-to-A mRNA changes in WT1 transcripts at two hotspots, proposing the APOBEC3A (apolipoprotein B mRNA editing enzyme, catalytic polypeptide 3A) as the enzyme implicated in this class of alternative mRNA editing. [66] It was also shown that alternative mRNA changes were associated with canonical WT1 splicing variants, indicating their functional significance.

RNA editing in plant mitochondria and plastids

It has been shown in previous studies that the only types of RNA editing seen in the plants' mitochondria and plastids are conversion of C-to-U and U-to-C (very rare). [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] RNA-editing sites are found mainly in the coding regions of mRNA, introns, and other non-translated regions. [69] In fact, RNA editing can restore the functionality of tRNA molecules. [71] [72] The editing sites are found primarily upstream of mitochondrial or plastid RNAs. While the specific positions for C to U RNA editing events have been fairly well studied in both the mitochondrion and plastid, [80] the identity and organization of all proteins comprising the editosome have yet to be established. Members of the expansive PPR protein family have been shown to function as trans-acting factors for RNA sequence recognition. [81] Specific members of the MORF (Multiple Organellar RNA editing Factor) family are also required for proper editing at several sites. As some of these MORF proteins have been shown to interact with members of the PPR family, it is possible MORF proteins are components of the editosome complex. [82] An enzyme responsible for the trans- or deamination of the RNA transcript remains elusive, though it has been proposed that the PPR proteins may serve this function as well.

RNA editing is essential for the normal functioning of the plant's translation and respiration activity. Editing can restore the essential base-pairing sequences of tRNAs, restoring functionality. [83] It has also been linked to the production of RNA-edited proteins that are incorporated into the polypeptide complexes of the respiration pathway. Therefore, it is highly probable that polypeptides synthesized from unedited RNAs would not function properly and hinder the activity of both mitochondria and plastids.

C-to-U RNA editing can create start and stop codons, but it cannot destroy existing start and stop codons. A cryptic start codon is created when the codon ACG is edited to be AUG.

Summary of the Various Functions of RNA Editing RNA Editing.png
Summary of the Various Functions of RNA Editing

RNA editing in viruses

Viruses (i.e., measles, mumps, or parainfluenza), especially viruses that have an RNA genome, have been shown to have evolved to utilize RNA modifications in many ways when taking over the host cell. Viruses are known to utilize the RNA modifications in different parts of their infection cycle from immune evasion to protein translation enhancement. [27] RNA editing is used for stability and generation of protein variants. [84] [85] Viral RNAs are transcribed by a virus-encoded RNA-dependent RNA polymerase, which is prone to pausing and "stuttering" at certain nucleotide combinations. In addition, up to several hundred non-templated A's are added by the polymerase at the 3' end of nascent mRNA. [86] These As help stabilize the mRNA. Furthermore, the pausing and stuttering of the RNA polymerase allows the incorporation of one or two Gs or As upstream of the translational codon. [86] The addition of the non-templated nucleotides shifts the reading frame, which generates a different protein.

Additionally, the RNA modifications are shown to have both positive and negative effects on the replication and translation efficiency depending on the virus.  For example, Courtney et al. [12] showed that an RNA modification called 5-methylcytosine is added to the viral mRNA in infected host cells in order to enhance the protein translation of HIV-1 virus. The inhibition of the m5C modification on viral mRNA results in significant reduction in viral protein translation, but interestingly it has no effect on the expression of viral mRNAs in the cell. On the other hand, Lichinchi et al. [87] showed that the N6-methyladenosine modification on ZIKV mRNA inhibits the viral replication.

Origin and Evolution of RNA editing

The RNA-editing system seen in the animal may have evolved from mononucleotide deaminases, which have led to larger gene families that include the apobec-1 and adar genes. These genes share close identity with the bacterial deaminases involved in nucleotide metabolism. The adenosine deaminase of E. coli cannot deaminate a nucleoside in the RNA; the enzyme's reaction pocket is too small for the RNA strand to bind to. However, this active site is widened by amino acid changes in the corresponding human analog genes, APOBEC1 and ADAR , allowing deamination. [88] [89] The gRNA-mediated pan-editing in trypanosome mitochondria, involving templated insertion of U residues, is an entirely different biochemical reaction. The enzymes involved have been shown in other studies to be recruited and adapted from different sources. [43] [90] But the specificity of nucleotide insertion via the interaction between the gRNA and mRNA is similar to the tRNA editing processes in the animal and Acanthamoeba mitochondria. [91] Eukaryotic ribose methylation of rRNAs by guide RNA molecules is a similar form of modification. [92]

Thus, RNA editing evolved more than once. Several adaptive rationales for editing have been suggested. [93] Editing is often described as a mechanism of correction or repair to compensate for defects in gene sequences. However, in the case of gRNA-mediated editing, this explanation does not seem possible because if a defect happens first, there is no way to generate an error-free gRNA-encoding region, which presumably arises by duplication of the original gene region. A more plausible alternative for the evolutionary origins of this system is through constructive neutral evolution, where the order of steps is reversed, with the gratuitous capacity for editing preceding the "defect". [94]

Therapeutic mRNA Editing

Directing edits to correct mutated sequences was first proposed and demonstrated in 1995. [95] This initial work used synthetic RNA antisense oligonucleotides complementary to a pre-mature stop codon mutation in a dystrophin sequence to activate A-to-I editing of the stop codon to a read through codon in a model xenopus cell system. [95] While this also led to nearby inadvertent A-to-I transitions, A to I (read as G) transitions can correct all three stop codons, but cannot create a stop codon. Therefore, the changes led >25% correction of the targeted stop codon with read through to a downstream luciferase reporter sequence. Follow on work by Rosenthal achieved editing of mutated mRNA sequence in mammalian cell culture by directing an oligonucleotide linked to a cytidine deaminase to correct a mutated cystic fibrosis sequence. [96] More recently, CRISPR-Cas13 fused to deaminases has been employed to direct mRNA editing. [97]

In 2022, therapeutic RNA editing for Cas7-11 was reported. [98] [99] It enables sufficiently targeted cuts and an early version of it was used for in vitro editing in 2021. [100]

Comparison to DNA editing

Unlike DNA editing, which is permanent, the effects of RNA editing − including potential off-target mutations in RNA − are transient and are not inherited. RNA editing is therefore considered to be less risky. Furthermore, it may only require a guide RNA by using the ADAR protein already found in humans and many other eukaryotes' cells instead of needing to introduce a foreign protein into the body. [101]

See also

Related Research Articles

<span class="mw-page-title-main">Protein biosynthesis</span> Assembly of proteins inside biological cells

Protein biosynthesis is a core biological process, occurring inside cells, balancing the loss of cellular proteins through the production of new proteins. Proteins perform a number of critical functions as enzymes, structural proteins or hormones. Protein synthesis is a very similar process for both prokaryotes and eukaryotes but there are some distinct differences.

<span class="mw-page-title-main">Gene expression</span> Conversion of a genes sequence into a mature gene product or products

Gene expression is the process by which information from a gene is used in the synthesis of a functional gene product that enables it to produce end products, proteins or non-coding RNA, and ultimately affect a phenotype. These products are often proteins, but in non-protein-coding genes such as transfer RNA (tRNA) and small nuclear RNA (snRNA), the product is a functional non-coding RNA. The process of gene expression is used by all known life—eukaryotes, prokaryotes, and utilized by viruses—to generate the macromolecular machinery for life.

<span class="mw-page-title-main">DNA methyltransferase</span> Class of enzymes

In biochemistry, the DNA methyltransferase family of enzymes catalyze the transfer of a methyl group to DNA. DNA methylation serves a wide variety of biological functions. All the known DNA methyltransferases use S-adenosyl methionine (SAM) as the methyl donor.

<span class="mw-page-title-main">Transfer RNA</span> RNA that facilitates the addition of amino acids to a new protein

Transfer RNA is an adaptor molecule composed of RNA, typically 76 to 90 nucleotides in length, that serves as the physical link between the mRNA and the amino acid sequence of proteins. Transfer RNA (tRNA) does this by carrying an amino acid to the protein-synthesizing machinery of a cell called the ribosome. Complementation of a 3-nucleotide codon in a messenger RNA (mRNA) by a 3-nucleotide anticodon of the tRNA results in protein synthesis based on the mRNA code. As such, tRNAs are a necessary component of translation, the biological synthesis of new proteins in accordance with the genetic code.

<span class="mw-page-title-main">Regulation of gene expression</span> Modifying mechanisms used by cells to increase or decrease the production of specific gene products

Regulation of gene expression, or gene regulation, includes a wide range of mechanisms that are used by cells to increase or decrease the production of specific gene products. Sophisticated programs of gene expression are widely observed in biology, for example to trigger developmental pathways, respond to environmental stimuli, or adapt to new food sources. Virtually any step of gene expression can be modulated, from transcriptional initiation, to RNA processing, and to the post-translational modification of a protein. Often, one gene regulator controls another, and so on, in a gene regulatory network.

Polyadenylation is the addition of a poly(A) tail to an RNA transcript, typically a messenger RNA (mRNA). The poly(A) tail consists of multiple adenosine monophosphates; in other words, it is a stretch of RNA that has only adenine bases. In eukaryotes, polyadenylation is part of the process that produces mature mRNA for translation. In many bacteria, the poly(A) tail promotes degradation of the mRNA. It, therefore, forms part of the larger process of gene expression.

<span class="mw-page-title-main">Pseudouridine</span> Chemical compound

Pseudouridine is an isomer of the nucleoside uridine in which the uracil is attached via a carbon-carbon instead of a nitrogen-carbon glycosidic bond.

<span class="mw-page-title-main">Apolipoprotein B</span> Protein-coding gene in the species Homo sapiens

Apolipoprotein B (ApoB) is a protein that in humans is encoded by the APOB gene. It is commonly used to detect risk of atherosclerotic cardiovascular disease.

Guide RNA (gRNA) or single guide RNA (sgRNA) is a short sequence of RNA that functions as a guide for the Cas9-endonuclease or other Cas-proteins that cut the double-stranded DNA and thereby can be used for gene editing. In bacteria and archaea, gRNAs are a part of the CRISPR-Cas system that serves as an adaptive immune defense that protects the organism from viruses. Here the short gRNAs serve as detectors of foreign DNA and direct the Cas-enzymes that degrades the foreign nucleic acid.

In molecular biology, small nucleolar RNAs (snoRNAs) are a class of small RNA molecules that primarily guide chemical modifications of other RNAs, mainly ribosomal RNAs, transfer RNAs and small nuclear RNAs. There are two main classes of snoRNA, the C/D box snoRNAs, which are associated with methylation, and the H/ACA box snoRNAs, which are associated with pseudouridylation. SnoRNAs are commonly referred to as guide RNAs but should not be confused with the guide RNAs that direct RNA editing in trypanosomes or the guide RNAs (gRNAs) used by Cas9 for CRISPR gene editing.

<span class="mw-page-title-main">Methyltransferase</span> Group of methylating enzymes

Methyltransferases are a large group of enzymes that all methylate their substrates but can be split into several subclasses based on their structural features. The most common class of methyltransferases is class I, all of which contain a Rossmann fold for binding S-Adenosyl methionine (SAM). Class II methyltransferases contain a SET domain, which are exemplified by SET domain histone methyltransferases, and class III methyltransferases, which are membrane associated. Methyltransferases can also be grouped as different types utilizing different substrates in methyl transfer reactions. These types include protein methyltransferases, DNA/RNA methyltransferases, natural product methyltransferases, and non-SAM dependent methyltransferases. SAM is the classical methyl donor for methyltransferases, however, examples of other methyl donors are seen in nature. The general mechanism for methyl transfer is a SN2-like nucleophilic attack where the methionine sulfur serves as the leaving group and the methyl group attached to it acts as the electrophile that transfers the methyl group to the enzyme substrate. SAM is converted to S-Adenosyl homocysteine (SAH) during this process. The breaking of the SAM-methyl bond and the formation of the substrate-methyl bond happen nearly simultaneously. These enzymatic reactions are found in many pathways and are implicated in genetic diseases, cancer, and metabolic diseases. Another type of methyl transfer is the radical S-Adenosyl methionine (SAM) which is the methylation of unactivated carbon atoms in primary metabolites, proteins, lipids, and RNA.

Missense mRNA is a messenger RNA bearing one or more mutated codons that yield polypeptides with an amino acid sequence different from the wild-type or naturally occurring polypeptide. Missense mRNA molecules are created when template DNA strands or the mRNA strands themselves undergo a missense mutation in which a protein coding sequence is mutated and an altered amino acid sequence is coded for.

<span class="mw-page-title-main">ADAR</span> Mammalian protein found in Homo sapiens

The double-stranded RNA-specific adenosine deaminase enzyme family are encoded by the ADAR family genes. ADAR stands for adenosine deaminase acting on RNA. This article focuses on the ADAR proteins; This article details the evolutionary history, structure, function, mechanisms and importance of all proteins within this family.

<span class="mw-page-title-main">APOBEC1</span> Protein-coding gene in the species Homo sapiens

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 also known as C->U-editing enzyme APOBEC-1 is a protein that in humans is encoded by the APOBEC1 gene.

<span class="mw-page-title-main">APOBEC3A</span> Protein-coding gene in the species Homo sapiens

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A, also known as APOBEC3A, or A3A is a gene of the APOBEC3 family found in humans, non-human primates, and some other mammals. It is a single-domain DNA cytidine deaminase with antiviral effects. While other members of the family such as APOBEC3G are believed to act by editing ssDNA by removing an amino group from cytosine in DNA, introducing a cytosine to uracil change which can ultimately lead to a cytosine to thymine mutation, one study suggests that APOBEC3A can inhibit parvoviruses by another mechanism. The cellular function of APOBEC3A is likely to be the destruction of foreign DNA through extensive deamination of cytosine.Stenglein MD, Burns MB, Li M, Lengyel J, Harris RS. "APOBEC3 proteins mediate the clearance of foreign DNA from human cells". Nature Structural & Molecular Biology. 17 (2): 222–9. doi:10.1038/nsmb.1744. PMC 2921484. PMID 20062055.

<span class="mw-page-title-main">APOBEC</span> Enzyme involved in messenger RNA editing

APOBEC is a family of evolutionarily conserved cytidine deaminases.

<i>N</i><sup>6</sup>-Methyladenosine Modification in mRNA, DNA

N6-Methyladenosine (m6A) was originally identified and partially characterised in the 1970s, and is an abundant modification in mRNA and DNA. It is found within some viruses, and most eukaryotes including mammals, insects, plants and yeast. It is also found in tRNA, rRNA, and small nuclear RNA (snRNA) as well as several long non-coding RNA, such as Xist.

Within the field of molecular biology, the epitranscriptome includes all the biochemical modifications of the RNA within a cell. In analogy to epigenetics that describes "functionally relevant changes to the genome that do not involve a change in the nucleotide sequence", epitranscriptomics involves all functionally relevant changes to the transcriptome that do not involve a change in the ribonucleotide sequence. Thus, the epitranscriptome can be defined as the ensemble of such functionally relevant changes.

<span class="mw-page-title-main">Epitranscriptomic sequencing</span>

In epitranscriptomic sequencing, most methods focus on either (1) enrichment and purification of the modified RNA molecules before running on the RNA sequencer, or (2) improving or modifying bioinformatics analysis pipelines to call the modification peaks. Most methods have been adapted and optimized for mRNA molecules, except for modified bisulfite sequencing for profiling 5-methylcytidine which was optimized for tRNAs and rRNAs.

The viral epitranscriptome includes all modifications to viral transcripts, studied by viral epitranscriptomics. Like the more general epitranscriptome, these modifications do not affect the sequence of the transcript, but rather have consequences on subsequent structures and functions.

References

  1. 1 2 3 4 5 Li S, Mason CE (2013). "The pivotal regulatory landscape of RNA modifications". Annual Review of Genomics and Human Genetics. 15: 127–150. doi: 10.1146/annurev-genom-090413-025405 . PMID   24898039.
  2. 1 2 3 4 Song CX, Yi C, He C (November 2012). "Mapping recently identified nucleotide variants in the genome and transcriptome". Nature Biotechnology. 30 (11): 1107–1116. doi:10.1038/nbt.2398. PMC   3537840 . PMID   23138310.
  3. 1 2 3 4 Meyer KD, Jaffrey SR (May 2014). "The dynamic epitranscriptome: N6-methyladenosine and gene expression control". Nature Reviews. Molecular Cell Biology. 15 (5): 313–326. doi:10.1038/nrm3785. PMC   4393108 . PMID   24713629.
  4. 1 2 Sun WJ, Li JH, Liu S, Wu J, Zhou H, Qu LH, Yang JH (January 2016). "RMBase: a resource for decoding the landscape of RNA modifications from high-throughput sequencing data". Nucleic Acids Research. 44 (D1): D259–D265. doi:10.1093/nar/gkv1036. PMC   4702777 . PMID   26464443.
  5. Su AA, Randau L (August 2011). "A-to-I and C-to-U editing within transfer RNAs". Biochemistry. Biokhimiia. 76 (8): 932–937. doi:10.1134/S0006297911080098. PMID   22022967. S2CID   11283810.
  6. "New genetic editing powers discovered in squid". phys.org. Retrieved 2020-04-05.
  7. Boccaletto P, Machnicka MA, Purta E, Piatkowski P, Baginski B, Wirecki TK, et al. (January 2018). "MODOMICS: a database of RNA modification pathways. 2017 update". Nucleic Acids Research. 46 (D1): D303–D307. doi:10.1093/nar/gkx1030. PMC   5753262 . PMID   29106616.
  8. Brennicke A, Marchfelder A, Binder S (June 1999). "RNA editing". FEMS Microbiology Reviews. 23 (3): 297–316. doi: 10.1111/j.1574-6976.1999.tb00401.x . PMID   10371035.
  9. Hoffmann A, Fallmann J, Vilardo E, Mörl M, Stadler PF, Amman F (April 2018). "Accurate mapping of tRNA reads". Bioinformatics. 34 (7). Oxford, England: 1116–1124. doi: 10.1093/bioinformatics/btx756 . PMID   29228294.
  10. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR (June 2012). "Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons". Cell. 149 (7): 1635–1646. doi:10.1016/j.cell.2012.05.003. PMC   3383396 . PMID   22608085.
  11. Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, et al. (April 2012). "Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq". Nature. 485 (7397): 201–206. Bibcode:2012Natur.485..201D. doi:10.1038/nature11112. PMID   22575960. S2CID   3517716.
  12. 1 2 3 Courtney DG, Tsai K, Bogerd HP, Kennedy EM, Law BA, Emery A, et al. (August 2019). "Epitranscriptomic Addition of m5C to HIV-1 Transcripts Regulates Viral Gene Expression". Cell Host & Microbe. 26 (2): 217–227.e6. doi:10.1016/j.chom.2019.07.005. PMC   6714563 . PMID   31415754.
  13. Hussain S, Sajini AA, Blanco S, Dietmann S, Lombard P, Sugimoto Y, et al. (July 2013). "NSun2-mediated cytosine-5 methylation of vault noncoding RNA determines its processing into regulatory small RNAs". Cell Reports. 4 (2): 255–261. doi:10.1016/j.celrep.2013.06.029. PMC   3730056 . PMID   23871666.
  14. 1 2 Ke S, Alemu EA, Mertens C, Gantman EC, Fak JJ, Mele A, et al. (October 2015). "A majority of m6A residues are in the last exons, allowing the potential for 3' UTR regulation". Genes & Development. 29 (19): 2037–2053. doi:10.1101/gad.269415.115. PMC   4604345 . PMID   26404942.
  15. Carlile TM, Rojas-Duran MF, Zinshteyn B, Shin H, Bartoli KM, Gilbert WV (November 2014). "Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells". Nature. 515 (7525): 143–146. Bibcode:2014Natur.515..143C. doi:10.1038/nature13802. PMC   4224642 . PMID   25192136.
  16. Schwartz S, Bernstein DA, Mumbach MR, Jovanovic M, Herbst RH, León-Ricardo BX, et al. (September 2014). "Transcriptome-wide mapping reveals widespread dynamic-regulated pseudouridylation of ncRNA and mRNA". Cell. 159 (1): 148–162. doi:10.1016/j.cell.2014.08.028. PMC   4180118 . PMID   25219674.
  17. Li X, Zhu P, Ma S, Song J, Bai J, Sun F, Yi C (August 2015). "Chemical pulldown reveals dynamic pseudouridylation of the mammalian transcriptome". Nature Chemical Biology. 11 (8): 592–597. doi:10.1038/nchembio.1836. PMID   26075521.
  18. Khoddami V, Cairns BR (May 2013). "Identification of direct targets and modified bases of RNA cytosine methyltransferases". Nature Biotechnology. 31 (5): 458–464. doi:10.1038/nbt.2566. PMC   3791587 . PMID   23604283.
  19. Birkedal U, Christensen-Dalsgaard M, Krogh N, Sabarinathan R, Gorodkin J, Nielsen H (January 2015). "Profiling of ribose methylations in RNA by high-throughput sequencing". Angewandte Chemie. 54 (2): 451–455. doi:10.1002/anie.201408362. PMID   25417815.
  20. 1 2 Chen LQ, Zhao WS, Luo GZ (2020). "Mapping and editing of nucleic acid modifications". Computational and Structural Biotechnology Journal. 18: 661–667. doi:10.1016/j.csbj.2020.03.010. PMC   7113611 . PMID   32257049.
  21. 1 2 Wetzel C, Limbach PA (January 2016). "Mass spectrometry of modified RNAs: recent developments". The Analyst. 141 (1): 16–23. Bibcode:2016Ana...141...16W. doi:10.1039/C5AN01797A. PMC   4679475 . PMID   26501195.
  22. Heiss M, Reichle VF, Kellner S (September 2017). "Observing the fate of tRNA and its modifications by nucleic acid isotope labeling mass spectrometry: NAIL-MS". RNA Biology. 14 (9): 1260–1268. doi:10.1080/15476286.2017.1325063. PMC   5699550 . PMID   28488916.
  23. Reichle VF, Weber V, Kellner S (December 2018). "NAIL-MS in E. coli Determines the Source and Fate of Methylation in tRNA". ChemBioChem. 19 (24): 2575–2583. doi:10.1002/cbic.201800525. PMC   6582434 . PMID   30328661.
  24. Reichle VF, Kaiser S, Heiss M, Hagelskamp F, Borland K, Kellner S (March 2019). "Surpassing limits of static RNA modification analysis with dynamic NAIL-MS". Methods. 156: 91–101. doi: 10.1016/j.ymeth.2018.10.025 . PMID   30395967.
  25. 1 2 McCown PJ, Ruszkowska A, Kunkler CN, Breger K, Hulewicz JP, Wang MC, et al. (September 2020). "Naturally occurring modified ribonucleosides". Wiley Interdisciplinary Reviews. RNA. 11 (5): e1595. doi:10.1002/wrna.1595. PMC   7694415 . PMID   32301288.
  26. Ontiveros RJ, Stoute J, Liu KF (April 2019). "The chemical diversity of RNA modifications". The Biochemical Journal. 476 (8): 1227–1245. doi:10.1042/BCJ20180445. PMC   9073955 . PMID   31028151. S2CID   135425191.
  27. 1 2 Pereira-Montecinos C, Valiente-Echeverría F, Soto-Rifo R (April 2017). "Epitranscriptomic regulation of viral replication". Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 1860 (4): 460–471. doi:10.1016/j.bbagrm.2017.02.002. PMID   28219769.
  28. Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. (January 2014). "N6-methyladenosine-dependent regulation of messenger RNA stability". Nature. 505 (7481): 117–120. Bibcode:2014Natur.505..117W. doi:10.1038/nature12730. PMC   3877715 . PMID   24284625.
  29. Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AA, Kol N, Salmon-Divon M, et al. (February 2015). "Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation". Science. 347 (6225): 1002–1006. doi: 10.1126/science.1261417 . PMID   25569111. S2CID   206562941.
  30. Karijolich J, Yu YT (June 2011). "Converting nonsense codons into sense codons by targeted pseudouridylation". Nature. 474 (7351): 395–398. doi:10.1038/nature10165. PMC   3381908 . PMID   21677757.
  31. Yang X, Yang Y, Sun BF, Chen YS, Xu JW, Lai WY, et al. (May 2017). "5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m5C reader". Cell Research. 27 (5): 606–625. doi:10.1038/cr.2017.55. PMC   5594206 . PMID   28418038.
  32. Bykhovskaya Y, Casas K, Mengesha E, Inbal A, Fischel-Ghodsian N (June 2004). "Missense mutation in pseudouridine synthase 1 (PUS1) causes mitochondrial myopathy and sideroblastic anemia (MLASA)". American Journal of Human Genetics. 74 (6): 1303–1308. doi:10.1086/421530. PMC   1182096 . PMID   15108122.
  33. Heiss NS, Knight SW, Vulliamy TJ, Klauck SM, Wiemann S, Mason PJ, et al. (May 1998). "X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions". Nature Genetics. 19 (1): 32–38. doi:10.1038/ng0598-32. PMID   9590285. S2CID   205342127.
  34. Gilbert WV, Bell TA, Schaening C (June 2016). "Messenger RNA modifications: Form, distribution, and function". Science. 352 (6292): 1408–1412. Bibcode:2016Sci...352.1408G. doi:10.1126/science.aad8711. PMC   5094196 . PMID   27313037.
  35. Kirchner S, Ignatova Z (February 2015). "Emerging roles of tRNA in adaptive translation, signalling dynamics and disease". Nature Reviews. Genetics. 16 (2): 98–112. doi:10.1038/nrg3861. PMID   25534324. S2CID   6727707.
  36. Lorenz C, Lünse CE, Mörl M (April 2017). "tRNA Modifications: Impact on Structure and Thermal Adaptation". Biomolecules. 7 (2): 35. doi: 10.3390/biom7020035 . PMC   5485724 . PMID   28375166.
  37. 1 2 Agris PF, Vendeix FA, Graham WD (February 2007). "tRNA's wobble decoding of the genome: 40 years of modification". Journal of Molecular Biology. 366 (1): 1–13. doi:10.1016/j.jmb.2006.11.046. PMID   17187822.
  38. 1 2 3 4 5 6 7 Sloan KE, Warda AS, Sharma S, Entian KD, Lafontaine DL, Bohnsack MT (September 2017). "Tuning the ribosome: The influence of rRNA modification on eukaryotic ribosome biogenesis and function". RNA Biology. 14 (9): 1138–1152. doi:10.1080/15476286.2016.1259781. PMC   5699541 . PMID   27911188.
  39. 1 2 3 4 Sharma, Sunny; Entian, Karl-Dieter (2022), Entian, Karl-Dieter (ed.), "Chemical Modifications of Ribosomal RNA", Ribosome Biogenesis: Methods and Protocols, Methods in Molecular Biology, vol. 2533, New York, NY: Springer US, pp. 149–166, doi:10.1007/978-1-0716-2501-9_9, ISBN   978-1-0716-2501-9, PMC   9761533 , PMID   35796987 , retrieved 2024-02-29
  40. 1 2 3 Borchardt, Erin K.; Martinez, Nicole M.; Gilbert, Wendy V. (2020-11-23). "Regulation and Function of RNA Pseudouridylation in Human Cells". Annual review of genetics. 54: 309–336. doi:10.1146/annurev-genet-112618-043830. ISSN   0066-4197. PMC   8007080 . PMID   32870730.
  41. 1 2 Borchardt, Erin K.; Martinez, Nicole M.; Gilbert, Wendy V. (2020-11-23). "Regulation and Function of RNA Pseudouridylation in Human Cells". Annual Review of Genetics. 54 (1): 309–336. doi:10.1146/annurev-genet-112618-043830. ISSN   0066-4197. PMC   8007080 .
  42. Benne R (April 1994). "RNA editing in trypanosomes". European Journal of Biochemistry. 221 (1): 9–23. doi: 10.1111/j.1432-1033.1994.tb18710.x . PMID   7513284.
  43. 1 2 Arts GJ, Benne R (June 1996). "Mechanism and evolution of RNA editing in kinetoplastida". Biochimica et Biophysica Acta (BBA) - Gene Structure and Expression. 1307 (1): 39–54. doi:10.1016/0167-4781(96)00021-8. PMID   8652667.
  44. Alfonzo JD, Thiemann O, Simpson L (October 1997). "The mechanism of U insertion/deletion RNA editing in kinetoplastid mitochondria". Nucleic Acids Research. 25 (19): 3751–3759. doi:10.1093/nar/25.19.3751 (inactive 2024-02-22). PMC   146959 . PMID   9380494.{{cite journal}}: CS1 maint: DOI inactive as of February 2024 (link)
  45. Blum B, Bakalara N, Simpson L (January 1990). "A model for RNA editing in kinetoplastid mitochondria: "guide" RNA molecules transcribed from maxicircle DNA provide the edited information". Cell. 60 (2): 189–198. doi:10.1016/0092-8674(90)90735-W. PMID   1688737. S2CID   19656609.
  46. Kable ML, Heidmann S, Stuart KD (May 1997). "RNA editing: getting U into RNA". Trends in Biochemical Sciences. 22 (5): 162–166. doi:10.1016/S0968-0004(97)01041-4. PMID   9175474.
  47. Simpson L, Thiemann OH (June 1995). "Sense from nonsense: RNA editing in mitochondria of kinetoplastid protozoa and slime molds". Cell. 81 (6): 837–840. doi: 10.1016/0092-8674(95)90003-9 . PMID   7781060. S2CID   4634304.
  48. Stuart K (February 1991). "RNA editing in mitochondrial mRNA of trypanosomatids". Trends in Biochemical Sciences. 16 (2): 68–72. doi:10.1016/0968-0004(91)90027-S. PMID   1713359.
  49. Hajduk SL, Sabatini RS (1998). "Mitochondrial mRNA editing in kinetoplastid protozoa". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC.: ASM Press. pp. 377–394.
  50. Takenaka M, Verbitskiy D, Zehrmann A, Härtel B, Bayer-Császár E, Glass F, Brennicke A (November 2014). "RNA editing in plant mitochondria—connecting RNA target sequences and acting proteins". Mitochondrion. Plant Mitochondria in Mitochondrion. 19 (Pt B): 191–197. doi:10.1016/j.mito.2014.04.005. PMID   24732437.
  51. Shikanai T (September 2015). "RNA editing in plants: Machinery and flexibility of site recognition". Biochimica et Biophysica Acta (BBA) - Bioenergetics. SI: Chloroplast Biogenesis. 1847 (9): 779–785. doi: 10.1016/j.bbabio.2014.12.010 . PMID   25585161.
  52. Nishikura K (2010). "Functions and regulation of RNA editing by ADAR deaminases". Annual Review of Biochemistry. 79 (1): 321–349. doi:10.1146/annurev-biochem-060208-105251. PMC   2953425 . PMID   20192758.
  53. Tajaddod M, Jantsch MF, Licht K (March 2016). "The dynamic epitranscriptome: A to I editing modulates genetic information". Chromosoma. 125 (1): 51–63. doi:10.1007/s00412-015-0526-9. PMC   4761006 . PMID   26148686.
  54. Licht K, Jantsch MF (April 2016). "Rapid and dynamic transcriptome regulation by RNA editing and RNA modifications". The Journal of Cell Biology. 213 (1): 15–22. doi:10.1083/jcb.201511041. PMC   4828693 . PMID   27044895.
  55. Licht K, Hartl M, Amman F, Anrather D, Janisiw MP, Jantsch MF (January 2019). "Inosine induces context-dependent recoding and translational stalling". Nucleic Acids Research. 47 (1): 3–14. doi:10.1093/nar/gky1163. PMC   6326813 . PMID   30462291.
  56. Ramaswami G, Li JB (January 2014). "RADAR: a rigorously annotated database of A-to-I RNA editing". Nucleic Acids Research. 42 (Database issue): D109–D113. doi:10.1093/nar/gkt996. PMC   3965033 . PMID   24163250.
  57. Stulić M, Jantsch MF (October 2013). "Spatio-temporal profiling of Filamin A RNA-editing reveals ADAR preferences and high editing levels outside neuronal tissues". RNA Biology. 10 (10): 1611–1617. doi:10.4161/rna.26216. PMC   3866242 . PMID   24025532.
  58. Licht K, Kapoor U, Mayrhofer E, Jantsch MF (July 2016). "Adenosine to Inosine editing frequency controlled by splicing efficiency". Nucleic Acids Research. 44 (13): 6398–6408. doi:10.1093/nar/gkw325. PMC   5291252 . PMID   27112566.
  59. Licht K, Kapoor U, Amman F, Picardi E, Martin D, Bajad P, Jantsch MF (September 2019). "A high resolution A-to-I editing map in the mouse identifies editing events controlled by pre-mRNA splicing". Genome Research. 29 (9): 1453–1463. doi:10.1101/gr.242636.118. PMC   6724681 . PMID   31427386.
  60. Kapoor U, Licht K, Amman F, Jakobi T, Martin D, Dieterich C, Jantsch MF (August 2020). "ADAR-deficiency perturbs the global splicing landscape in mouse tissues". Genome Research. 30 (8): 1107–1118. doi:10.1101/gr.256933.119. PMC   7462079 . PMID   32727871.
  61. Tang SJ, Shen H, An O, Hong H, Li J, Song Y, et al. (February 2020). "Cis- and trans-regulations of pre-mRNA splicing by RNA editing enzymes influence cancer development". Nature Communications. 11 (1): 799. Bibcode:2020NatCo..11..799T. doi:10.1038/s41467-020-14621-5. PMC   7005744 . PMID   32034135.
  62. Sharma PM, Bowman M, Madden SL, Rauscher FJ, Sukumar S (March 1994). "RNA editing in the Wilms' tumor susceptibility gene, WT1". Genes & Development. 8 (6): 720–731. doi: 10.1101/gad.8.6.720 . PMID   7926762.
  63. Klimek-Tomczak K, Mikula M, Dzwonek A, Paziewska A, Karczmarski J, Hennig E, et al. (February 2006). "Editing of hnRNP K protein mRNA in colorectal adenocarcinoma and surrounding mucosa". British Journal of Cancer. 94 (4): 586–592. doi:10.1038/sj.bjc.6602938. PMC   2361188 . PMID   16404425.
  64. Grohmann M, Hammer P, Walther M, Paulmann N, Büttner A, Eisenmenger W, et al. (January 2010). "Alternative splicing and extensive RNA editing of human TPH2 transcripts". PLOS ONE. 5 (1): e8956. Bibcode:2010PLoSO...5.8956G. doi: 10.1371/journal.pone.0008956 . PMC   2813293 . PMID   20126463.
  65. Castandet B, Araya A (August 2011). "RNA editing in plant organelles. Why make it easy?". Biochemistry. Biokhimiia. 76 (8): 924–931. doi:10.1134/S0006297911080086. PMID   22022966. S2CID   2174535.
  66. Niavarani A, Currie E, Reyal Y, Anjos-Afonso F, Horswell S, Griessinger E, et al. (2015). "APOBEC3A is implicated in a novel class of G-to-A mRNA editing in WT1 transcripts". PLOS ONE. 10 (3): e0120089. Bibcode:2015PLoSO..1020089N. doi: 10.1371/journal.pone.0120089 . PMC   4373805 . PMID   25807502.
  67. Covello PS, Gray MW (October 1989). "RNA editing in plant mitochondria". Nature. 341 (6243): 662–666. Bibcode:1989Natur.341..662C. doi:10.1038/341662a0. PMID   2552326. S2CID   4373041.
  68. Gualberto JM, Lamattina L, Bonnard G, Weil JH, Grienenberger JM (October 1989). "RNA editing in wheat mitochondria results in the conservation of protein sequences". Nature. 341 (6243): 660–662. Bibcode:1989Natur.341..660G. doi:10.1038/341660a0. PMID   2552325. S2CID   19402913.
  69. 1 2 Hiesel R, Wissinger B, Schuster W, Brennicke A (December 1989). "RNA editing in plant mitochondria". Science. 246 (4937): 1632–1634. Bibcode:1989Sci...246.1632H. doi:10.1126/science.2480644. PMID   2480644.
  70. Hoch B, Maier RM, Appel K, Igloi GL, Kössel H (September 1991). "Editing of a chloroplast mRNA by creation of an initiation codon". Nature. 353 (6340): 178–180. Bibcode:1991Natur.353..178H. doi:10.1038/353178a0. PMID   1653905. S2CID   4303733.
  71. 1 2 Pring D, Brennicke A, Schuster W (March 1993). "RNA editing gives a new meaning to the genetic information in mitochondria and chloroplasts". Plant Molecular Biology. 21 (6): 1163–1170. doi:10.1007/BF00023611. PMID   8490134. S2CID   30396182.
  72. 1 2 Wissinger B, Brennicke A, Schuster W (September 1992). "Regenerating good sense: RNA editing and trans splicing in plant mitochondria". Trends in Genetics. 8 (9): 322–328. doi:10.1016/0168-9525(92)90265-6. PMID   1365399.
  73. Grienenberger JM (1993). "RNA editing in plant organelles". RNA Editing (Benne, R., Ed.), Ellis Harwood, New York.
  74. Malek O, Lättig K, Hiesel R, Brennicke A, Knoop V (March 1996). "RNA editing in bryophytes and a molecular phylogeny of land plants". The EMBO Journal. 15 (6): 1403–1411. doi:10.1002/j.1460-2075.1996.tb00482.x. PMC   450045 . PMID   8635473.
  75. Freyer R, Kiefer-Meyer MC, Kössel H (June 1997). "Occurrence of plastid RNA editing in all major lineages of land plants". Proceedings of the National Academy of Sciences of the United States of America. 94 (12): 6285–6290. Bibcode:1997PNAS...94.6285F. doi: 10.1073/pnas.94.12.6285 . PMC   21041 . PMID   9177209.
  76. Dietrich A, Small I, Cosset A, Weil JH, Maréchal-Drouard L (1996). "Editing and import: strategies for providing plant mitochondria with a complete set of functional transfer RNAs". Biochimie. 78 (6): 518–529. doi:10.1016/0300-9084(96)84758-4. PMID   8915541.
  77. Bock R, Hermann M, Fuchs M (October 1997). "Identification of critical nucleotide positions for plastid RNA editing site recognition". RNA. 3 (10): 1194–1200. PMC   1369561 . PMID   9326494.
  78. Gray MW, Covello PS (January 1993). "RNA editing in plant mitochondria and chloroplasts". FASEB Journal. 7 (1): 64–71. doi: 10.1096/fasebj.7.1.8422976 . PMID   8422976. S2CID   26005486.
  79. Marchfelder A, Binder S, Brennicke A, Knoop V (1998). "Preface". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC: ASM Press. pp. 307–323.
  80. Takenaka M, Zehrmann A, Verbitskiy D, Härtel B, Brennicke A (2013). "RNA editing in plants and its evolution". Annual Review of Genetics. 47: 335–352. doi:10.1146/annurev-genet-111212-133519. PMID   24274753.
  81. Barkan A, Small I (2014). "Pentatricopeptide repeat proteins in plants". Annual Review of Plant Biology. 65: 415–442. doi: 10.1146/annurev-arplant-050213-040159 . PMID   24471833.
  82. Bentolila S, Oh J, Hanson MR, Bukowski R (June 2013). "Comprehensive high-resolution analysis of the role of an Arabidopsis gene family in RNA editing". PLOS Genetics. 9 (6): e1003584. doi: 10.1371/journal.pgen.1003584 . PMC   3688494 . PMID   23818871.
  83. Price DH, Gray MW (1998). "Editing of tRNA". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC: ASM Press. pp. 289–306.
  84. Curran J, Boeck R, Kolakofsky D (October 1991). "The Sendai virus P gene expresses both an essential protein and an inhibitor of RNA synthesis by shuffling modules via mRNA editing". The EMBO Journal. 10 (10): 3079–3085. doi:10.1002/j.1460-2075.1991.tb07860.x. PMC   453024 . PMID   1655410.
  85. Zheng H, Fu TB, Lazinski D, Taylor J (August 1992). "Editing on the genomic RNA of human hepatitis delta virus". Journal of Virology. 66 (8): 4693–4697. doi:10.1128/jvi.66.8.4693-4697.1992. PMC   241294 . PMID   1629949.
  86. 1 2 Kolakofsky D, Hausmann S (1998). "Chapter 23: Cotranscriptional Paramyxovirus mRNA Editing: a Contradiction in Terms?". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC: ASM Press. pp. 413–420.
  87. Lichinchi G, Zhao BS, Wu Y, Lu Z, Qin Y, He C, Rana TM (November 2016). "Dynamics of Human and Viral RNA Methylation during Zika Virus Infection". Cell Host & Microbe. 20 (5): 666–673. doi:10.1016/j.chom.2016.10.002. PMC   5155635 . PMID   27773536.
  88. Carter CW (1998). "Nucleoside deaminases for cytidine and adenosine: comparisons with deaminases acting on RNA". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC: ASM Press. pp. 363–376.
  89. Navaratnam N, Fujino T, Bayliss J, Jarmuz A, How A, Richardson N, et al. (January 1998). "Escherichia coli cytidine deaminase provides a molecular model for ApoB RNA editing and a mechanism for RNA substrate recognition". Journal of Molecular Biology. 275 (4): 695–714. doi:10.1006/jmbi.1997.1506. PMID   9466941.
  90. Covello PS, Gray MW (August 1993). "On the evolution of RNA editing". Trends in Genetics. 9 (8): 265–268. doi:10.1016/0168-9525(93)90011-6. PMID   8379005.
  91. Lonergan KM, Gray MW (September 1993). "Predicted editing of additional transfer RNAs in Acanthamoeba castellanii mitochondria". Nucleic Acids Research. 21 (18): 4402. doi:10.1093/nar/21.18.4402. PMC   310088 . PMID   8415006.
  92. Bachellerie JP, Cavaille J (1998). "Small nucleolar RNAs guide the ribose methylations of eukaryotic rRNAs". In Grosjean H, Benne R (eds.). Modification and Editing of RNA. Washington, DC: ASM Press. pp. 255–272.
  93. Speijer D (May 2011). "Does constructive neutral evolution play an important role in the origin of cellular complexity? Making sense of the origins and uses of biological complexity". BioEssays. 33 (5): 344–349. doi:10.1002/bies.201100010. PMID   21381061. S2CID   205470421.
  94. Stoltzfus A (August 1999). "On the possibility of constructive neutral evolution". Journal of Molecular Evolution. 49 (2): 169–181. Bibcode:1999JMolE..49..169S. CiteSeerX   10.1.1.466.5042 . doi:10.1007/PL00006540. PMID   10441669. S2CID   1743092.
  95. 1 2 Woolf TM, Chase JM, Stinchcomb DT (August 1995). "Toward the therapeutic editing of mutated RNA sequences". Proceedings of the National Academy of Sciences of the United States of America. 92 (18): 8298–8302. Bibcode:1995PNAS...92.8298W. doi: 10.1073/pnas.92.18.8298 . PMC   41144 . PMID   7545300.
  96. Montiel-Gonzalez MF, Vallecillo-Viejo I, Yudowski GA, Rosenthal JJ (November 2013). "Correction of mutations within the cystic fibrosis transmembrane conductance regulator by site-directed RNA editing". Proceedings of the National Academy of Sciences of the United States of America. 110 (45): 18285–18290. Bibcode:2013PNAS..11018285M. doi: 10.1073/pnas.1306243110 . PMC   3831439 . PMID   24108353.
  97. Cox DB, Gootenberg JS, Abudayyeh OO, Franklin B, Kellner MJ, Joung J, Zhang F (November 2017). "RNA editing with CRISPR-Cas13". Science. 358 (6366): 1019–1027. Bibcode:2017Sci...358.1019C. doi:10.1126/science.aaq0180. PMC   5793859 . PMID   29070703.
  98. Williams S. "Neuroscientists expand CRISPR toolkit with new, compact Cas7-11 enzyme". Massachusetts Institute of Technology. Retrieved 22 June 2022.
  99. Kato K, Zhou W, Okazaki S, Isayama Y, Nishizawa T, Gootenberg JS, et al. (June 2022). "Structure and engineering of the type III-E CRISPR-Cas7-11 effector complex". Cell. 185 (13): 2324–2337.e16. doi: 10.1016/j.cell.2022.05.003 . PMID   35643083. S2CID   249103058.
  100. Özcan A, Krajeski R, Ioannidi E, Lee B, Gardner A, Makarova KS, et al. (September 2021). "Programmable RNA targeting with the single-protein CRISPR effector Cas7-11". Nature. 597 (7878): 720–725. Bibcode:2021Natur.597..720O. doi:10.1038/s41586-021-03886-5. PMID   34489594. S2CID   237432753.
  101. Cross R (25 March 2019). "Watch out, CRISPR. The RNA editing race is on". Chemical & Engineering News. 97 (12). Retrieved 30 September 2020.