Abstract
Background
RNA trans-splicing joins exons from different pre-mRNA transcripts to generate a chimeric product. Trans-splicing can also occur at the protein level, with split inteins mediating the ligation of separate gene products to generate a mature protein.
Sources of data
Comprehensive literature search of published research papers and reviews using Pubmed.
Areas of agreement
Trans-splicing techniques have been used to target a wide range of diseases in both in vitro and in vivo models, resulting in RNA, protein and functional correction.
Areas of controversy
Off-target effects can lead to therapeutically undesirable consequences. In vivo efficacy is typically low, and delivery issues remain a challenge.
Growing points
Trans-splicing provides a promising avenue for developing novel therapeutic approaches. However, much more research needs to be done before developing towards preclinical studies.
Areas timely for developing research
Increasing trans-splicing efficacy and specificity by rational design, screening and competitive inhibition of endogenous cis-splicing.
Keywords: trans-splicing, spliceosome-mediated RNA trans-splicing (SMaRT), ribozyme-mediated trans-splicing, split intein-mediated trans-splicing, genetic disease, infectious disease, cancer, gene therapy
Introduction
In order to become fully functional mature messenger RNA (mRNA), a pre-mRNA molecule needs to undergo various post-transcriptional modifications. One of these is splicing, whereby introns are removed from the pre-mRNA transcript(s) and the remaining exons are joined together. The predominant form of splicing in humans is cis-splicing, in which a single pre-mRNA transcript produces a single mRNA molecule (Fig. 1). However, the spotlight has increasingly been cast on the role of RNA trans-splicing in human health and disease. RNA trans-splicing involves the joining of exons from more than one pre-mRNA transcript to form a single chimeric mRNA molecule (Fig. 1), and was initially proposed as a possible mechanism involved in transcription of trypanosome variant surface glycoprotein.1, 2 It was subsequently observed in in vitro cell-free systems,3, 4 before being shown to occur naturally in trypanosomes,5, 6 Caenorhabditis elegans7 and other lower eukaryotes. Further studies have described trans-splicing in viruses,8–12 bacteriophages13 and prokaryotes,14, 15 suggesting an early evolutionary origin for this form of RNA processing. In vertebrates, RNA trans-splicing occurs far less frequently, but it has been observed in many species including Homo sapiens. In humans, RNA trans-splicing is a physiological phenomenon seen in genes coding for certain enzymes, transcription factors, cancer biomarkers, gene expression regulators and more (reviewed by Lei et al.16).
Fig. 1.

Schematic depiction of different types of pre-mRNA splicing. (a) Cis-splicing. Exons from a single pre-mRNA molecule are ligated together to generate a mature linear transcript. (b) Intergenic trans-splicing. Exons from different genes are joined to produce a chimeric molecule. (c) Intragenic trans-splicing. Exons from different pre-mRNAs of the same gene are spliced together, resulting in exon duplication and sense-antisense fusion. (d) SL trans-splicing. SL exon is spliced onto multiple genes of a polycistronic pre-mRNA, resulting in a number of mature transcripts containing a common 5′ sequence.
RNA trans-splicing can be categorized as intergenic or intragenic, depending on the pre-mRNA source (Fig. 1). In intergenic trans-splicing, exons from separate genes are joined together to produce a chimeric mRNA transcript, while in intragenic trans-splicing, the exons which are spliced together originate from different pre-mRNAs of the same gene (whether they are identical transcripts of the same strand, resulting in exon duplication, or complementary transcripts of different strands, resulting in sense-antisense fusion). Spliced leader (SL) trans-splicing is a special form of trans-splicing, widespread in lower eukaryotes but not yet observed in vertebrates, in which an SL sequence is joined to the 5′-end of multiple pre-mRNAs17 (Fig. 1).
On a post-translational level, polypeptides can undergo protein splicing, where protein sequences with autocatalytic activity (inteins) excise themselves from the precursor polypeptide and ligate the flanking regions (exteins) with a peptide bond. Some inteins—known as split inteins—are derived from two separate genes, one encoding the N-terminus and the other encoding the C-terminus, whose products undergo protein trans-splicing to generate a single mature protein.18
In this article, we review the latest development of RNA and protein trans-splicing technology as potential therapeutic strategies for human diseases.
Trans-splicing therapeutic strategies and techniques
Research has predominantly focused on two main therapeutic strategies: (a) using RNA trans-splicing to repair mutations at the mRNA level, and (b) inducing cell death by introducing trans-splicing constructs encoding toxins, apoptotic factors or suicide genes into target cells. Other therapeutic strategies include using trans-splicing to produce therapeutic proteins and to generate probes for direct molecular imaging of gene expression.
Therapeutic techniques used to carry out these strategies include spliceosome-mediated RNA trans-splicing (SMaRT), ribozyme-mediated trans-splicing and at the protein level, split intein-mediated trans-splicing. First developed by Puttaraju et al.19 in 1999, SMaRT exploits the cell’s own splicing machinery to trans-splice therapeutic coding sequences into endogenous pre-mRNA targets. The therapeutic coding sequences are delivered by artificially engineered pre-mRNA trans-splicing molecules (PTMs). In order to successfully produce the desired product, the trans-splicing reaction must be induced in preference to the cis-splicing reaction, and the specificity of the trans-splicing reaction must be maintained. Thus, in addition to the coding domain, the PTM must also be containing a splicing domain (an artificial intron, containing functional splice sites and spliceosomal recognition sites, which regulates the splicing reaction catalysed by the spliceosome) and a binding domain that hybridizes specifically to an intron within the target pre-mRNA. Only the coding domain is retained in the mature mRNA product.
PTMs can be introduced into the target pre-mRNA by 3′-, 5′- or internal-exon replacement (3′ER, 5′ER or IER, respectively; Fig. 2). 3′ER, the most commonly described SMaRT approach in literature, corrects the distal exons of a target gene by trans-splicing the therapeutic coding sequence downstream of a 5′ splice site. 5′ER was first described in cystic fibrosis (CF) research using the ΔF508 CFTR minigene system20 and involves trans-splicing the coding sequence upstream of a 3′ splice site in the target gene. IER involves a double trans-splicing reaction within the same pre-mRNA target, and as such requires a PTM with two binding domains and two splicing domains to insert the coding sequence in the middle of the target gene. Due to the relative difficulty of designing PTMs for IER and the low repair efficacy of this approach, research into the therapeutic applications of IER has been limited compared to 3′ER and 5′ER, although there have been a few IER studies in minigene21–23 and endogenous RNA systems.24
Fig. 2.

Schematic depiction of different types of exon replacement. * indicates a mutation within the targeted exon.
RNA trans-splicing can also be carried out by non-spliceosomal systems. Group I intron ribozymes have been designed to cleave a target pre-mRNA transcript upstream of the sequences to be replaced, and subsequently catalyse a trans-splicing reaction to attach therapeutic sequences to the 3′ end (reviewed by Lee et al.25). Because ribozyme-mediated RNA trans-splicing is limited to 3′ exon replacement, this is an obvious limitation of this technique compared to SMaRT.
Protein trans-splicing techniques use split inteins to separately deliver gene fragments, which are translated to polypeptides that trans-splice themselves together to reconstitute the full-length protein within target cells (Fig. 3), thus overcoming the limited packaging capacity of gene therapy vectors such as adeno-associated viruses (AAV).26 Split intein-mediated trans-splicing can also be used as a platform for biotechnological development, including protein purification, protein labelling, protein cyclization, protein engineering and design of molecular biosensors.27–29
Fig. 3.

Schematic depiction of protein trans-splicing mediated by split inteins. The DNA sequence coding for the desired product is split into two pieces, and the N-intein and C-intein coding sequences added to each gene fragment. The two genetic constructs are co-delivered to target cells and undergo transcription and translation to produce two precursor polypeptides. The N-intein and C-intein self-assemble, excise themselves out of the polypeptide, and ligate the flanking regions together to generate the mature full-length protein.
Therapeutic applications in disease
Both RNA- (Table 1) and protein- (Table 2) mediated trans-splicing have been explored in a variety of diseases.
Table 1.
Summary of the applications of RNA trans-splicing
| (a) Repairing mutations | ||||
|---|---|---|---|---|
| Application | Disease | Target gene | Mutation | Models in vitro/vivo |
| Repairing mutations | Cancer | p1673 | - | vitro |
| Cancer: BPL | p5375–77 | - | vitro + vivo | |
| CD2278, 79 | - | vitro + vivo | ||
| Cystic fibrosis | CFTR20, 30–33 | Recessive | vitro + vivo | |
| Duchenne muscular dystrophy | Dystrophin22,23,45 | Recessive | vitro + vivo | |
| Dysferlinopathies | DYSF38, 47 | Recessive | vitro + vivo | |
| Dystrophia myotonica type 1 | DMPK40, 41 | Dominant | vitro | |
| Epidermolysis bullosa | COL7A124, 65, 66 COL17A121, 67 Plectin168 Keratin1469, 70 |
Recessive | vitro | |
| Frontotemporal dementia with parkinsonism | Tau-162,63 | Recessive | vitro | |
| Haemophilia A | FVIII49 | Recessive | vitro + vivo | |
| Huntington’s disease | Huntingtin60,61 | Dominant | vitro | |
| Hypertrophic cardiomyopathy | Cardiac myosin-binding protein C55 | Dominant | vitro | |
| GNE myopathy | GNE39 | Recessive | vitro | |
| L-CMD | LMNA46 | Dominant | vitro + vivo | |
| Leber congenital amaurosis type 10, Joubert syndrome | CEP29058, 59 | Recessive | vitro + vivo | |
| Myotonia congenita | ClC-142 | Dominant or recessive | vitro | |
| SCID | DNA-PKcs64 | Recessive | vitro | |
| Sickle cell anaemia, beta-thalassaemia | β-globin50–53 | Recessive | vitro | |
| Spinal muscular atrophy | SMN234–37 | Recessive | vitro + vivo | |
| X-linked immunodeficiency with hyper IgM | CD40L56 | Recessive | vitro | |
| (b) Inducing cell death | ||||
| Application | Target | Models in vitro/vivo | ||
| Inducing cell death | Inducing apoptosis | CHIKV101 | vitro | |
| DENV99,100 | vitro | |||
| Intracellular toxin delivery | HCV97,98 | vitro | ||
| MMP981 | vitro | |||
| Suicide gene therapy (HSV-tk/GCV system) | AFP90 | vitro | ||
| AIMP292 | vitro | |||
| CEA91 | vitro | |||
| Ct-OATP1B396 | vitro + vivo | |||
| hCKAP293 | vitro + vivo | |||
| hTERT25, 83–89 | vitro + vivo | |||
| HIV103 | vitro | |||
| HPV-16102 | vitro | |||
| KRAS94 | vitro + vivo | |||
| SLCO1B395 | vitro + vivo | |||
| (c) Other applications | ||||
| Application | ||||
| Generating therapeutic proteins71, 104, 105 | ||||
| Molecular imaging25,106 | ||||
Table 2.
Summary of the applications of split intein-mediated protein trans-splicing
| Application | Disease | Target gene | Mutation | Models in vitro/vivo |
|---|---|---|---|---|
| Repairing mutations | Amyotrophic lateral sclerosis | SOD143 | Dominant | vitro + vivo |
| Becker muscular dystrophy | Dystrophin44 | Recessive | vitro + vivo | |
| Haemophilia A | FVIII48 | Recessive | vitro | |
| Stargardt disease | ABCA459 | Recessive | vitro + vivo | |
| Biotechnological development 27–29, 107–116 | ||||
| Facilitating delivery of other gene therapies72 | ||||
Genetic diseases
Trans-splicing techniques have been most enthusiastically embraced in the development of therapy for genetic diseases, particularly in the correction of monogenic recessive mutations at the mRNA level, since even partial correction may be able to achieve a heterozygous non-disease phenotype. CF was one of the first genetic diseases targeted using RNA trans-splicing. As a monogenic autosomal recessive disorder whose most common disease-causing mutation, ΔF508 CFTR, has been extensively investigated, CF has proved an attractive target for gene therapy. In in vitro disease models, both 3′ER and 5′ER SMaRT have been found to be effective at correcting the ΔF508 mutation.20, 30–32 Song et al.32 used a segmental RNA trans-splicing approach by co-transfection of two AAV vectors to deliver the 5′ and 3′ halves of CFTR cDNA, which resulted in expression of full-length CFTR mRNA and protein as well as correction of CFTR Cl− conductance. In in vivo disease models, Liu et al.33 found that 3′ER SMaRT corrected CFTR Cl− conductance to 22% of normal function.
Trans-splicing approaches have also been investigated for the treatment of inheritable neuromuscular disease. An optimized 3′ER SMaRT system, where the RNA trans-splicing construct was co-expressed with antisense RNA that competitively inhibited endogenous downstream splice sites and thus increased trans-splicing efficacy, has been used to functionally correct aberrant survival motor neuron-2 (SMN2) expression in mice with spinal muscular atrophy and increase their lifespan.34–37 SMaRT 3′ER has also been used to correct dysferlin (DYSF),38 bifunctional UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE)39 and dystrophin myotonica protein kinase (DMPK)40 mutations. In addition, RNA trans-splicing ribozymes have been developed to target DMPK41 and a mutant canine skeletal muscle chloride channel (ClC-1), which causes myotonia congenita.42 A split intein-based trans-splicing system has been used to deliver the Streptococcus pyogenes Cas9 protein into a mouse model of amyotrophic lateral sclerosis in order to introduce a nonsense-coding substitution in the mutated superoxide dismutase 1 (SOD1) gene, resulting in reduced expression of mutant SOD1 in vivo and improved therapeutic outcomes.43
The dystrophin gene has been targeted using split intein protein trans-splicing in vitro and in vivo in order to treat Becker muscular dystrophy,44 and it has also been targeted using RNA trans-splicing 3′ER and IER to treat Duchenne muscular dystrophy (DMD).22, 23, 45 However, while in vitro dystrophin rescue has been seen,22, 23 only 1% of dystrophin transcripts were repaired in vivo23, and when Koo et al.45 used a novel triple-AAV RNA trans-splicing vector system to deliver the full-length dystrophin coding sequence to skeletal muscle of DMD mice, only low levels of protein were expressed. Similarly, RNA trans-splicing efficacy was extremely low in vivo when using 5′ER SMaRT to target the Lamin A/C (LMNA) gene mutated in autosomal dominant LMNA-related congenital muscular dystrophy (L-CMD).46 While 3′ER SMaRT was effective at repairing DYSF and titin RNA expression in vivo, the corresponding protein was not sufficiently expressed.47 Moreover, undesired proteins were translated from the PTM due to both the presence of a previously unmapped open reading frame (ORF), as well as cis-splicing within the PTM itself giving rise to a number of unexpected gene products with toxic potential.
Further studies demonstrated that haematological disorders could be targeted and repaired by trans-splicing including haemophilia A, sickle cell anaemia (SCA) and beta-thalassaemia. In vitro split intein protein trans-splicing48 and in vivo 3′ER SMaRT49 have been used to correct Factor VIII (FVIII) RNA and protein production. In mouse models, sufficient functional FVIII was produced to correct the haemophilia A phenotype. Ribozyme-mediated RNA trans-splicing50, 51 and 5′ER SMaRT52, 53 have been used to convert the mutated SCA β-globin to anti-sickling γ-globin. However, repair efficacy has been low, perhaps because normal β-globin splicing is very efficient54 so RNA trans-splicing constructs cannot outcompete endogenous splice sites for the splicing machinery. Splicing-impaired targets such as the beta-thalassaemia splice mutants investigated by Kierlin-Duncan and Sullenger52 show higher repair efficiencies of up to 36%.
A variety of other genes and genetic conditions have been investigated using in vivo mouse model SMaRT studies, including cardiac myosin-binding protein C in hypertrophic cardiomyopathy (reviewed by Prondzynski et al.55), CD40L in X-linked immunodeficiency with hyper IgM,56 CEP290 associated with Leber congenital amaurosis type 10 and Joubert syndrome,57 and rhodopsin in autosomal dominant retinitis pigmentosa.58 Moreover, Tornabene et al.59 demonstrated that split intein protein trans-splicing improved CEP290 and ABCA4 expression (associated with Stargardt disease) in vitro and in vivo mouse and pig retina, as well as improving the retinal phenotype of mouse models. They also showed that split intein protein trans-splicing can be used to reconstitute marker proteins in 3D human retinal organoids. Furthermore, in in vitro disease models, aberrant huntingtin in Huntington’s disease has been corrected using 5′ER SMaRT,60, 61 mutant tau-1 in frontotemporal dementia with parkinsonism has been corrected using 3′ER SMaRT,62, 63 and DNA protein kinase, catalytic subunit (DNA-PKcs) deficiency in severe combined immunodeficiency (SCID) has been corrected using 3′ER SMaRT delivered by a non-viral delivery system.64 Moreover, RNA, protein and functional correction of genes involved in epidermolysis bullosa—including collagen type VII (COL7A1),24, 65, 66 collagen type XVII (COL17A1),21, 67 plectin168 and keratin1469, 70—has been achieved in vitro using 3′,65, 67 5’66, 68–70 and IER SMaRT.21, 24
Besides mutation repair, other strategies include using trans-splicing to express therapeutic proteins in vivo. Wang et al.71 demonstrated that therapeutic proteins such as disease-specific antibodies and functional FVIII can be produced by RNA trans-splicing on to highly abundant transcripts, in this case albumin pre-mRNA. Another interesting avenue of research is in exploiting the ability of protein trans-splicing to overcome the packaging limitations of gene therapy vectors: Truong et al.72 investigated a split-Cas9 system, delivered via a recombinant AAV vector, and found that its nuclease activity was reconstituted efficiently in target cells.
Cancer
Since the vast majority of cancers show multifactorial aetiologies and involve mutations in multiple genes, cancer has been a less popular target for trans-splicing techniques compared to genetic disease. Nonetheless, single gene repair has been shown to have some efficacy: mutated tumour suppressors such as p1673 and p53, the latter of which is among the most frequently mutated genes in human cancer,74 have been targeted for correction and restoration of function. Both RNA trans-splicing ribozyme75, 76 and SMART techniques have been used to repair mutant p53 transcripts, with He et al.77 demonstrating that SMART-mediated 3′ER is capable of correcting transcripts and restoring wildtype function in vivo, suppressing the growth of hepatocellular carcinoma xenograft tumours in mice. Beside tumour suppressor defects, SMART-mediated repair of the CD22ΔE12 defect found in paediatric B-precursor leukaemia (BPL) has been shown to reduce in vitro and in vivo clonogenicity of cells derived from BPL patients.78, 79
Another approach is to use trans-splicing to deliver intracellular toxins, thus inducing cancer cell death. Using segmental trans-splicing—a technique in which 5′ and 3′ segments of the pre-mRNA of a toxin gene are encoded in separate vectors, delivered to target cells, and then trans-spliced together by the cell’s own spliceosome to generate a functional toxin—Nakayama et al.80 inhibited tumour growth in vivo by inducing expression of Shigatoxin1A1. Using 3′ER SMART, Gruber et al.81 targeted the matrix metalloproteinase-9 (MMP9) transcript linked to epidermolysis bullosa-associated squamous cell carcinoma, inserting the toxin streptolysin O into the MMP9 gene to trigger cell death.
RNA trans-splicing has also been used in suicide gene therapy, where genes which convert non-toxic compounds into active metabolites are delivered to cancer cells. One such suicide system is the Herpes simplex virus thymidine kinase-ganciclovir (HSV-tk/GCV) system, in which the HSV-tk enzyme catalyses the phosphorylation of the prodrug ganciclovir into an active cytotoxin that causes cell death by DNA replication chain termination and possibly other mechanisms (Fig. 4). To avoid off-target effects, HSV-tk must be specifically expressed only in the cancer cells. This has been achieved by trans-splicing HSV-tk onto mRNA transcripts of genes that are overexpressed in cancer cells, such as the human telomerase reverse transcriptase catalytic subunit (hTERT).82 RNA trans-splicing group I intron ribozymes containing the HSV-tk gene as a 3′ exon have been designed to specifically target hTERT (reviewed by Lee et al.25), with the incorporation of tissue-specific promoters such as the liver-specific phosphoenolpyruvate carboxykinase and apolipoprotein E endowing further specificity in vitro83 and in vivo84–86. Specificity has been further honed by developing hTERT-targeting RNA trans-splicing ribozymes regulated by hypoxia response elements,87 and by liver-specific microRNAs, the latter of which produced efficient hepatocellular carcinoma regression in vivo88. Kim et al.89 reported an hTERT-targeting RNA trans-splicing ribozyme regulated exogenously by theophylline in vitro, pointing to a further level of control over the expression of therapeutic transgenes.
Fig. 4.

Mechanism of action of the HSV-tk/GCV suicide system. HSV-tk is delivered to and specifically expressed in target cells, followed by application of the inactive prodrug ganciclovir (GCV). HSV-tk and other endogenous enzymes phosphorylate GCV to produce the activated drug, which induces DNA replication chain termination and consequently cell death.
Besides hTERT, HSV-tk has been trans-spliced via ribozymes onto a number of other targets, including alpha-fetoprotein (AFP),90 carcinoembryonic antigen (CEA),91 the splicing variant AIMP2-DX2,92 human cytoskeleton-associated protein 2 (hCKAP2) in vivo93 and the KRAS-G12V mutant in vivo94. In addition, SMART has been used in vivo to trans-splice HSV-tk onto SLCO1B3, a marker gene in EB-SCC95, and Ct-OATP1B3, a cancer-specific transcript found in a number of tumours.96
Infectious diseases
Ribozyme-mediated 3′ER has been used to trans-splice the diphtheria toxin (DTA) chain onto hepatitis C (HCV) mRNA and induce apoptosis in transfected human cells.97 In order to specifically target HCV-infected cells, Nawtaisong et al.98 modified apoptosis-inducing genes by replacing their endogenous cleavage sites with the HCV NS5A/5B cleavage recognition site, such that the apoptotic cascade would only be initiated in the presence of HCV protease in infected cells, and trans-spliced the modified constructs onto HCV mRNA using ribozyme-mediated 3′ER. This not only cleaves and destroys the target viral genome, but also induces cytotoxicity in infected cells, thus producing a two-pronged inhibition of viral replication in vitro. However, rapid simultaneous killing of HCV-infected cells may not be therapeutically desirable since it may cause widespread liver cell death.
Group I introns have also been used to target and suppress dengue virus (DENV) replication in mosquito cells by trans-splicing the pro-apoptotic Bax C-terminal domain onto DENV mRNA.99, 100 DENV shares a mosquito vector with the Chikungunya (CHIKV) virus, and cases of co-infection are rising; the same group engineered the DENV-targeting group I ribozyme so that it would also target the highly conserved NS1 region in the CHIKV genome,101 demonstrating for the first time the capability of a single dual-acting ribozyme to trans-splice two different RNA sequences.
Besides its applications in anticancer therapy, the HSV-tk/GCV suicide system has been used to induce selective killing of HPV-16102 and HIV-infected103 human cells in vitro. Ingemarsdotter et al.103 performed extensive bioinformatic analyses to identify target HIV splice sites and design HIV binding domains, and found that trans-splicing constructs targeting HIV donor site D4 for 3′ER and HIV acceptor sites A5, A7 and A8 for 5′ER successfully reduced the viability of HIV-expressing cells in the presence of ganciclovir, with the constructs targeting D4 and A8 being most efficient. Current HIV treatment inhibits viral replication but does not eradicate the viral reservoir that resides latently in memory T cells. Latency does not equate to total transcriptional inactivity for HIV. Thus, RNA trans-splicing techniques that selectively kill HIV-expressing cells may potentially contribute to a cure strategy which targets both actively replicating virus and latently infected cells.
Other therapeutic applications
SMaRT has been used to generate antibody-reporter enzyme fusion proteins104 and to develop modular protein expression systems that allow the production of different antibody formats from the same phage display vector without clone reformatting.105 Both SMaRT and RNA trans-splicing ribozymes have been used as molecular imaging tools, as reviewed by Wally et al.106 and Lee et al.,25 respectively.
Split intein protein trans-splicing has been widely used in biotechnological development, as reviewed by Sarmiento et al.,27 Li28 and Wood et al.29 Protein trans-splicing has been used in protein labelling and ligation, such as in segmental isotopic labelling, where specific segment(s) within a protein are selectively labelled by stable isotopes in order to reduce nuclear magnetic resonance (NMR) signal overlap.107 It has also been used to label proteins within living cells,108–110 as well as in purification of recombinant proteins for research,111–114 and in the purification of toxic proteins.115 Moreover, split inteins have been used as biosensors27, 29 and in targeting AAV vectors to specific cell types.116 Unmodified AAV vectors are not selective, so Muik et al.116 used split intein trans-splicing to couple targeting ligands to AAV capsid protein VP2, resulting in specific gene delivery into target receptor-positive cell types in vitro and in vivo.
Conclusion
Trans-splicing techniques have been used for a wide array of therapeutic applications targeting cancer, genetic and infectious diseases, and demonstrate numerous advantages compared to more traditional therapeutic approaches. Since only the therapeutic sequence needs to be delivered, rather than the full gene, trans-splicing techniques can overcome vector packaging limitations; segmental RNA trans-splicing and intein-mediated protein trans-splicing take this a step further by enabling even smaller gene fragments to be delivered separately and assembled within target cells. Off-target effects are reduced because RNA trans-splicing can only occur in cells where the target pre-mRNA is expressed. By correcting mutated mRNA to wild type, RNA trans-splicing not only induces therapeutic gene expression but also ablates disease-associated gene expression. In addition, it allows endogenous regulation of the transcript to be maintained, which is particularly important when constitutive expression has negative consequences: for example, gene transfer has been successful in correcting CD40L deficiency in HIGM1 model mice, but subsequently resulted in development of lymphoproliferative disease.117 A later trans-splicing study, on the other hand, found no evidence of lymphoproliferative disease after CD40L correction, emphasizing the importance of preserving regulated expression.56
However, there are still some significant limitations. Firstly, non-specific trans-splicing, activation of cryptic splice sites and undesired splicing within the trans-splicing constructs themselves can all lead to off-target effects.47 Furthermore, although functional and phenotypic correction has been achieved in some cases, trans-splicing efficacy in vivo is typically low. This makes it insufficient for repair of dominantly inherited diseases, and risks the escape of tumour cells from anticancer treatment as well as the development of escape mutants when used in antiviral therapy. Some diseases will require sustained expression of repaired mRNA, and in vivo delivery issues are still challenging. The use of viral vectors to deliver RNA trans-splicing constructs may have some limitations, as there are risks of insertional mutagenesis, cytopathic effects and undesired immune responses to the viral vector.118 Non-viral delivery methods that could feasibly be adapted for clinical practice include the biolistic gene gun approach investigated by Peking et al.,66 where non-viral minicircle plasmids expressing 5’ PTM were delivered into murine skin and efficiently corrected mutant COL7A1 in vivo. This could potentially provide a pain-reduced alternative to local intradermal injections, which is of particular relevance to patients with epidermolysis bullosa.
Off-target effects may be reduced by screening trans-splicing constructs to exclude sequence homologies, as well as by more specific delivery to target cells. There are also several strategies that can be pursued to increase trans-splicing efficacy. Screening methods, such as the in vivo rescue assay described by Olson and Müller,119 have been developed to select the most efficient trans-splicing constructs, and in silico techniques have been used to aid rational design of RNA trans-splicing constructs.102, 103 Poddar et al.102 developed bispecific PTMs, targeting multiple disease markers, which increased the efficacy of the HSV/tk-GCV suicide system at triggering cell death, suggesting an avenue for future improvement of RNA trans-splicing activity and specificity. Moreover, antisense oligonucleotides have been used to competitively inhibit endogenous cis-splicing and thus improve trans-splicing efficacy.21, 34–37, 120 Overall, trans-splicing technology is a tool of great therapeutic potential with the capacity to target a diverse spectrum of disease, but there is still much progress to be made before trans-splicing therapy is ready to reach the clinic.
Acknowledgement
This project was supported by funding from the clinical academic reserve (to A.M.L.L.) and Cancer Research UK (Grant number C63549/A25332 to C.K.I.).
Contributor Information
Elizabeth M Hong, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 2QQ, UK.
Carin K Ingemarsdotter, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 2QQ, UK.
Andrew M L Lever, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 2QQ, UK.
Data Availability Statement
No new data were generated or analysed in support of this review.
Conflict of Interest
Authors claim no conflict of interest.
References
- 1. Boothroyd JC, Cross GA. Transcripts coding for variant surface glycoproteins of Trypanosoma brucei have a short, identical exon at their 5′ end. Gene 1982;20:281–9. [DOI] [PubMed] [Google Scholar]
- 2. Van der Ploeg LH, Liu AY, Michels PA et al. RNA splicing is required to make the messenger RNA for a variant surface antigen in trypanosomes. Nucleic Acids Res 1982;10:3591–604. doi: 10.1093/nar/10.12.3591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Konarska MM, Padgett RA, Sharp PA. Trans splicing of mRNA precursors in vitro. Cell 1985;42:165–71. [DOI] [PubMed] [Google Scholar]
- 4. Solnick D. Trans splicing of mRNA precursors. Cell 1985;42:157–64. [DOI] [PubMed] [Google Scholar]
- 5. Murphy WJ, Watkins KP, Agabian N. Identification of a novel Y branch structure as an intermediate in trypanosome mRNA processing: evidence for trans splicing. Cell 1986;47:517–25. [DOI] [PubMed] [Google Scholar]
- 6. Sutton RE, Boothroyd JC. Evidence for trans splicing in trypanosomes. Cell 1986;47:527–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Krause M, Hirsh D. A trans-spliced leader sequence on actin mRNA in C. elegans. Cell 1987;49:753–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Caudevilla C, Da Silva-Azevedo L, Berg B et al. Heterologous HIV-nef mRNA trans-splicing: a new principle how mammalian cells generate hybrid mRNA and protein molecules. FEBS Lett 2001;507:269–79. [DOI] [PubMed] [Google Scholar]
- 9. Kikumori T, Cote GJ, Gagel RF. Naturally occurring heterologous trans-splicing of adenovirus RNA with host cellular transcripts during infection. FEBS Lett 2002;522:41–6. [DOI] [PubMed] [Google Scholar]
- 10. Eul J, Patzel V. Homologous SV40 RNA trans-splicing: a new mechanism for diversification of viral sequences and phenotypes. RNA Biol 2013;10:1689–99. doi: 10.4161/rna.26707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gao Z, Dong Q, Jiang Y et al. Identification and characterization of two novel transcription units of porcine circovirus 2. Virus Genes 2013;47:268–75. doi: 10.1007/s11262-013-0933-z. [DOI] [PubMed] [Google Scholar]
- 12. Sherrill-Mix S, Ocwieja KE, Bushman FD. Gene activity in primary T cells infected with HIV89.6: intron retention and induction of genomic repeats. Retrovirology 2015;12:79. doi: 10.1186/s12977-015-0205-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Galloway Salvo JL, Coetzee T, Belfort M. Deletion-tolerance and trans-splicing of the bacteriophage T4 td intron. Analysis of the P6-L6a region. J Mol Biol 1990;211:537–49. [DOI] [PubMed] [Google Scholar]
- 14. Randau L, Münch R, Hohn MJ et al. Nanoarchaeum equitans creates functional tRNAs from separate genes for their 5′- and 3′-halves. Nature 2005;433:537–41. doi: 10.1038/nature03233. [DOI] [PubMed] [Google Scholar]
- 15. Belhocine K, Mak AB, Cousineau B. Trans-splicing of the Ll.LtrB group II intron in Lactococcus lactis. Nucleic Acids Res 2007;35:2257–68. doi: 10.1093/nar/gkl1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lei Q, Li C, Zuo Z et al. Evolutionary insights into RNA trans-splicing in vertebrates. Genome Biol Evol 2016;8:562–77. doi: 10.1093/gbe/evw025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Bitar M, Boroni M, Macedo AM et al. The spliced leader trans-splicing mechanism in different organisms: molecular details and possible biological roles. Front Genet 2013;4:199. doi: 10.3389/fgene.2013.00199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Mills KV, Johnson MA, Perler FB. Protein splicing: how inteins escape from precursor proteins. J Biol Chem 2014;289:14498–505. doi: 10.1074/jbc.R113.540310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Puttaraju M, Jamison SF, Mansfield SG et al. Spliceosome-mediated RNA trans-splicing as a tool for gene therapy. Nat Biotechnol 1999;17:246–52. doi: 10.1038/6986. [DOI] [PubMed] [Google Scholar]
- 20. Mansfield SG, Clark RH, Puttaraju M et al. 5′ exon replacement and repair by spliceosome-mediated RNA trans-splicing. RNA 2003;9:1290–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Koller U, Wally V, Mitchell LG et al. A novel screening system improves genetic correction by internal exon replacement. Nucleic Acids Res 2011;39:e108. doi: 10.1093/nar/gkr465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lorain S, Peccate C, Le Hir M, Garcia L. Exon exchange approach to repair Duchenne dystrophin transcripts. PLoS ONE 2010;5:e10894. doi: 10.1371/journal.pone.0010894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lorain S, Peccate C, Le Hir M et al. Dystrophin rescue by trans-splicing: a strategy for DMD genotypes not eligible for exon skipping approaches. Nucleic Acids Res 2013;41:8391–402. doi: 10.1093/nar/gkt621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Hüttner C, Murauer EM, Hainzl S et al. Designing efficient double RNA trans-splicing molecules for targeted RNA repair. Int J Mol Sci 2016;17. doi: 10.3390/ijms17101609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lee CH, Han SR, Lee S-W. Therapeutic applications of group I intron-based trans-splicing ribozymes. Wiley Interdiscip Rev RNA 2018;9:e1466. doi: 10.1002/wrna.1466. [DOI] [PubMed] [Google Scholar]
- 26. Duan D, Yue Y, Engelhardt JF. Expanding AAV packaging capacity with trans-splicing or overlapping vectors: a quantitative comparison. Mol Ther 2001;4:383–91. doi: 10.1006/mthe.2001.0456. [DOI] [PubMed] [Google Scholar]
- 27. Sarmiento C, Camarero JA. Biotechnological applications of protein splicing. Curr Protein Pept Sci 2019;20:408–24. doi: 10.2174/1389203720666190208110416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Li Y. Split-inteins and their bioapplications. Biotechnol Lett 2015;37:2121–37. doi: 10.1007/s10529-015-1905-2. [DOI] [PubMed] [Google Scholar]
- 29. Wood DW, Camarero JA. Intein applications: from protein purification and labeling to metabolic control methods. J Biol Chem 2014;289:14512–9. doi: 10.1074/jbc.R114.552653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Mansfield SG, Kole J, Puttaraju M et al. Repair of CFTR mRNA by spliceosome-mediated RNA trans-splicing. Gene Ther 2000;7:1885–95. doi: 10.1038/sj.gt.3301307. [DOI] [PubMed] [Google Scholar]
- 31. Liu X, Luo M, Zhang LN et al. Spliceosome-mediated RNA trans-splicing with recombinant adeno-associated virus partially restores cystic fibrosis transmembrane conductance regulator function to polarized human cystic fibrosis airway epithelial cells. Hum Gene Ther 2005;16:1116–23. doi: 10.1089/hum.2005.16.1116. [DOI] [PubMed] [Google Scholar]
- 32. Song Y, Lou HH, Boyer JL et al. Functional cystic fibrosis transmembrane conductance regulator expression in cystic fibrosis airway epithelial cells by AAV6.2-mediated segmental trans-splicing. Hum Gene Ther 2009;20:267–81. doi: 10.1089/hum.2008.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Liu X, Jiang Q, Mansfield SG et al. Partial correction of endogenous DeltaF508 CFTR in human cystic fibrosis airway epithelia by spliceosome-mediated RNA trans-splicing. Nat Biotechnol 2002;20:47–52. doi: 10.1038/nbt0102-47. [DOI] [PubMed] [Google Scholar]
- 34. Coady TH, Baughan TD, Shababi M et al. Development of a single vector system that enhances trans-splicing of SMN2 transcripts. PLoS ONE. 2008;3:e3468. doi: 10.1371/journal.pone.0003468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Shababi M, Glascock J, Lorson CL. Combination of SMN trans-splicing and a neurotrophic factor increases the life span and body mass in a severe model of spinal muscular atrophy. Hum Gene Ther 2011;22:135–44. doi: 10.1089/hum.2010.114. [DOI] [PubMed] [Google Scholar]
- 36. Coady TH, Lorson CL. Trans-splicing-mediated improvement in a severe mouse model of spinal muscular atrophy. J Neurosci 2010;30:126–30. doi: 10.1523/JNEUROSCI.4489-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Shababi M, Lorson CL. Optimization of SMN trans-splicing through the analysis of SMN introns. J Mol Neurosci 2012;46:459–69. doi: 10.1007/s12031-011-9614-3. [DOI] [PubMed] [Google Scholar]
- 38. Philippi S, Lorain S, Beley C et al. Dysferlin rescue by spliceosome-mediated pre-mRNA trans-splicing targeting introns harbouring weakly defined 3′ splice sites. Hum Mol Genet 2015;24:4049–60. doi: 10.1093/hmg/ddv141. [DOI] [PubMed] [Google Scholar]
- 39. Tal-Goldberg T, Lorain S, Mitrani-Rosenbaum S. Correction of the Middle Eastern M712T mutation causing GNE myopathy by trans-splicing. Neuromolecular Med 2014;16:322–31. doi: 10.1007/s12017-013-8278-2. [DOI] [PubMed] [Google Scholar]
- 40. Chen HY, Kathirvel P, Yee WC, Lai PS. Correction of dystrophia myotonica type 1 pre-mRNA transcripts by artificial trans-splicing. Gene Ther 2009;16:211–7. doi: 10.1038/gt.2008.150. [DOI] [PubMed] [Google Scholar]
- 41. Phylactou LA, Darrah C, Wood MJ. Ribozyme-mediated trans-splicing of a trinucleotide repeat. Nat Genet 1998;18:378–81. doi: 10.1038/ng0498-378. [DOI] [PubMed] [Google Scholar]
- 42. Rogers CS, Vanoye CG, Sullenger BA, George AL. Functional repair of a mutant chloride channel using a trans-splicing ribozyme. J Clin Invest 2002;110:1783–9. doi: 10.1172/JCI16481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lim CKW, Gapinske M, Brooks AK et al. Treatment of a mouse model of ALS by in vivo base editing. Mol Ther 2020;28:1177–89. doi: 10.1016/j.ymthe.2020.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Li J, Sun W, Wang B et al. Protein trans-splicing as a means for viral vector-mediated in vivo gene therapy. Hum Gene Ther 2008;19:958–64. doi: 10.1089/hum.2008.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Koo T, Popplewell L, Athanasopoulos T, Dickson G. Triple trans-splicing adeno-associated virus vectors capable of transferring the coding sequence for full-length dystrophin protein into dystrophic mice. Hum Gene Ther 2014;25:98–108. doi: 10.1089/hum.2013.164. [DOI] [PubMed] [Google Scholar]
- 46. Azibani F, Brull A, Arandel L et al. Gene therapy via trans-splicing for LMNA-related congenital muscular dystrophy. Mol Ther Nucleic Acids 2018;10:376–86. doi: 10.1016/j.omtn.2017.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Monjaret F, Bourg N, Suel L et al. Cis-splicing and translation of the pre-trans-splicing molecule combine with efficiency in spliceosome-mediated RNA trans-splicing. Mol Ther 2014;22:1176–87. doi: 10.1038/mt.2014.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhu F, Liu Z, Chi X, Qu H. Protein trans-splicing based dual-vector delivery of the coagulation factor VIII gene. Sci China Life Sci 2010;53:683–9. doi: 10.1007/s11427-010-4011-7. [DOI] [PubMed] [Google Scholar]
- 49. Chao H, Mansfield SG, Bartel RC et al. Phenotype correction of hemophilia A mice by spliceosome-mediated RNA trans-splicing. Nat Med 2003;9:1015–9. doi: 10.1038/nm900. [DOI] [PubMed] [Google Scholar]
- 50. Lan N, Howrey RP, Lee SW et al. Ribozyme-mediated repair of sickle beta-globin mRNAs in erythrocyte precursors. Science 1998;280:1593–6. doi: 10.1126/science.280.5369.1593. [DOI] [PubMed] [Google Scholar]
- 51. Byun J, Lan N, Long M, Sullenger BA. Efficient and specific repair of sickle beta-globin RNA by trans-splicing ribozymes. RNA 2003;9:1254–63. doi: 10.1261/rna.5450203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kierlin-Duncan MN, Sullenger BA. Using 5’-PTMs to repair mutant beta-globin transcripts. RNA 2007;13:1317–27. doi: 10.1261/rna.525607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Uchida N, Washington KN, Mozer B et al. RNA trans-splicing targeting endogenous β-globin pre-messenger RNA in human erythroid cells. Hum Gene Ther Methods 2017;28:91–9. doi: 10.1089/hgtb.2016.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Audibert A, Weil D, Dautry F. In vivo kinetics of mRNA splicing and transport in mammalian cells. Mol Cell Biol 2002;22:6706–18. doi: 10.1128/mcb.22.19.6706-6718.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Prondzynski M, Mearini G, Carrier L. Gene therapy strategies in the treatment of hypertrophic cardiomyopathy. Pflugers Arch 2019;471:807–15. doi: 10.1007/s00424-018-2173-5. [DOI] [PubMed] [Google Scholar]
- 56. Tahara M, Pergolizzi RG, Kobayashi H et al. Trans-splicing repair of CD40 ligand deficiency results in naturally regulated correction of a mouse model of hyper-IgM X-linked immunodeficiency. Nat Med 2004;10:835–41. doi: 10.1038/nm1086. [DOI] [PubMed] [Google Scholar]
- 57. Dooley SJ, McDougald DS, Fisher KJ et al. Spliceosome-mediated pre-mRNA trans-splicing can repair CEP290 mRNA. Mol Ther Nucleic Acids 2018;12:294–308. doi: 10.1016/j.omtn.2018.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Berger A, Lorain S, Joséphine C et al. Repair of rhodopsin mRNA by spliceosome-mediated RNA trans-splicing: a new approach for autosomal dominant retinitis pigmentosa. Mol Ther 2015;23:918–30. doi: 10.1038/mt.2015.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Tornabene P, Trapani I, Minopoli R et al. Intein-mediated protein trans-splicing expands adeno-associated virus transfer capacity in the retina. Sci Transl Med 2019;11. doi: 10.1126/scitranslmed.aav4523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Rindt H, Yen P-F, Thebeau CN et al. Replacement of huntingtin exon 1 by trans-splicing. Cell Mol Life Sci 2012;69:4191–204. doi: 10.1007/s00018-012-1083-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Rindt H, Tom CM, Lorson CL, Mattis VB. Optimization of trans-splicing for Huntington’s disease RNA therapy. Front Neurosci 2017;11:544. doi: 10.3389/fnins.2017.00544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Rodriguez-Martin T, Garcia-Blanco MA, Mansfield SG et al. Reprogramming of tau alternative splicing by spliceosome-mediated RNA trans-splicing: implications for tauopathies. Proc Natl Acad Sci U S A 2005;102:15659–64. doi: 10.1073/pnas.0503150102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Rodriguez-Martin T, Anthony K, Garcia-Blanco MA et al. Correction of tau mis-splicing caused by FTDP-17 MAPT mutations by spliceosome-mediated RNA trans-splicing. Hum Mol Genet 2009;18:3266–73. doi: 10.1093/hmg/ddp264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zayed H, Xia L, Yerich A et al. Correction of DNA protein kinase deficiency by spliceosome-mediated RNA trans-splicing and sleeping beauty transposon delivery. Mol Ther 2007;15:1273–9. doi: 10.1038/sj.mt.6300178. [DOI] [PubMed] [Google Scholar]
- 65. Murauer EM, Gache Y, Gratz IK et al. Functional correction of type VII collagen expression in dystrophic epidermolysis bullosa. J Invest Dermatol 2011;131:74–83. doi: 10.1038/jid.2010.249. [DOI] [PubMed] [Google Scholar]
- 66. Peking P, Koller U, Hainzl S et al. A gene gun-mediated nonviral RNA trans-splicing strategy for Col7a1 repair. Mol Ther Nucleic Acids. 2016;5:e287. doi: 10.1038/mtna.2016.3. [DOI] [PubMed] [Google Scholar]
- 67. Dallinger G, Puttaraju M, Mitchell LG et al. Development of spliceosome-mediated RNA trans-splicing (SMaRT) for the correction of inherited skin diseases. Exp Dermatol 2003;12:37–46. doi: 10.1034/j.1600-0625.2003.120105.x. [DOI] [PubMed] [Google Scholar]
- 68. Wally V, Klausegger A, Koller U et al. 5′ trans-splicing repair of the PLEC1 gene. J Invest Dermatol 2008;128:568–74. doi: 10.1038/sj.jid.5701152. [DOI] [PubMed] [Google Scholar]
- 69. Wally V, Brunner M, Lettner T et al. K14 mRNA reprogramming for dominant epidermolysis bullosa simplex. Hum Mol Genet 2010;19:4715–25. doi: 10.1093/hmg/ddq405. [DOI] [PubMed] [Google Scholar]
- 70. Peking P, Breitenbach JS, Ablinger M et al. An ex vivo RNA trans-splicing strategy to correct human generalized severe epidermolysis bullosa simplex. Br J Dermatol 2019;180:141–8. doi: 10.1111/bjd.17075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Wang J, Mansfield SG, Cote CA et al. Trans-splicing into highly abundant albumin transcripts for production of therapeutic proteins in vivo. Mol Ther 2009;17:343–51. doi: 10.1038/mt.2008.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Truong D-JJ, Kühner K, Kühn R et al. Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic Acids Res 2015;43:6450–8. doi: 10.1093/nar/gkv601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Kastanos E, Hjiantoniou E, Phylactou LA. Restoration of protein synthesis in pancreatic cancer cells by trans-splicing ribozymes. Biochem Biophys Res Commun 2004;322:930–4. doi: 10.1016/j.bbrc.2004.07.203. [DOI] [PubMed] [Google Scholar]
- 74. Kandoth C, McLellan MD, Vandin F et al. Mutational landscape and significance across 12 major cancer types. Nature 2013;502:333–9. doi: 10.1038/nature12634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Watanabe T, Sullenger BA. Induction of wild-type p53 activity in human cancer cells by ribozymes that repair mutant p53 transcripts. Proc Natl Acad Sci U S A 2000;97:8490–4. doi: 10.1073/pnas.150104097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Shin K-S, Sullenger BA, Lee S-W. Ribozyme-mediated induction of apoptosis in human cancer cells by targeted repair of mutant p53 RNA. Mol Ther 2004;10:365–72. doi: 10.1016/j.ymthe.2004.05.007. [DOI] [PubMed] [Google Scholar]
- 77. He X, Liu F, Yan J et al. Trans-splicing repair of mutant p53 suppresses the growth of hepatocellular carcinoma cells in vitro and in vivo. Sci Rep 2015;5:8705. doi: 10.1038/srep08705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Uckun FM, Qazi S, Ma H et al. CD22ΔE12 as a molecular target for corrective repair using RNA trans-splicing: anti-leukemic activity of a rationally designed RNA trans-splicing molecule. Integr Biol (Camb) 2015;7:237–49. doi: 10.1039/c4ib00221k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Uckun FM, Mitchell LG, Qazi S et al. Development of polypeptide-based nanoparticles for non-viral delivery of CD22 RNA trans-splicing molecule as a new precision medicine candidate against B-lineage ALL. EBioMedicine 2015;2:649–59. doi: 10.1016/j.ebiom.2015.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Nakayama K, Pergolizzi RG, Crystal RG. Gene transfer-mediated pre-mRNA segmental trans-splicing as a strategy to deliver intracellular toxins for cancer therapy. Cancer Res 2005;65:254–63. [PubMed] [Google Scholar]
- 81. Gruber C, Gratz IK, Murauer EM et al. Spliceosome-mediated RNA trans-splicing facilitates targeted delivery of suicide genes to cancer cells. Mol Cancer Ther 2011;10:233–41. doi: 10.1158/1535-7163.MCT-10-0669. [DOI] [PubMed] [Google Scholar]
- 82. Schwaederle M, Krishnamurthy N, Daniels GA et al. Telomerase reverse transcriptase promoter alterations across cancer types as detected by next-generation sequencing: a clinical and molecular analysis of 423 patients. Cancer 2018;124:1288–96. doi: 10.1002/cncr.31175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Song M-S, Lee S-W. Cancer-selective induction of cytotoxicity by tissue-specific expression of targeted trans-splicing ribozyme. FEBS Lett 2006;580:5033–43. doi: 10.1016/j.febslet.2006.08.021. [DOI] [PubMed] [Google Scholar]
- 84. Hong S-H, Jeong J-S, Lee Y-J et al. In vivo reprogramming of hTERT by trans-splicing ribozyme to target tumor cells. Mol Ther 2008;16:74–80. doi: 10.1038/sj.mt.6300282. [DOI] [PubMed] [Google Scholar]
- 85. Song M-S, Jeong J-S, Ban G et al. Validation of tissue-specific promoter-driven tumor-targeting trans-splicing ribozyme system as a multifunctional cancer gene therapy device in vivo. Cancer Gene Ther 2009;16:113–25. doi: 10.1038/cgt.2008.64. [DOI] [PubMed] [Google Scholar]
- 86. Kim Y-H, Kim KT, Lee S-J et al. Image-aided suicide gene therapy utilizing multifunctional hTERT-targeting adenovirus for clinical translation in hepatocellular carcinoma. Theranostics 2016;6:357–68. doi: 10.7150/thno.13621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kim SJ, Lee S-W. Selective expression of transgene using hypoxia-inducible trans-splicing group I intron ribozyme. J Biotechnol 2014;192:22–7. doi: 10.1016/j.jbiotec.2014.10.001. [DOI] [PubMed] [Google Scholar]
- 88. Kim J, Won R, Ban G et al. Targeted regression of hepatocellular carcinoma by cancer-specific RNA replacement through microRNA regulation. Sci Rep 2015;5:12315. doi: 10.1038/srep12315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kim J, Jeong S, Kertsburg A et al. Conditional and target-specific transgene induction through RNA replacement using an allosteric trans-splicing ribozyme. ACS Chem Biol 2014;9:2491–5. doi: 10.1021/cb500567v. [DOI] [PubMed] [Google Scholar]
- 90. Won Y-S, Lee S-W. Targeted retardation of hepatocarcinoma cells by specific replacement of alpha-fetoprotein RNA. J Biotechnol 2007;129:614–9. doi: 10.1016/j.jbiotec.2007.02.004. [DOI] [PubMed] [Google Scholar]
- 91. Jung H-S, Lee S-W. Ribozyme-mediated selective killing of cancer cells expressing carcinoembryonic antigen RNA by targeted trans-splicing. Biochem Biophys Res Commun 2006;349:556–63. doi: 10.1016/j.bbrc.2006.08.073. [DOI] [PubMed] [Google Scholar]
- 92. Won Y-S, Lee S-W. Selective regression of cancer cells expressing a splicing variant of AIMP2 through targeted RNA replacement by trans-splicing ribozyme. J Biotechnol 2012;158:44–9. doi: 10.1016/j.jbiotec.2012.01.006. [DOI] [PubMed] [Google Scholar]
- 93. Ban G, Jeong J-S, Kim A et al. Selective and efficient retardation of cancers expressing cytoskeleton-associated protein 2 by targeted RNA replacement. Int J Cancer 2011;129:1018–29. doi: 10.1002/ijc.25988. [DOI] [PubMed] [Google Scholar]
- 94. Kim SJ, Kim JH, Yang B et al. Specific and efficient regression of cancers harboring KRAS mutation by targeted RNA replacement. Mol Ther 2017;25:356–67. doi: 10.1016/j.ymthe.2016.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Gruber C, Koller U, Murauer EM et al. The design and optimization of RNA trans-splicing molecules for skin cancer therapy. Mol Oncol 2013;7:1056–68. doi: 10.1016/j.molonc.2013.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Sun Y, Piñón Hofbauer J, Harada M et al. Cancer-type organic anion transporting polypeptide 1B3 is a target for cancer suicide gene therapy using RNA trans-splicing technology. Cancer Lett 2018;433:107–16. doi: 10.1016/j.canlet.2018.06.032. [DOI] [PubMed] [Google Scholar]
- 97. Ryu K-J, Kim J-H, Lee S-W. Ribozyme-mediated selective induction of new gene activity in hepatitis C virus internal ribosome entry site-expressing cells by targeted trans-splicing. Mol Ther 2003;7:386–95. doi: 10.1016/s1525-0016(02)00063-1. [DOI] [PubMed] [Google Scholar]
- 98. Nawtaisong P, Fraser ME, Carter JR, Fraser MJ. Trans-splicing group I intron targeting hepatitis C virus IRES mediates cell death upon viral infection in Huh7.5 cells. Virology 2015;481:223–34. doi: 10.1016/j.virol.2015.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Carter JR, Keith JH, Barde PV et al. Targeting of highly conserved dengue virus sequences with anti-dengue virus trans-splicing group I introns. BMC Mol Biol 2010;11:84. doi: 10.1186/1471-2199-11-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Carter JR, Keith JH, Fraser TS et al. Effective suppression of dengue virus using a novel group-I intron that induces apoptotic cell death upon infection through conditional expression of the Bax C-terminal domain. Virol J 2014;11:111. doi: 10.1186/1743-422X-11-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Carter JR, Taylor S, Fraser TS et al. Suppression of the arboviruses dengue and chikungunya using a dual-acting group-I intron coupled with conditional expression of the Bax C-terminal domain. PLoS ONE 2015;10:e0139899. doi: 10.1371/journal.pone.0139899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Poddar S, Loh PS, Ooi ZH et al. RNA structure design improves activity and specificity of trans-splicing-triggered cell death in a suicide gene therapy approach. Mol Ther Nucleic Acids. 2018;11:41–56. doi: 10.1016/j.omtn.2018.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ingemarsdotter CK, Poddar S, Mercier S et al. Expression of herpes simplex virus thymidine kinase/ganciclovir by RNA trans-splicing induces selective killing of HIV-producing cells. Mol Ther Nucleic Acids 2017;7:140–54. doi: 10.1016/j.omtn.2017.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Iwasaki R, Kiuchi H, Ihara M et al. Trans-splicing as a novel method to rapidly produce antibody fusion proteins. Biochem Biophys Res Commun 2009;384:316–21. doi: 10.1016/j.bbrc.2009.04.122. [DOI] [PubMed] [Google Scholar]
- 105. Shang Y, Tesar D, Hötzel I. Modular protein expression by RNA trans-splicing enables flexible expression of antibody formats in mammalian cells from a dual-host phage display vector. Protein Eng Des Sel 2015;28:437–44. doi: 10.1093/protein/gzv018. [DOI] [PubMed] [Google Scholar]
- 106. Wally V, Murauer EM, Bauer JW. Spliceosome-mediated trans-splicing: the therapeutic cut and paste. J Invest Dermatol 2012;132:1959–66. doi: 10.1038/jid.2012.101. [DOI] [PubMed] [Google Scholar]
- 107. Schubeis T, Nagaraj M, Ritter C. Segmental isotope labeling of insoluble proteins for solid-state NMR by protein trans-splicing. Methods Mol Biol 2017;1495:147–60. doi: 10.1007/978-1-4939-6451-2_10. [DOI] [PubMed] [Google Scholar]
- 108. Charalambous A, Andreou M, Skourides PA. Intein-mediated site-specific conjugation of quantum dots to proteins in vivo. J Nanobiotechnology 2009;7:9. doi: 10.1186/1477-3155-7-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Charalambous A, Andreou M, Antoniades I et al. In vivo, site-specific, covalent conjugation of quantum dots to proteins via split-intein splicing. Methods Mol Biol 2012;906:157–69. doi: 10.1007/978-1-61779-953-2_11. [DOI] [PubMed] [Google Scholar]
- 110. Borra R, Dong D, Elnagar AY et al. In-cell fluorescence activation and labeling of proteins mediated by FRET-quenched split inteins. J Am Chem Soc 2012;134:6344–53. doi: 10.1021/ja300209u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Volkmann G, Liu X-Q. Protein C-terminal labeling and biotinylation using synthetic peptide and split-intein. PLoS ONE 2009;4:e8381. doi: 10.1371/journal.pone.0008381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Lu W, Sun Z, Tang Y et al. Split intein facilitated tag affinity purification for recombinant proteins with controllable tag removal by inducible auto-cleavage. J Chromatogr A 2011;1218:2553–60. doi: 10.1016/j.chroma.2011.02.053. [DOI] [PubMed] [Google Scholar]
- 113. Guan D, Ramirez M, Chen Z. Split intein mediated ultra-rapid purification of tagless protein (SIRP). Biotechnol Bioeng 2013;110:2471–81. doi: 10.1002/bit.24913. [DOI] [PubMed] [Google Scholar]
- 114. Shi C, Meng Q, Wood DW. A dual ELP-tagged split intein system for non-chromatographic recombinant protein purification. Appl Microbiol Biotechnol 2013;97:829–35. doi: 10.1007/s00253-012-4601-3. [DOI] [PubMed] [Google Scholar]
- 115. Shi C, Tarimala A, Meng Q, Wood DW. A general purification platform for toxic proteins based on intein trans-splicing. Appl Microbiol Biotechnol 2014;98:9425–35. doi: 10.1007/s00253-014-6080-1. [DOI] [PubMed] [Google Scholar]
- 116. Muik A, Reul J, Friedel T et al. Covalent coupling of high-affinity ligands to the surface of viral vector particles by protein trans-splicing mediates cell type-specific gene transfer. Biomaterials 2017;144:84–94. doi: 10.1016/j.biomaterials.2017.07.032. [DOI] [PubMed] [Google Scholar]
- 117. Brown MP, Topham DJ, Sangster MY et al. Thymic lymphoproliferative disease after successful correction of CD40 ligand deficiency by gene transfer in mice. Nat Med 1998;4:1253–60. doi: 10.1038/3233. [DOI] [PubMed] [Google Scholar]
- 118. Lukashev AN, Zamyatnin AA. Viral vectors for gene therapy: current state and clinical perspectives. Biochemistry Mosc 2016;81:700–8. doi: 10.1134/S0006297916070063. [DOI] [PubMed] [Google Scholar]
- 119. Olson KE, Müller UF. An in vivo selection method to optimize trans-splicing ribozymes. RNA 2012;18:581–9. doi: 10.1261/rna.028472.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Liemberger B, Piñón Hofbauer J, Wally V et al. RNA trans-splicing modulation via antisense molecule interference. Int J Mol Sci 2018;19. doi: 10.3390/ijms19030762. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analysed in support of this review.
