Chimeric enzymes and their applications

Information

  • Patent Grant
  • 11760982
  • Patent Number
    11,760,982
  • Date Filed
    Friday, July 27, 2018
    5 years ago
  • Date Issued
    Tuesday, September 19, 2023
    8 months ago
  • Inventors
  • Original Assignees
  • Examiners
    • Monshipouri; Maryam
    Agents
    • SALIWANCHIK, LLOYD & EISENSCHENK
Abstract
The present invention relates to a chimeric enzyme comprising or consisting of at least one catalytic domain of a capping enzyme and at least one RNA-binding domain of a protein-RNA tethering system as well as its application for the production of an RNA molecule with a 5′-terminal cap.
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a National Stage Application of International Application No. PCT/EP2018/070479, filed Jul. 27, 2018; which claims priority to European Application No. 17306006.2, filed Jul. 27, 2017.


The present invention relates to the field of expression systems, particularly in eukaryotic cells.


In particular, the invention relates to a chimeric enzyme useful for the production of RNA molecules with 5′-terminal cap structures and preferably with a 3′ poly(A) tail.


In the eukaryotes, precursors of messenger RNA (mRNA), i.e. the pre-mRNAs, are synthesized by the RNA polymerase II and then undergoes multiple post-transcriptional modifications, which are required for their biological activities including translation, stability or immune response modulation. Two of these modifications are particularly critical for mRNA metabolism and its translation: the addition of a cap at their 5′-end and a polyadenylation tail at their 3′-end.


The capping is a specialized structure found at the 5′-end of nearly all eukaryotic messenger RNAs. The simplest cap structure, cap-0, results of the addition of a guanine nucleoside methylated at N7 that is joined by 5′-5′ triphosphate bound to the end of primary RNA (i.e. m7GpppN where N is any base, p denotes a phosphate and m a methyl group). In the so called canonical pathway, the formation of the cap-0 involves a series of three enzymatic reactions: RNA triphosphatase (RTPase) removes the γ phosphate residue of 5′ triphosphate end of nascent pre-mRNA to diphosphate, RNA guanylyltransferase (GTase) transfers GMP from GTP to the diphosphate 5′ end of nascent RNA terminus, and RNA N7-guanine methyltransferase (N7-MTase) adds a methyl residue on azote 7 of guanine to the GpppN cap (Furuichi and Shatkin 2000). In higher eukaryotes and some viruses, the 2′-hydroxyl group of the ribose of the first (i.e. cap-1 structures; m7GpppNm2′-OpN) and second (i.e. cap2 structures; m7 GpppNm2′-OpNm2′-O) transcribed nucleotides can be methylated by two separate ribose-2′-O MTases, respectively named cap1- and cap2-specific MTases (Langberg and Moss 1981). However, In contrast to the cellular N7-MTase activity that is exclusively nuclear, cap-1 ribose-2′-O MTase activity has been detected in both the cytoplasm and nucleus of HeLa cells, whereas cap2 MTase activity is exclusively found in their cytoplasm (Langberg and Moss 1981).


The formation of the 5′-terminal m7GpppN cap is the first step of pre-mRNA processing. The m7GpppN cap plays important roles in mRNA stability and its transport from the nucleus to the cytoplasm (Huang and Steitz 2005, Kohler and Hurt 2007). In addition, the 5′-terminal m7GpppN cap is important for the translation of mRNA to protein by anchoring the eukaryotic translation initiation factor 4F (eIF4F) complex, which mediates the recruitment of the 16S portion of the small ribosomal subunit to mRNA (Furuichi, LaFiandra et al. 1977, Gingras, Raught et al. 1999, Rhoads 1999). The 5′-terminal m7GpppN cap therefore enhances drastically the translation of mRNA both in vitro (Lo, Huang et al. 1998), and in cellulo (Malone, Feigner et al. 1989, Gallie 1991, Lo, Huang et al. 1998, Kozak 2005). The cap-0, cap-1 and cap-2 modifications participate in the innate immune response, by distinguishing self from non-self RNA through the RNA sensor RIG-1 and MDAS, which in turn induce an interferon type-I response (Hornung, Ellegast et al. 2006, Daffis, Szretter et al. 2010).


Since they are widely used in the life sciences, biotechnology and medicines, many expression systems have been designed to efficiently produce proteins and/or RNAs particularly in eukaryotic cells.


The inventor has developed in the past an artificial expression system (i.e. a chimeric enzyme) for efficient transgenesis in eukaryotic cells, which autonomously generates mRNA molecules, in particular in the cytoplasm of said cells (WO 2011/128444). Using this system, RNA chains are synthesized by RNA polymerase moiety of this chimeric enzyme and are capped at 5′-end by its mRNA capping enzyme moiety. In addition, a poly(A) tail can be produced at the 3′-end of transcripts by transcription of a polyadenosine track from DNA templates. This system has notably the advantage of not using the endogenous RNA transcription machinery of eukaryotic cells, e.g. RNA polymerase II and associated factors involved in transcription and post-transcription.


Other attempts to couple capping to transcription and thus to improve the translatability of uncapped transcripts produced by the T7 RNA polymerase by fusing the carboxyl-terminal domain (CTD) of the largest subunit of the RNA polymerase II (POLR2A), have to enhance the capping of both constitutively and alternatively spliced substrates in cellulo (Kaneko, Chu et al. 2007, Natalizio, Robson-Dixon et al. 2009). The CTD comprises 25-52 heptapeptide repeats of the consensus sequence 1YSPTSPS7, which is highly conserved throughout evolution and subject to reversible phosphorylation during the transcription cycle (Palancade and Bensaude 2003). When phosphorylated, the CTD is thought to mediate the coupling of transcription and capping of nascent transcripts, by binding one or more subunits of the mRNA capping enzymes in yeast (Cho, Takagi et al. 1997, McCracken, Fong et al. 1997) and mammals (McCracken, Fong et al. 1997, Yue, Maldonado et al. 1997). Noticeably, RNA polymerase II with Ser5-phosphorylated CTD repeats undergoes promoter proximal pausing which is coincident with the co-transcriptional capping of the nascent transcripts (Komarnitsky, Cho et al. 2000, Schroeder, Schwer et al. 2000). However, in contrast to what could be expected intuitively, the fusion of the CTD to the single-unit T7 RNA polymerase is not sufficient to enhance the capping of both constitutively and alternatively spliced substrates in vivo (Kaneko, Chu et al. 2007, Natalizio, Robson-Dixon et al. 2009).


There remains therefore a significant need in the art for new and improved expression systems, in particular in eukaryotic cells, which are appropriate for gene therapy and large-scale protein production without cytotoxicity or induced-cytotoxicity. The present inventor has made a significant step forward with the invention disclosed herein.


The purpose of the invention is to fulfill this need by providing new chimeric enzymes, which make it possible to solve in whole or part the problems mentioned-above.


Unexpectedly, the inventor has notably demonstrated that monomeric or oligomeric chimeric (non-natural) enzymes comprising catalytic domains of a capping enzyme, particularly a catalytic domain of a RNA triphosphatase, a catalytic domain of a guanylyltransferase, a catalytic domain of a N7-guanine methyltransferase, and a RNA-binding domain of a protein-RNA tethering system, said RNA-binding domain binding specifically to a RNA element consisting of a specific RNA sequence and/or structure, allows to highly increase the capping rate of specific mRNAs produced by a RNA polymerase.


These results are surprising since the formation of the cap is a complex process and that the capping of exogenous transcripts cannot be achieved by most other approaches, such as the fusion enzyme CTD-T7 RNA polymerase (Kaneko, Chu et al. 2007, Natalizio, Robson-Dixon et al. 2009).


The U.S. Pat. No. 5,866,680 suggests but without any demonstration the use of the nuclear 70K RNA binding-domain to direct RNA modifying activity to specific site in RNAs including different enzymes such as RNA capping enzymes.


Unexpectedly, the inventor has also demonstrated that cytoplasmic monomeric or oligomeric chimeric enzymes comprising catalytic domains of a capping enzyme and a bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system allows to highly increase the capping rate of specific mRNAs produced by a bacteriophage DNA-dependant RNA polymerase, compared to other RNA-binding domains of protein-RNA tethering systems, such as the U1-RNA 70K (also known as SNRNP70)-U1snRNA system described in said U.S. Pat. No. 5,866,680.


The inventor has also demonstrated that surprisingly monomeric or oligomeric chimeric enzymes comprising catalytic domains of a capping enzyme, a RNA-binding domain of a protein-RNA tethering system and catalytic domain of a poly(A) polymerase, allows to highly increase, synergistically, the capping rate of specific mRNAs produced by RNA polymerase, compared to the combination of a capping enzyme fused to a RNA-binding domain in presence of a poly(A) polymerase fused to a RNA-binding domain. These results are unexpected since capping enzymes and poly(A) polymerases are not physically linked in the nature and contain no known predicted binding domain for a specific RNA sequence. One skilled in the art could have expected to obtain the same expression rate since the components are the same.


Thus, in one aspect, the invention relates to a chimeric enzyme, in particular cytoplasmic, comprising or consisting of:

    • at least one catalytic domain of a capping enzyme, in particular selected in the group consisting of cap-0 canonical capping enzymes, cap-0 non-canonical capping enzymes, cap-1 capping enzymes and cap-2 capping enzymes; and
    • at least one RNA-binding domain of a protein-RNA tethering system wherein said RNA-binding domain binds specifically to a RNA element of said protein-RNA tethering system, consisting of a specific RNA sequence and/or structure.


The chimeric enzyme according to the invention has in particular the following advantages:

    • It increases the rate of mRNAs produced either by an endogenous DNA-dependent RNA polymerase or a non-endogenous DNA-dependent RNA polymerase;
    • It is not expensive, quick and easy to implement and thus appropriate for large-scale assays and protein production.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one skilled in the relevant art.


For convenience, the meaning of certain terms and phrases employed in the specification, examples, and claims are provided.


As used herein, the term “chimeric enzyme” refers to an enzyme that is not a native enzyme found in the nature (that is non-natural). Accordingly, a chimeric enzyme may comprise catalytic domains that are derived from different sources (e.g. from different enzymes) or catalytic domains derived from the same source (e.g. from the same enzyme), but arranged in a different manner than that found in nature.


The term “chimeric enzyme” encompasses monomeric (i.e. single-unit) enzyme but also oligomeric (i.e. multi-unit) enzyme, in particular hetero-oligomeric enzyme.


More specifically, the term “chimeric enzyme” may encompasses an enzyme that comprises or consists of a RNA-binding domain of a protein-RNA tethering system linked covalently or noncovalently with one (i.e. single unit) or several (i.e. multi-unit) catalytic domain(s) (at least one catalytic domain of a capping enzyme) or protein(s) (at least one capping enzyme).


The term “catalytic domain” of an enzyme relates to a protein domain, which is necessary and sufficient, in particular in its three-dimensional structure, to assure the enzymatic function. For example, a catalytic domain of a RNA triphosphatase is the domain, which is necessary and sufficient to assure the RNA triphosphatase function. The term “catalytic domain” encompasses catalytic domain of wild type or mutant enzyme.


The term “protein domain” defines distinct functional and/or structural building blocks and elements in a protein which folds and functions independently.


In particular, the chimeric enzyme according to the invention is a monomeric or oligomeric non-natural enzyme.


As used herein, the term “monomeric enzyme” relates to a single-unit enzyme that consists of only one polypeptide chain.


As used herein, the term “oligomeric enzyme” refers to a multi-unit enzyme that consists of at least two polypeptides chains, linked together covalently or noncovalently. The term “oligomeric enzyme” encompasses a multi-unit enzyme, wherein at least two units of said enzyme are linked together covalently or noncovalently. The term “oligomeric enzyme” encompasses homo-oligomeric enzyme that is a multi-unit enzyme consisting of only one type of monomers (subunits) and hetero-oligomeric enzyme consisting of different types of monomers (subunits).


As used herein, the term “protein-RNA tethering system” refers to a system wherein a protein (or a peptide) recognizes and specifically binds (with high affinity) via its RNA-binding domain to a specific RNA element consisting of a specific RNA sequence and/or structure, therefore making possible to tether this protein (or peptide) with this RNA element. The specific binding between the protein (or the peptide) via its RNA binding domain and the specific RNA element implies that the protein (or peptide) and the specific RNA element interact with high affinity. Interaction with high affinity includes interaction with an affinity of about 10−6 M or stronger, in particular at least 10−7M, at least 10−8M, at least 10−9M and more particularly at least 10−10 M. Whether a RNA-binding domain specifically binds with high affinity to a specific RNA element can be tested readily by, inter alia, comparing the reaction of said RNA-binding domain with a specific RNA element with the reaction of said RNA-binding domain with RNA other than the specific RNA element.


The RNA-protein affinity can be determined by various methods, well known by one skilled in the art. These methods include, but are not limited to, steady-state fluorescence or electrophoretic measurements, RNA electrophoretic mobility shift assay.


As used herein, the term “RNA-binding domain of a protein-RNA tethering system” refers to the domain of a protein (or a peptide) which is necessary and sufficient, in particular in its three-dimensional structure, to assure the recognition and the interaction with high affinity with a specific RNA element consisting of a specific RNA sequence and/or structure, therefore making possible to tether this protein (or peptide) via said RNA-binding domain with this RNA element. Thus, said “RNA-binding domain” (RNA-tethering domain) binds to RNA with sequence and/or structure specificity, i.e. binds specifically to a RNA element consisting of a specific RNA sequence and/or structure. The term “RNA-binding domain of a protein-RNA tethering system” encompasses RNA-biding domain of wild type or mutant protein (or peptide).


The chimeric enzyme according to the invention comprises at least said RNA-binding domain but can further comprise the whole or part of the protein (or peptide) containing said RNA-binding domain. In fact, according to one embodiment of the chimeric enzyme according to the invention, said RNA-binding domain of a protein-RNA tethering system can be included in the whole or part of a protein (or a peptide) of a protein-RNA tethering system.


Some characterized protein-RNA tethering systems include bacteriophage protein-RNA tethering systems such as the MS2 coat protein-RNA tethering system, the R17 coat protein-RNA tethering system and the lambdoid N antitermination protein-RNA tethering systems. The MS2 coat protein and the R17 coat protein recognize and interact with high affinity with specific RNA elements consisting of stem-loop RNA structures (Valegard, Murray et al. 1994, Valegard, Murray et al. 1997). The lambdoid N antitermination protein-RNA tethering systems recognize and interact with high affinity with specific RNA elements consisting of boxBL and boxBR stem loop RNA structures (Das 1993, Greenblatt, Nodwell et al. 1993, Friedman and Court 1995). The bacteriophages characterized so far that belong either to the lambdoid family (i.e. λ, P22, ϕ21, HK97 and 933W viruses) or the MS2-related family (i.e. MS2, and R17).


Other well-characterized protein-RNA tethering systems include: (a) TAT binding domain from the Human immunodeficiency virus-1 (HIV-1; e.g. NCBI reference sequence ABY50660.1) (Dingwall, Ernberg et al. 1990, Weeks, Ampe et al. 1990, Karn, Dingwall et al. 1991, Puglisi, Tan et al. 1992, Frankel and Young 1998) and Bovine Immunodeficiency Virus (Puglisi, Chen et al. 1995), (b) Rev protein from HIV-1 and mutants (e.g. UniProtKB P69718) bind to the Rev-binding element (RBE), a short stem-internal stem-loop structure (Karn, Dingwall et al. 1991, Tan and Frankel 1995, Battiste, Mao et al. 1996, Frankel and Young 1998), (c) Jembrana disease virus Tat protein (UniProtKB Q82854) bind its own TAR protein, as well as TAR proteins from human and bovine immunodeficiency viruses (Smith, Calabro et al. 2000), (d) Iron regulatory proteins, such as the iron-responsive element-binding protein 1 (IREB1, e.g. UniProtKB P21399) and 2 (IREB2, e.g. UniProtKB P48200), which bind iron-responsive elements within 5′UTR or 3′UTR of iron metabolism mRNAs (Theil 1994, Rouault 2006), (e) Brome mosaic virus (BMV) coat protein (UniProtKB Q5KSV1_BMV) binds an hairpin of the MP coding region required for packaging of viral RNA (Sacher and Ahlquist 1989, Choi and Rao 2003), (f) U1A small nuclear ribonucleoprotein subunit 70K (SNRNP70), which binds with high specificity and affinity to a 30-nucleotide RNA hairpin within the 3′UTR of U1snRNA (Keene, Query et al. 1999), (g) SLBP (stem-loop binding protein UniProtKB Q14493) that binds the stem-loop structure in the 3′UTR of histone pre-mRNAs (Marzluff, Wagner et al. 2008), (h) 60S ribosomal protein L7 (e.g. UniProtKB P18124) that binds to G-rich structures in 28S rRNA (Hemmerich, Bosbach et al. 1997), (i) Cowpea chlorotic mottle virus capsid protein (UniProtKB P03601) that binds an hairpin required for packaging of viral RNA (Annamalai, Apte et al. 2005), (j) human T-cell leukemia virus type I (HTLV-1) rex protein and related mutants (e.g. HTLV-1 isolate Caribbea HS-35 subtype A, UniProtKB P0C206), which bind to rex-response element (RxRE) located in the 3′ long terminal repeat of all human T-cell leukemia virus type I-specific mRNAs (Ballaun, Farrington et al. 1991, Jiang, Gorin et al. 1999), (j) HTLV-2 Rex protein (UniProtKB Q85601) that binds 5′ long terminal repeat RNA (Yip, Dynan et al. 1991), (m) RNA splicing component U2 snRNP auxiliary factor U2AF65 (e.g. UniProtKB P26368) that binds a polypyrimidine tract that precedes 3′ splice sites of pre-mRNA (Zamore, Patton et al. 1992, Banerjee, Rahn et al. 2004), (n) bacterial ribosomal protein S7 (UniProtKB P02359) that binds to the lower half of the 3′ major domain of 16S ribosomal RNA (Robert, Gagnon et al. 2000), (o) the archeal L7Ae protein binds to RNA containing a kink-turn with nanomolar affinity (Turner, Melcher et al. 2005, Ye, Yang et al. 2013), (p) RNA binding dormain from the SELENBP2 gene product (Selenium Binding Protein 2, SBP2) binds to the SECIS element in the 3′-UTR of some mRNAs encoding selenoproteins (Mix, Lobanov et al. 2007), (q) the N-terminal domain of Brome mosaic virus (BMV) that can bind a BoxB (Yi, Vaughan et al. 2009), (r) the N-terminus of gp10 head-tail connector which binds ϕ29 pRNA sequence (Xiao, Moll et al. 2005), (s) Streptavidin that binds the artificial RNA aptamer (Leppek and Stoecklin 2014).


In one embodiment of the chimeric enzyme according to the invention, said RNA-binding domain is a bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system, in particular selected in the group consisting of: the MS2 coat protein-RNA tethering system, the R17 coat protein-RNA tethering system and the lambdoid N antitermination protein-RNA tethering systems, more particularly the lambdoid N antitermination protein-RNA tethering systems selected from the group consisting of the lambda N antitermination protein-RNA tethering system, the phi21 N antitermination protein-RNA tethering system, the HK97 N antitermination protein-RNA tethering system and the p22 N antitermination protein-RNA tethering system, and even more particularly the lambda N antitermination protein-RNA tethering system.


Particularly, when the chimeric enzyme of the invention is used to add a 5′-terminal cap to an RNA molecule synthetized by a bacteriophage DNA-dependent polymerase (comprised or not in said chimeric enzyme), said RNA-binding domain is a bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system.


In fact, unexpectedly, the inventor has demonstrated that cytoplasmic monomeric or oligomeric chimeric enzymes comprising catalytic domains of a capping enzyme and a bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system allows to highly increase the capping rate of specific mRNAs produced by a bacteriophage DNA-dependant RNA polymerase, compared to other RNA-binding domains of protein-RNA tethering systems, including the U1-RNA 70K system described in the U.S. Pat. No. 5,866,680.


In particular, said RNA-binding domain is a bacteriophage RNA-binding domain of a bacteriophage protein selected in the group consisting of: the wild type MS2 coat protein (NCBI accession number NC_001417.2, UniProtKB/Swiss-Prot P03612), the wild type R17 coat protein (NCBI accession numbers EF108465.1, UniProtKB/Swiss-Prot P69170) and the wild type lambdoid N antitermination proteins and mutants and derivatives thereof which are able to recognize and interact with high affinity with the specific RNA element, more particularly the wild type lambdoid N antitermination proteins selected from the group consisting of the wild type lambda N antitermination protein (NCBI accession number NC_001416.1, complete genome sequence; UniProtKB/Swiss-Prot accession number P03045), the wild type phi21 N antitermination protein (NCBI accession number AH007390.1, partial genome sequence; UniProtKB/Swiss-Prot accession number P07243), the wild type HK97 N antitermination protein-RNA tethering system (NCBI accession number NC_002167.1, complete genome sequence; NCBI protein accession number NP_037732.1) and the wild type p22 N antitermination protein (particularly NCBI sequence (NCBI accession number NC_002371.2, complete genome sequence; UniProtKB/Swiss-Prot accession number P04891), and even more particularly the wild type lambda N antitermination protein (NCBI accession number NC_001416.1, complete genome sequence; UniProtKB/Swiss-Prot accession number P03045).


The entire or nearly entire MS2/R17 proteins are needed for proficient binding to the tethered RNA, since multiple amino-acid residues spread in these proteins are involved in stem-loop interaction (Valegard, Murray et al. 1994, Valegard, Murray et al. 1997).


So, in one embodiment, the chimeric enzyme of the invention comprises the wild type MS2 coat protein (NCBI accession number NC_001417.2, UniProtKB/Swiss-Prot P03612) or its isolate the wild type R17 coat protein (NCBI accession numbers EF108465.1, UniProtKB/Swiss-Prot P69170), or a mutant or derivative thereof which is able to recognize and interact with high affinity with the specific RNA element.


Importantly, the 18- to 22-amino-acid region from the N-terminal sequences of the lambdoid N-proteins bind to cognate RNA sequences with an affinity and specificity similar to that of the full-length N-proteins (Franklin 1985, Cilley and Williamson 1997).


In one embodiment of the chimeric of the invention, said RNA-binding domain of a protein-RNA tethering system comprising or consisting of the peptide consisting of amino acids at position 1 to 22, in particular 1 to 18 of the wild type lambda N antitermination protein (NCBI accession number NC_001416.1, complete genome sequence; UniProtKB/Swiss-Prot accession number P03045), or of the wild type phi21 N antitermination protein (NCBI accession number AH007390.1, partial genome sequence; UniProtKB/Swiss-Prot accession number P07243), or of the wild type HK97 N antitermination protein-RNA tethering system (NCBI accession number NC_002167.1, complete genome sequence; NCBI protein accession number NP_037732.1) or of the wild type P22 N antitermination protein (NCBI accession number NC_002371.2, complete genome sequence; UniProtKB/Swiss-Prot accession number P04891) or a mutant or derivative thereof which is able to recognize and interact with high affinity with the specific RNA element, particularly of the wild type lambda N antitermination protein (NCBI accession number NC_001416.1, complete genome sequence; UniProtKB/Swiss-Prot accession number P03045).


In particular, said RNA-binding domain of a protein-RNA tethering system comprising or consisting of the peptide consisting of amino acids at position 1 to 22, of the wild type lambda N antitermination protein (NCBI accession number NC_001416.1, complete genome sequence; UniProtKB/Swiss-Prot accession number P03045), in particular SEQ ID No 2, preferably encoded by SEQ ID No 1.


In particular, said RNA-binding domain of a protein-RNA tethering system does not derive from the same source (e.g. from the same enzyme) than the different catalytic domains of the chimeric enzyme of the invention.


The chimeric enzyme according to the invention can be a nuclear enzyme, a subcellular compartment enzyme or a cytoplasmic enzyme. Thus, the chimeric enzyme according to the invention can comprise a signal peptide well known by one skilled in the art, which directs the transport of the enzyme in cells. For example, the chimeric enzyme according to the invention can comprise a nuclear localization signal (NLS), which directs the enzyme to the nucleus. Such NLS is often a unit consisting of five basic, plus-charged amino acids. The NLS can be located anywhere on the peptide chain.


Preferably, the chimeric enzyme according to the invention is a cytoplasmic chimeric enzyme. In particular, it does not comprise signal peptide that directs the transport of the enzyme, except to the cytoplasm.


The cytoplasmic localization of the chimeric enzyme according to the invention has the advantage that it optimizes the levels of transgene expression by avoiding the active transfer of large DNA molecules (i.e. transgene) from the cytoplasm to the nucleus of eukaryotic cells and the export of RNA molecules from the nucleus to the cytoplasm.


These cytoplasmic chimeric enzymes according to the invention can thus be useful to generate a host-independent, eukaryotic gene expression system that is able to work in the cytoplasm in which significantly higher amounts of transfected DNA are usually found as compared to the nucleus.


There is no competition between the endogenous gene transcription and the transgene transcription, since the endogenous gene transcription occurs in the nucleus of eukaryotic cells in contrast to the transgene transcription, which occurs in the cytoplasm.


The cytoplasmic chimeric enzyme according to the invention is thus notably appropriate for large-scale assays and protein production.


As used herein, the term “capping enzyme” refers to any enzyme able to add a m7 GpppG cap at 5′-end of mRNA and/or to modify the ultimate or penultimate bases of a RNA sequence, including cap-0 canonical or non-canonical capping enzymes and cap-1 or cap-2 nucleoside 2′ methyltransferases, N6-methyl-adenosine transferase.


As used herein, the term “cap-0 canonical capping enzymes” refers to enzymes able to add cap-0 structure at the 5′end of RNA molecules by involving a series of three enzymatic reactions: RNA triphosphatase (RTPase) that removes the γ phosphate residue of 5′ triphosphate end of nascent pre-mRNA to diphosphate ppRNA, RNA guanylyltransferase (GTase) that transfers GMP from GTP to the diphosphate ppRNA nascent RNA terminus, and RNA N7-guanine methyltransferase (N7-MTase) that adds a methyl residue on nitrogen 7 of guanine to the GpppRNA cap (Furuichi and Shatkin 2000).


The enzymatic domains of eukaryotic organisms and viruses, which are involved in the canonical formation of cap-0 structure, can be assembled in a variable number of protein subunits:

    • Single subunit capping enzymes with all three critical enzymatic domains, i.e. RTPase, GTase and N7-MTase. These enzymes include, but are not limited to: (i) Acanthamoeba polyphaga mimivirus capping enzyme R382 (Raoult, Audic et al. 2004, Benarroch, Smith et al. 2008) (NCBI APMV genomic sequence NC_006450; UniProtKB/Swiss-Prot accession number Q5UQX1), (ii) ORF3 capping enzyme from yeast Kluyveromyces lactis linear extra-chromosomal episome pGKL2 (Tommasino, Ricci et al. 1988, Tiggemann, Jeske et al. 2001) (NCBI Kluyveromyces lactis CB 2359 pGKL2 genomic sequence NC_010187; UniProtKB/Swiss-Prot accession number P05469), (iii) African swine fever virus NP868R capping enzyme (Pena, Yanez et al. 1993, Jais 2011, Dixon, Chapman et al. 2013, Jais, Decroly et al. 2018) (NCBI ASFV genomic sequence strain BA71V NC_001659; UniProtKB/Swiss-Prot accession number P32094), VP4 Bluetongue virus capping enzyme (NCBI BTV serotype 10 genomic sequence Y00421; UniProtKB/Swiss-Prot accession number P07132).
    • Capping enzymes consisting of two subunits which include, but are not limited to: (i) the mammalian capping enzymes that consists of the RNGTT subunit having both RTPase and GTase enzymatic activities (Yue, Maldonado et al. 1997, Pillutla, Yue et al. 1998, Tsukamoto, Shibagaki et al. 1998, Yamada-Okabe, Doi et al. 1998) (also named HCE1; human and mouse UniProtKB/Swiss-Prot accession number O60942 and O55236, respectively) and RNMT having N7-MTase enzymatic activity (Pillutla, Yue et al. 1998, Tsukamoto, Shibagaki et al. 1998) (human and mouse UniProtKB/Swiss-Prot accession number Q05D80 and D3YYS7, respectively), (ii) the vaccinia capping enzyme that consists of the D1R gene product having RTPase, GTase and N7-MTase enzymatic domains (Cong and Shuman 1993, Niles and Christen 1993, Mao and Shuman 1994, Cong and Shuman 1995, Mao and Shuman 1996, Myette and Niles 1996, Yu and Shuman 1996, Yu, Martins et al. 1997, Gong and Shuman 2003) (genomic sequence strain Western Reserve NC_006998.1; UniProtKB/Swiss-Prot accession number P04298) and D12L gene product that has no intrinsic enzymatic activity but enhances drastically the RNA N7-MTase activity of the D1R subunit (Higman, Bourgeois et al. 1992, Higman, Christen et al. 1994, Mao and Shuman 1994, Schwer, Hausmann et al. 2006, De la Pena, Kyrieleis et al. 2007) (genomic sequence strain Western Reserve NC_006998.1; Gene 3707515; UniProtKB/Swiss-Prot accession number P04318).
    • Capping enzymes that consist of three subunits, such Saccharomyces cerevisiae CET1 with RTPase (Tsukamoto, Shibagaki et al. 1997, Gu, Rajashankar et al. 2010) (UniProtKB/Swiss-Prot accession number O13297), CEG1 with GTase (Shibagaki, Itoh et al. 1992, Yamada-Okabe, Doi et al. 1998, Gu, Rajashankar et al. 2010) (UniProtKB/Swiss-Prot accession number Q01159), and ABD1 having N7-MTase catalytic activities (Mao, Schwer et al. 1995, Schwer, Saha et al. 2000) (UniProtKB/Swiss-Prot accession number P32783).


As used herein, the term “cap-0 non canonical capping enzymes” refers to enzymes able to add a cap-0 structure at the 5′ end of RNA molecules but in a pathway which differs from the canonical enzymatic process. As of today, three non-canonical 5′ RNA cap synthesis mechanisms have been described:

    • Firstly, the m7GTP RNA capping pathway of various ss(+)RNA viruses of the alphavirus (e.g. Semliki Forest virus and Sindbis virus), potexvirus (e.g. Bamboo mosaic virus), tobamovirus (e.g. Tobacco mosaic virus), Togaviridae (e.g. Rubella virus and Chikungunya virus) and Hepeviridae (e.g. Hepatitis E virus) families (Decroly, Ferron et al. 2011). This RNA capping pathway relies on three sequential enzymatic reactions: (a) RTPase similar to the conventional pathway (for example, nsP2 protein of Semliki Forest virus resulting from the apparent cleavage of the non-structural P123 polyprotein; UniProtKB/Swiss-Prot accession number P08411), hydrolyzes the γ-β bond at the 5′-end of the RNA, (b) methylation of GTP molecule by an atypical N7-MTase (for example, nsP1 protein of Semliki Forest virus also resulting from the apparent cleavage of the non-structural P123 polyprotein for example; UniProtKB/Swiss-Prot accession number P08411), (c) m7GTP is then recognized as a substrate by an atypical GTase (also nsP1 of protein of Semliki Forest virus for example) and transferred onto the 5′-end of the acceptor ppRNA, to yield a typical m7GpppN cap-0 structure (Decroly, Ferron et al. 2011). These three enzymatic activities, in addition to a RNA-dependent RNA polymerase catalytic domain, can be found in a single viral protein, i.e. the Bamboo Mosaic Virus mRNA capping enzyme ORF1 (Li, Shih et al. 2001, Huang, Han et al. 2004, Huang, Hsu et al. 2005, Han, Tsai et al. 2007) (NCBI BMV isolate BaMV-O genomic sequence NC_001642; UniProtKB/Swiss-Prot accession number Q65005).
    • Secondly, the GDP RNA capping pathway of many ss(−)RNA viruses of the Rhabdoviridae (e.g. vesicular stomatitis virus and Rabies virus), paramyxoviridae (e.g. human respiratory syncytial virus and Measles virus), Bornaviridae (e.g. bornavirus), and Filoviridae (e.g. Ebola virus and Marburg virus) families (Decroly, Ferron et al. 2011), which catalyzes the formation of a cap-0/cap-1 structure in four enzymatic steps. For instance, the single subunit large L protein from the human respiratory syncytial virus (UniProtKB/Swiss-Prot accession number P28887) can complete these four enzymatic steps by itself, in addition of having an RNA dependent RNA polymerase activity: (a) the NTPase activity is responsible for the hydrolysis of a GTP into a GDP, (b) the L protein hydrolyzes the α-β bond of the pppRNA triphosphate moiety, thereby releasing pyrophosphate and creating a covalent enzyme-pRNA intermediate (i.e. RNA with monophosphate 5′-end), (c) the pRNA moiety is then transferred onto the GDP to form a GpppN block RNA. In this case, only the α-phosphate originates from the RNA whereas both the β and γ-phosphates are contributed by the GDP, (d) finally, synthesis of the cap-0 then cap-1 structures is completed by two successive methylations at m7pppN and 2′-residue on the first transcribed nucleotide, respectively (Grdzelishvili, Smallwood et al. 2005, Li, Fontaine-Rodriguez et al. 2005, Grdzelishvili, Smallwood et al. 2006, Ogino and Banerjee 2007, Li, Rahmeh et al. 2008, Ogino and Banerjee 2008, Rahmeh, Li et al. 2009).
    • Thirdly, RNA cap snatching, which is a process by which some viruses unable to synthesize their own cap structures, acquire capping by stealing it from host mRNA. Viruses belonging to this class include representatives of the Orthomyxoviridae (e.g. Influenza virus, Thogoto virus), Arenaviridae (e.g. Lassa virus, Machupo virus) and Bunyaviridae families (e.g. Hantaan virus, La Crosse virus, Tomato Spotted Wilt virus) (Decroly, Ferron et al. 2011). To acquire their cap structure, nucleotide sequence between 10 and 20 nucleotides in size is cleaved from the 5′ end of host capped mRNAs by an endonuclease activity encompassed within the viral RNA dependent RNA polymerase and transferred to the viral genomic RNA. The capped leader obtained is subsequently used to prime transcription on the viral genomic RNA, which ultimately leads to the synthesis of capped, translatable viral mRNAs. The Arenaviridae and Bunyaviridae express a large monomeric polymerase to ensure cap snatching. Orthomyxoviridae influenza virus have heterotrimeric polymerase, consisting of PB1 (UniProtKB/Swiss-Prot accession number strain A/Puerto Rico/8/1934 H1 N1 P03431), PB2 (UniProtKB/Swiss-Prot accession number strain A/Puerto Rico/8/1934 H1 N1 P03428) and PA (UniProtKB/Swiss-Prot accession number strain A/Puerto Rico/8/1934 H1 N1 P03433). All these three subunits are required for endonuclease activity but the enzymatic activity is thought to reside in the amino-terminal domain of the PA subunit (Ohlmann, Rau et al. 1995).


As used herein, the term “cap-1 capping enzymes” refers to enzymes able to add cap-1 structure at the 5′end of RNA molecules.


As used herein, the term “cap-2 capping enzymes” refers to enzymes able to add cap-2 structure at the 5′end of RNA molecules.


In mammalians, higher eukaryotes and some viruses, two cap modifications are found, which are lacking in yeast and plant mRNAs (Langberg and Moss 1981) and are generated by methylation of the 2′ hydroxy-groups of the ribose moiety of nucleotides at the 5′ end of the mRNA: cap-1 at the first mRNA nucleotide, and cap-2 at the second one. Cap-1 methylation is found on nearly all mammalian mRNA molecules, while only half of the mRNA contain a 2′-O-methylated residue on the second transcribed nucleotide.


In mammalians, cap-1 and cap-2 modifications are performed by two ribose-2′-O methyltransferases, (also named nucleoside-2′-methyltransferase or 2′-O-MTases) (Belanger, Stepinski et al. 2010). Firstly, MTR1 (cap-1 ribose-2′-O MTase activity, also named FTSJD2, KIAA0082 or ISG95; UniProtKB/Swiss-Prot accession number Q8N1G2), which is exclusively found in the nucleus and contains a putative nuclear localization signal and a G-patch domain that is potentially involved in RNA binding (Haline-Vaz, Silva et al. 2008). Noticeably, MRT1 associates with the CTD of RNA polymerase II, which indicate that cap-1 formation occurs early in the synthesis of mRNA (Langberg and Moss 1981). Secondly, MTR2 (cap 2 ribose-2′-O MTase, also named FTSJD1 or FLJ11171; UniProtKB/Swiss-Prot accession number Q8IYT2) transfers a methyl group from S-adenosylmethionine to the 2′-O-ribose of the second nucleotide of mRNA and small nuclear RNA. Nor N7 methylation of the guanosine cap-0 or cap-1 modification is required for MTR2, but the presence of cap-1 increases MTR2 activity. The MTR2 protein is distributed throughout the nucleus and cytosol, in contrast to the nuclear MTR1 (Keith, Ensinger et al. 1978).


Some eukaryotic viruses have their own cap-1 and/or cap-2 2′-O-MTases, including:

    • VP39 from the vaccinia virus (NCBI genomic sequence NC_006998.1; UniProtKB/Swiss-Prot accession number YP_232977) that has both cap-1 and cap-2 2′-O-MTase enzymatic activities (Schnierle, Gershon et al. 1994, Shi, Yao et al. 1996, Hu, Gershon et al. 1999),
    • Orf69 from the Autographa californica Nucleopolyhedrovirus (NCBI genomic sequence NC_001623.1; UniProtKB/Swiss-Prot accession number P41469) (Wu and Guarino 2003),
    • nsp16 from coronavirus (residues 6776-7073 of the polyprotein 1 ab of the human SARS coronavirus NCBI genomic sequence NC_004718.3; UniProtKB/Swiss-Prot accession number POC6X7) (Reinisch, Nibert et al. 2000, Decroly, Imbert et al. 2008, Chen, Cai et al. 2009, Lugari, Betzi et al. 2010),
    • λ2 protein from Reovirus (e.g. mammalian orthoreovirus type 3, strain Dearing; NCBI genomic sequence J03488; UniProtKB/Swiss-Prot accession number P11079), which has 2′-O-MTase in addition to GTase and N7-MTase enzymatic activities (Bujnicki and Rychlewski 2001), VP4 from the bluetongue virus (NCBI BTV serotype 10 genomic sequence ID Y00421; UniProtKB/Swiss-Prot accession number P07132), NS5 from the flaviviruses that include dengue virus yellow fever virus, Zika virus, West Nile virus, Meaban virus, Yokose virus, St. Louis encephalitis virus, Japanese encephalitis virus, tick-borne encephalitis virus (e.g. polyprotein from Dengue virus type 1 strain Nauru/West Pac/1974; NCBI genomic sequence U88535; UniProtKB/Swiss-Prot accession number P17763) can also methylate internal adenosine residues of mRNA (Dong, Chang et al. 2012).


In one embodiment, the chimeric enzyme according to the invention comprises or consists of:

    • at least one catalytic domain of a RNA triphosphatase;
    • at least one catalytic domain of a guanylyltransferase;
    • at least one catalytic domain of a N7-guanine methyltransferase; and
    • at least one RNA-binding domain of a protein-RNA tethering system;


      in particular, wherein at least one of said catalytic domains is a catalytic domain of a cap-0 canonical capping enzyme, more particularly of a virus cap-0 canonical capping enzyme.


As used herein, the term “RNA triphosphatase” (RTPase) relates to the enzyme, which removes the γ phosphate residue of 5′ triphosphate end of nascent pre-mRNA to diphosphate (Furuichi and Shatkin 2000).


As used herein, the term “RNA guanylyltransferase” (GTase) refers to the enzyme, which transfers GMP from GTP to the diphosphate nascent RNA terminus (Furuichi and Shatkin 2000).


As used herein, the term “N7-guanine methyltransferase” (N7-MTase) relates to the enzyme, which adds a methyl residue on azote 7 of guanine to the GpppN cap (Furuichi and Shatkin 2000).


Said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase, can be of the same or of different capping enzymes.


Preferably, said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase are from one or several cytoplasmic enzymes, which have advantageously relatively simple structure and well-characterized enzymatic activities. Thus, in particular, said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase can be catalytic domains of one or several virus capping enzymes, or of capping enzymes of cytoplasmic episomes.


In one embodiment, said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase are from one or several virus capping enzymes, in particular selected in the group consisting of the wild type vaccinia virus capping enzyme, the wild type bluetongue virus capping enzyme, the wild type bamboo mosaic virus capping enzyme, the wild type African swine fever virus capping enzyme, the wild type Acanthamoeba polyphaga mimivirus capping enzyme, the wild type Organic Lake phycodnavirus 1 (OLPV1) capping enzyme, the wild type Organic Lake phycodnavirus 2 (OLPV2) capping enzyme, the wild type Phaeocystis globosa virus capping enzyme, the wild type Chrysochromulina ericina virus capping enzyme and mutants or derivatives thereof which are able respectively to remove the γ phosphate residue of 5′ triphosphate end of nascent pre-mRNA to diphosphate or transfer GMP from GTP to the diphosphate nascent RNA terminus or add a methyl residue on azote 7 of guanine to the GpppN cap, more particularly of the wild type African swine fever virus capping enzyme and mutants or derivatives thereof which are able respectively to remove the γ phosphate residue of 5′ triphosphate end of nascent pre-mRNA to diphosphate or transfer GMP from GTP to the diphosphate nascent RNA terminus or add a methyl residue on azote 7 of guanine to the GpppN cap.


As used herein the term “vaccinia virus capping enzyme” relates to the heterodimeric D1R/D12L capping enzyme of vaccinia virus. The D1R gene product having RTPase, GTase and N7-MTase enzymatic domains (Cong and Shuman 1993, Niles and Christen 1993, Mao and Shuman 1994, Cong and Shuman 1995, Mao and Shuman 1996, Myette and Niles 1996, Yu and Shuman 1996, Yu, Martins et al. 1997, Gong and Shuman 2003) (genomic sequence strain Western Reserve NC_006998.1; UniProtKB/Swiss-Prot accession number P04298) and D12L gene product that has no intrinsic enzymatic activity but enhances drastically the RNA N7-MTase activity of the D1R subunit (Higman, Bourgeois et al. 1992, Higman, Christen et al. 1994, Mao and Shuman 1994, Schwer, Hausmann et al. 2006, De la Pena, Kyrieleis et al. 2007) (genomic sequence strain Western Reserve NC_006998.1; Gene 3707515; UniProtKB/Swiss-Prot accession number P04318).


As used herein the term “bluetongue virus capping enzyme” relates to the single-unit VP4 capping enzyme of Bluetongue virus (BTV), which is a 76 kDa protein (644 amino-acids; for sequence, see for instance NCBI BTV serotype 10 genomic sequence Y00421; Gene 2943157; UniProtKB/Swiss-Prot P07132). This capping enzyme is likely able to homodimerize through the leucine zipper located near its carboxyl-terminus (Ramadevi, Rodriguez et al. 1998). VP4 catalyze all three enzymatic steps required for mRNA m7GpppN capping synthesis: RTPase (Martinez-Costas, Sutton et al. 1998), GTase (Martinez-Costas, Sutton et al. 1998, Ramadevi, Burroughs et al. 1998) and N7-MTase (Ramadevi, Burroughs et al. 1998).


As used herein, the term “bamboo mosaic virus capping enzyme” relates to ORF1, the Bamboo Mosaic Virus (BMV) mRNA capping enzyme, which is a single-unit 155-kDa protein (1365-amino acids; NCBI BMV isolate BaMV-O genomic sequence NC_001642; Gene 1497253; UniProtKB/Swiss-Prot Q65005). ORF1 protein has all the enzymatic activities required to generate m7GpppN mRNA capping, i.e. RTPase (Li, Shih et al. 2001, Han, Tsai et al. 2007), GTase and N7-MTase (Li, Chen et al. 2001, Li, Shih et al. 2001).


As used herein, the term “African swine fever virus capping enzyme” (ASFV) relates to the NP868R capping enzyme (G4R), which is a single-unit 100 kDa protein (868 amino-acids; NCBI ASFV genomic sequence strain BA71V NC_001659; Gene 1488865; UniProtKB/Swiss-Prot P32094), which has all the enzymatic activities required to generate m7GpppN mRNA capping, i.e. RTPase, GTase and N7-MTase (Pena, Yanez et al. 1993, Jais 2011, Dixon, Chapman et al. 2013, Jais, Decroly et al. 2018).


As used herein, the term “Acanthamoeba Polyphaga mimivirus capping enzyme” relates to R382, (APMV), which is another single-unit 136.5 kDa protein (1170 amino-acids; NCBI APMV genomic sequence NC_006450; Gene 3162607; UniProtKB/Swiss-Prot Q5UQX1).


As used herein, the term “Organic Lake phycodnavirus 1 (OLPV1) capping enzyme” refers to a 889 single-unit amino-acids, as referenced with NCBI accession number ADX05869.1.


As used herein, the term “Organic Lake phycodnavirus 2 (OLPV2) capping enzyme” refers to a 942 single-unit amino-acids, as referenced with NCBI accession number ADX06468.1.


As used herein, the term “Phaeocystis globosa virus capping enzyme” refers to a 1066 single-unit amino-acids as referenced with UniProtKB/Swiss-Prot YP_008052553.1.


As used herein, the term “Chrysochromulina ericina virus capping enzyme” refers to a 965 single-unit amino-acids as referenced with UniProtKB/Swiss-Prot YP_009173557.1.


In one embodiment, said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase are from the capping enzymes of cytoplasmic episomes, like pGKL2. In particular, said catalytic domains of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase are included in the whole or part of the capping enzyme of the yeast linear extra-chromosomal episome pGKL2 encoded by the ORF3 gene of Kluyveromyces lactis pGKL2 (NCBI Kluyveromyces lactis CB 2359 pGKL2 genomic sequence NC_010187; UniProtKB/Swiss-Prot P05469) and which is a 594 amino-acid protein (70.6 kDa protein).


In one embodiment of the chimeric enzyme according to the invention, said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, said catalytic domain of a N7-guanine methyltransferase, are included in a monomer, i.e. in one polypeptide. For example, said monomer can be a monomeric capping enzyme or a monomeric chimeric enzyme according to the invention.


In particular, said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, and said catalytic domain of a N7-guanine methyltransferase are included in a monomeric capping enzyme. In this case, the chimeric enzyme according to the invention comprise a monomeric capping enzyme, which includes said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, and said catalytic domain of a N7-guanine methyltransferase. Said monomeric capping enzyme can be a monomeric virus capping enzyme, in particular selected in the group consisting of the wild type bluetongue virus capping enzyme, the wild type bamboo mosaic virus capping enzyme, the wild type African swine fever virus capping enzyme, the wild type Acanthamoeba polyphaga mimivirus capping enzyme, the wild type OLPV1 capping enzyme, the wild type OLPV2 capping enzyme, the wild type Phaeocystis globosa virus capping enzyme, the wild type Chrysochromulina ericina virus capping enzyme and mutants and derivatives thereof which are able to add a m7GpppN cap at the 5′-terminal end of RNA molecules and, more particularly of the wild type African swine fever virus capping enzyme and mutants and derivatives thereof which are able to add a m7GpppN cap at the 5′-terminal end of RNA molecules, and even more particularly the wild type African swine fever virus capping enzyme


The chimeric enzyme according to the invention can also further comprise a domain, which enhances the activity of at least one catalytic domain of the chimeric enzyme of the invention, in particular of at least one catalytic domain of a capping enzyme, more particularly of at least one catalytic domain selected in the group consisting of a catalytic domain of a RNA triphosphatase, a catalytic domain of a guanylyltransferase, a catalytic domain of a N7-guanine methyltransferase, preferably of a N7-guanine methyltransferase.


For example said domain, which enhance the activity of at least one catalytic domain of the chimeric enzyme of the invention, can be a 31-kDa subunit encoded by the vaccinia virus D12L gene (genomic sequence NC_006998.1; Gene3707515; UniProtKB/Swiss-Prot YP_232999.1), which has no intrinsic enzymatic activity, but enhances drastically the RNA N7-guanine methyltransferase activity of the D1R subunit of the vaccinia mRNA capping enzyme (Higman, Bourgeois et al. 1992, Higman, Christen et al. 1994, Mao and Shuman 1994).


In one embodiment, the chimeric enzyme according to the invention further comprises at least one catalytic domain of a 5′-end RNA processing enzyme other than cap-0, cap-1 and cap-2 capping enzymes.


As used herein, the term “5′-end RNA processing enzyme other than cap-0, cap-1 and cap-2 capping enzymes” relates to enzymes able to modify the ultimate or penultimate bases of a mRNA sequence, other than cap-0, cap-1 and cap-2 capping enzymes, including N6-methyl-adenosine transferase and enzymes able to add 2,2,7-trimethylguanosine (TMG) and 2,7-trimethylguanosine (DMG) cap modifications at the 5′end of RNA molecules.


Other capping modifications than cap-0, cap-1, cap-2 modifications have been characterized in eukaryotes or viruses mRNA. For instance, the m6A methylation by N6-methyl-adenosine transferase of the first base is a reversible modification that influences cellular mRNA fate (Mauer, Luo et al. 2017). 2,2,7-trimethylguanosine (TMG) and 2,7-trimethylguanosine (DMG) cap modifications, which are present on snRNAs, telomerase RNAs, trans-spliced nematode mRNAs and certain viral mRNAs, can confer an advantage of translation (Darzynkiewicz, Stepinski et al. 1988, Cai, Jankowska-Anyszka et al. 1999). TMG and DMG are performed by specialized enzymes from viruses (e.g. L320 from DNA mimivirus, UniProtKB/Swiss-Prot accession number Q5UQR2 (Benarroch, Qiu et al. 2009); protozoan (e.g. Giardia lamblia trimethylguanosine synthase (NCBI accession number EAA46438 (Hausmann and Shuman 2005); lower eukaryotes (e.g. Schizosaccharomyces pombe trimethylguanosine synthase, UniProtKB/Swiss-Prot accession number Q09814 (Hausmann and Shuman 2005, Hausmann, Zheng et al. 2008, Benarroch, Jankowska-Anyszka et al. 2010) and mammalian (e.g. human trimethylguanosine synthase 1 UniProtKB/Swiss-Prot accession number Q96RS0 (Zhu, Qi et al. 2001, Hausmann, Zheng et al. 2008, Benarroch, Jankowska-Anyszka et al. 2010).


In one embodiment, the chimeric enzyme according to the invention further comprises at least one catalytic domain of a poly(A) polymerase.


In fact, unexpectidely, the inventor has demonstrated that monomeric or oligomeric chimeric enzymes comprising at least one catalytic domain of a capping enzyme, at least one RNA-binding domain of a protein-RNA tethering system and at least one catalytic domain of a poly(A) polymerase allows to highly increase, synergistically, the capping rate of specific mRNAs produced by RNA polymerase, compared to the combination of a capping enzyme fused to a RNA-binding domain in presence of a poly(A) polymerase fused to a RNA-binding domain. These results are unexpected since capping enzymes and poly(A) polymerases are not physically linked in the nature and contain no binding domain for a specific RNA sequence. One skilled in the art could have expected to obtain the same expression rate since the components are the same.


As used herein, the term “poly(A) polymerase” relates to any enzyme able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules.


In one embodiment, said catalytic domain of a poly(A) polymerase is a catalytic domain of a canonical poly(A) polymerase including mammalian (such as PAPOLA, PAPOLG), yeast, (such as Saccharomyces cerevisiae PAP1, Schizosaccharomyces pombe PLA1, Candida albicans PAP, Pneumocystis carinii PAP), protozoan, viral and bacterial canonical poly(A) polymerases.


In particular said catalytic domain of a poly(A) polymerase is a cytoplasmic canonical poly(A) polymerase, more particularly selected in the group consisting of:

    • mammalian cytoplasmic poly(A) polymerase including PAPOLB (human and mouse PAPOLB, UniProtKB/Swiss-Prot accession number Q9NRJ5 and O9WVP6, respectively), which is at least in part a cytoplasmic enzyme (Kashiwabara, Zhuang et al. 2000, Lee, Lee et al. 2000, Kashiwabara, Tsuruta et al. 2016); mutants of PAPOLA (human and mouse PAPOLA, UniProtKB/Swiss-Prot accession number P51003 and Q61183, respectively) wherein mutation or deletion of the nuclear localization signal can relocate the nuclear enzyme to the cytoplasm (Raabe, Murthy et al. 1994, Vethantham, Rao et al. 2008), and mutants of PAPOLG (human and mouse PAPOLG, UniProtKB/Swiss-Prot accession number Q9BWT3 and Q6PCL9, respectively) wherein mutation or deletion of the nuclear localization signal is likely to relocate the nuclear enzyme to the cytoplasm (Kyriakopoulou, Nordvarg et al. 2001),
    • yeast or protozoan poly(A) polymerases, e.g. mutants of Saccharomyces cerevisiae PAP1, UniProtKB/Swiss-Prot accession number P29468; Schizosaccharomyces pombe PLA1, UniProtKB/Swiss-Prot accession number Q10295), Candida albicans PAP (UniProtKB/Swiss-Prot accession number Q9UW26), Pneumocystis carinii PAP (also named Pneumocystis jiroveci; UniProtKB/Swiss-Prot accession number A0A0W4ZDF2), wherein mutation or deletion of the nuclear localization signal is likely to relocate the nuclear enzyme to the cytoplasm (Lingner, Kellermann et al. 1991), as well as other psychrotrophic, mesophilic, thermophilic or hyperthermophilic yeast or protozoan strains,
    • viral poly(A) polymerases, including the heterodimeric vaccinia virus poly(A) polymerase that consists of the VP55 catalytic subunit (UniProtKB/Swiss-Prot accession number strain Western Reserve P23371) and VP39 that acts as a processivity factor (UniProtKB/Swiss-Prot accession number strain Western Reserve P07617) (Gershon, Ahn et al. 1991), other poxvirus poly(A) polymerases (e.g. Cowpox virus, Monkeypox virus or Camelpox virus), African Swine Fever Virus (C475L, UniProtKB/Swiss-Prot accession number A0A0A1E081), Acanthamoeba polyphaga mimivirus R341 (UniProtKB/Swiss-Prot accession number E3VZZ8) and the Megavirus chilensis Mg561 poly(A) polymerases (NCBI Accession number: YP_004894612), Moumouvirus (NCBI accession number AEX62700), Mamavirus (NCBI accession number AEQ60527), Cafeteria roenbergensis BV-PW1 virus (NCBI accession number YP_003969918), Megavirus Iba (NCBI accession number AGD92490), Yellowstone lake mimivirus (NCBI accession number YP_009174112), Chrysochromulina ericina virus (NCBI accession number YP_009173345), organic lake phycodnavirus 1 (NCBI accession number ADX05881), organic lake phycodnavirus 2 (NCBI accession number ADX06298), Faustovirus (NCBI accession number AMN83802) and Phaeocystis globosa virus (NCBI accession number YP_008052392).
    • and mutants or derivatives thereof which are able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules.


In still another embodiment, said catalytic domain of a poly(A) polymerase is a catalytic domain of a non-canonical poly(A) polymerase, in particular of a cytoplasmic non-canonical poly(A) polymerase. “noncanonical poly(A) polymerases”, refers to enzymes, that do not have a tripartite structure involving a N-terminal nucleotidyltransferase (NT) catalytic domain, a central domain, and a C-terminal domain corresponding to the RNA binding domain (RBD) (Trippe, Sandrock et al. 1998). As of today, the following non-canonical poly(A) polymerases have been described in mammalians:

    • Firstly, the poly(A) polymerase GLD2 (also named PAPD4; human and mouse GLD2, UniProtKB/Swiss-Prot accession number Q6PIY7 and 091Y16, respectively), which was initially identified by sequence analogy with the Caenorhabditis elegans GLD2 (Wang, Eckmann et al. 2002). In addition, GLD4 (UniProtKB/Swiss-Prot accession number GSEFL0), a GLD2 homolog, has been characterized in Caenorhabditis elegans but is lacking in mammals (Schmid, Kuchler et al. 2009)
    • Secondly, the single-subunit mitochondrial poly(A) polymerase (also named PAPD1, TUTase1 or mtPAP; human and mouse PAPD1, UniProtKB/Swiss-Prot accession number Q9NVV4 and Q9D0D3, respectively),
    • Thirdly, the nucleolar RBM21 poly(A) polymerase (also named U6 TUTase, TUT1 or Star-PAP; Speckle Targeted PIPKIa Regulated Poly(A) Polymerase; human and mouse RBM21, UniProtKB/Swiss-Prot accession number Q9H6E5 and Q8R3F9, respectively),
    • Fourthly and fifthly, the putative cytoplasmic/nuclear PAPD5 (human and mouse PAPD5, UniProtKB/Swiss-Prot accession number Q8NDF8 and Q68ED3, respectively) and PAPD7 (also named POLS, human and mouse PAPD7, UniProtKB/Swiss-Prot accession number Q5XG87 and Q6PB75, respectively), which are orthologs to the yeast poly(A) polymerase TRF4 or TRF5 from the nuclear TRAMP complex (Trf4/Air2/Mtr4p Polyadenylation complex) (Haracska, Johnson et al. 2005),
    • Sixthly and seventhly, the cytoplasmic ZCCHC6 (human and mouse ZCCHC6, UniProtKB/Swiss-Prot accession number Q5VYS8 and Q5BLK4, respectively) and ZCCHC11 poly(A) polymerases (human and mouse ZCCHC11, UniProtKB/Swiss-Prot accession number Q5TAX3 and B2RX14, respectively).


In an embodiment, said catalytic domain of a poly(A) polymerase is a catalytic domain of a yeast or protozoan poly(A) polymerase, in particular selected in the group consisting of the wild type Saccharomyces cerevisiae PAP1 poly(A) polymerase, Schizosaccharomyces pombe PLA1, Candida albicans PAP (UniProtKB/Swiss-Prot accession number Q9UW26), Pneumocystis carinii PAP (UniProtKB/Swiss-Prot accession number A0A0W4ZDF2), the cytoplasmic mutants of the Saccharomyces cerevisiae, Schizosaccharomyces pombe, Candida albicans or Pneumocystis carinii poly(A) polymerases (wherein the nuclear localization signal is non-functional or deleted) and mutants or derivatives thereof, which are able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules.


The yeast or protozoan poly(A) polymerases have notably the advantage of its reduced molecular weight in comparison to mammalian canonical poly(A) polymerases (e.g. 568 and 566 amino-acids vs. 745 amino-acids for the Saccharomyces cerevisiae PAP1 and Schizosaccharomyces pombe PLA1 poly(A) polymerases vs. the human PAPOLA, UniProtKB/Swiss-Prot accession number P51003, respectively), as well as high processivity in tethering assays (Dickson, Thompson et al. 2001), As used herein, the term “PAP1 poly(A) polymerase” refers to the Saccharomyces cerevisiae PAP1 poly(A) polymerase (UniProtKB/Swiss-Prot accession number P29468),


As used herein, the term “PLA1 poly(A) polymerase” refers to the Schizosaccharomyces pombe PLA1 poly(A) polymerase, UniProtKB/Swiss-Prot accession number Q10295),


Said catalytic domain of a poly(A) polymerase can be a catalytic domain of a virus poly(A) polymerase, in particular selected in the group consisting of the wild type VP55 poly(A) polymerase, the wild type C475L poly(A) polymerase, the wild type R341 poly(A) polymerase and the wild type MG561 poly(A) polymerase and mutants or derivatives thereof, which are able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules.


The virus poly(A) polymerases have notably the advantage of their reduced molecular weight in comparison to mammalian poly(A) polymerases, their strong enzymatic activity and presence in the cytoplasmic compartment when expressed in mammalian cells. In addition, some of these enzymes, such as R341 and MG561 poly(A) polymerases do not require any know accessory protein cofactor.


As used herein, the term “VP55 poly(A) polymerase” relates to the catalytic subunit (UniProtKB/Swiss-Prot accession number strain Western Reserve P23371) of the heterodimeric vaccinia virus poly(A) polymerase. VP39, the second subunit, acts as a processivity factor (UniProtKB/Swiss-Prot accession number strain Western Reserve P07617).


As used herein, the term “C475L poly(A) polymerase” relates to the African Swine Fever Virus poly(A) polymerase (UniProtKB/Swiss-Prot accession number A0A0A1E081).


As used herein, the term “R341 poly(A) polymerase” relates to the Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase (UniProtKB/Swiss-Prot accession number E3VZZ8).


As used herein, the term “MG561 poly(A) polymerase” relates to the Megavirus chilensis MG561 poly(A) polymerases (NCBI Accession number: YP_004894612).


In one embodiment, the chimeric enzyme according to the invention further comprises at least one catalytic domain of a DNA-dependent RNA polymerase.


As used herein, the term “DNA-dependent RNA polymerase” (RNAPs) relates to nucleotidyl transferases that synthesize complementary strand of RNA from a single- or double-stranded DNA template in the 5′→3′ direction.


Preferably, said catalytic domain of a DNA-dependent RNA polymerase is a catalytic domain of an enzyme, which have a relatively simple structure and more preferably, which have characterized genomic enzymatic regulation elements (i.e. promoter and transcription termination signal). Thus, in particular, said catalytic domain of a DNA-dependent RNA polymerase can be a catalytic domain of a bacteriophage DNA-dependent RNA polymerase, of a bacterial DNA-dependent RNA polymerase or of a DNA-dependent RNA polymerase of various eukaryotic organelles (e.g. mitochondria, chloroplast and proplastids).


In one embodiment, said catalytic domain of a DNA-dependent RNA polymerase is a catalytic domain of a bacteriophage DNA-dependent RNA polymerase.


The bacteriophage DNA-dependent RNA polymerases have notably the advantage that they optimize the levels of transgene expression, in particular by having a higher processivity than the eukaryotic RNA polymerases. The bacteriophage DNA-dependent RNA polymerases have also a much simpler structure than most nuclear eukaryotic polymerases, which consist of multiple subunits (e.g. RNA polymerase II) and transcription factors. Most of the bacteriophage DNA-dependent RNA polymerases characterized so far are single-subunit enzymes, which require no accessory proteins for initiation, elongation, or termination of transcription (Chen and Schneider 2005). Several of these enzymes, which are named for the bacteriophages from which they have been cloned, have also well-characterized regulation genomic elements (i.e. promoter and termination signals), which are important for transgenesis.


There is also no competition between the endogenous gene transcription and the transgene transcription. The chimeric enzymes according to the invention, which comprise bacteriophage DNA-dependent RNA-polymerase moieties, allow the production of RNA transcripts in any eukaryotic species (e.g. yeast, rodents, and humans). They are not expensive, quick and easy to implement and thus appropriate for large-scale assays and protein productions; it allows the production of RNA transcripts in any biological system (e.g. acellular reaction mix, cultured cells, and living organisms), since in contrast to eukaryotic RNA polymerase such as RNA polymerase II, most of bacteriophage DNA-dependent RNA polymerases do not require associated factors for initiation, elongation or termination of transcription.


Said catalytic domain of a bacteriophage DNA-dependent RNA polymerase can be a catalytic domain of a bacteriophage DNA-dependent RNA polymerase, in particular selected in the group consisting of the wild type T7 RNA polymerase (NCBI genomic sequence NC_001604; Gene 1261050; UniProtKB/Swiss-Prot P00573), the wild type T3 RNA polymerase (NCBI genomic sequence NC_003298; Gene 927437; UniProtKB/Swiss-Prot Q778M8), the wild type K1E RNA polymerase (NCBI genome sequence AM084415.1, UniProtKB/Swiss-Prot Q2WC24), the wild type K1-5 RNA polymerase (NCBI genome sequence AY370674.1, NCBI YP_654105.1), the wild type K11 RNA polymerase (NCBI genomic K11 RNAP sequence NC_004665; Gene 1258850; UniProtKB/Swiss-Prot Q859H5), the wild type φA1122 RNA polymerase (NCBI genomic sequence NC_004777; Gene 1733944; UniProtKB/Swiss-Prot protein Q858N4), the wild type φYeo3-12 RNA polymerase (NCBI genomic sequence NC_001271; Gene 1262422; UniProtKB/Swiss-Prot Q9T145) and the wild type gh-1 RNA polymerase (NCBI genomic sequence NC_004665; Gene 1258850; UniProtKB/Swiss-Prot protein Q859H5), the wild type SP6 RNA polymerase (NCBI genomic sequence NC_004831; Gene 1481778; UniProtKB/Swiss-Prot protein Q7Y5R1), and mutants or derivatives thereof, which are able to synthesize single-stranded RNA complementary in sequence to the double-stranded template DNA in the 5′→3′ direction, more particularly the wild type T7 RNA polymerase, the wild type T3 RNA polymerase, the wild type SP6 RNA polymerase, the wild type K1-5 RNA polymerase and the wild type K1E RNA polymerase and mutants or derivatives thereof, which are able to synthesize single-stranded RNA complementary in sequence to the double-stranded template DNA in the 5′→3′ direction.


The prototype of bacteriophage RNA polymerases, i.e. the bacteriophage T7 RNA polymerase, has in particular the advantage that, in vitro, the enzyme is extremely processive and elongates 240-250 nucleotides/s at 37° C. in the 5′→3′ direction (Golomb and Chamberlin 1974, Lyakhov, He et al. 1997, Zhang and Studier 1997, Finn, MacLachlan et al. 2005). Moreover, when expressed in eukaryotic cells, the bacteriophage T7 RNA polymerase, remains largely in the cytoplasm (Elroy-Stein and Moss 1990, Gao and Huang 1993, Brisson, He et al. 1999, Jais, Decroly et al. 2018), and thus optimizes the levels of transgene expression by avoiding the active transfer of large DNA molecules (i.e. transgene) from the cytoplasm to the nucleus of eukaryotic cells and the export of RNA molecules from the nucleus to the cytoplasm.


The catalytic domain of a DNA-dependent RNA polymerase can be the one of the wild type of the K1E or K1-5 RNA polymerase but also of mutants of the K1E or K1-5 RNA polymerases, which are able to synthesize single-stranded RNA complementary in sequence to the double-stranded template DNA in the 5′→3′ direction, even with processivity. For example, said mutants can be selected in the group consisting of the K1E RNA polymerase mutants R551S (Jais, Decroly et al. 2018), F644A, Q649S, G645A, R627S, 1810S, D812E (Makarova, Makarov et al. 1995), and K631M (Osumi-Davis, de Aguilera et al. 1992, Osumi-Davis, Sreerama et al. 1994), in particular R551 S (Jais, Decroly et al. 2018).


Preferably, said catalytic domain of the DNA-dependent RNA-polymerase of the chimeric enzyme according to the invention is from different enzymes than those of the host cell to prevent the competition between the endogenous gene transcription and said DNA sequence transcription.


The chimeric enzyme according to the invention comprises at least said catalytic domains but can further comprise the whole or part of the enzymes containing said catalytic domains. In fact, according to one embodiment of the chimeric enzyme according to the invention, said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase and said catalytic domain of a N7-guanine methyltransferase can be included in the whole or part of a capping enzyme, preferably of a monomeric capping enzyme. Said catalytic domain of a poly(A) polymerase can also be included in the whole or part of a poly(A) polymerase. Said catalytic domain of a DNA-dependent RNA polymerase can also be included in the whole or part of a DNA-dependent RNA polymerase, preferably of a monomeric DNA-dependent RNA polymerase.


Unexpectedly, the inventor has demonstrated that a chimeric enzyme of the invention is able to generate translatable RNA in the cells, therefore generating the key modifications required for its recognition and use by the host-cell ribosomal machinery.


As used herein, the terms «link» and «bound» encompass covalent and non-covalent linkage.


Said RNA-binding domain can be linked by covalent (directly or indirectly by a linking peptide) linkage to one catalytic domain of the chimeric enzyme according to the invention, in particular selected in the group consisting of:

    • said catalytic domain of a capping enzyme, in particular said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, said catalytic domain of a N7-guanine methyltransferase;
    • said catalytic domain of a poly(A) polymerase,
    • said domain which enhances the activity of at least one catalytic domain of the chimeric enzyme of the invention, preferably of at least one catalytic domain selected in the group consisting of a catalytic domain of a RNA triphosphatase, a catalytic domain of a guanylyltransferase, a catalytic domain of a N7-guanine methyltransferase, particularly of a N7-guanine methyltransferase; and
    • said catalytic domain of a 5′end RNA processing enzyme other than cap-0, cap-1 and cap-2 capping enzymes.


Linking peptide has the advantage of generating fusion proteins in which steric hindrance is minimizes and enough space is provided for the components of the fusion protein to remain in their native conformation.


Said linking peptide of the invention can be selected from the group consisting of:

    • peptides of formula (GlymSerp)n, in which:
    • m represents an integer from 0 to 12, in particular from 1 to 8, and more particularly from 3 to 6 and even more particularly 4;
    • p represents an integer from 0 to 6, in particular from 0 to 5, more particularly from 0 to 3 and more particularly 1; and
    • n represents an integer from 0 to 30, in particular from 0 to 12, more particularly from 0 to 8 and even more particularly between 1 and 6 inclusive;


      in particular peptides of formula (GlymSerp)n, in which:
    • m represents 4;
    • p represents 0 or 1; and
    • n represents 1, 2 or 4;


      more particularly peptides of formula Gly4, (Gly4Ser)1 (Gly4Ser)2 and (Gly4Ser)4.


The flexible linker peptides of formula (GlymSerp)n have the advantages that the glycine residues confer peptide flexibility, while the serine provide some solubility (Huston, Levinson et al. 1988). Furthermore, the absence of sensitive sites for chymotrypsin I, factor Xa, papain, plasmin, thrombin and trypsin in the (GlymSerp)n linker sequences is supposed to increase the overall stability of the resulting fusion proteins.


(GlymSerp)n linkers of variable lengths are commonly used to engineer single-chain Fv fragment (sFv) antibodies (Huston, Levinson et al. 1988). In addition, (GlymSerp)n linkers have been used to generate various fusion proteins, which frequently retain the biological activities of each of their components (Newton, Xue et al. 1996, Lieschke, Rao et al. 1997, Shao, Zhang et al. 2000, Hu, Li et al. 2004).


Other types of peptide linkers can be also considered to generate chimeric enzymes according to the invention, such as GGGGIAPSMVGGGGS (SEQ ID No 48) (Turner, Ritter et al. 1997), SPNGASNSGSAPDTSSAPGSQ (SEQ ID No 49) (Hennecke, Krebber et al. 1998), EGKSSGSGSESKSTE (SEQ ID No 50) (Bird, Hardman et al. 1988), EGKSSGSGSESKEF (SEQ ID No 51) (Newton, Xue et al. 1996), GGGSGGGSGGGTGGGSGGG (SEQ ID No 52) (Robinson and Sauer 1998), GSTSGSGKSSEGKG (SEQ ID No 53) (Bedzyk, Weidner et al. 1990), YPRSIYIRRRHPSPSLTT (SEQ ID No 54) (Tang, Jiang et al. 1996), GSTSGSGKPGSGEGS (SEQ ID No 55) (Ting, Kain et al. 2001), SSADDAKKDAAKKDDAKKDDAKKDA (SEQ ID No 56) (Pantoliano, Bird et al. 1991), GSADDAXXDAAXKDDAKKDDAKKDGS (SEQ ID No 57) (Gregoire, Lin et al. 1996), LSADDAKKDAAKKDDAKKDDAKKDL (SEQ ID No 58) (Pavlinkova, Beresford et al. 1999), AEAAAKEAAAKEAAAKA (SEQ ID No 59) (Wickham, Carrion et al. 1995), GSTSGSGKPGSGEGSTGAGGAGSTSGSGKPSGEG (SEQ ID No 60) (Ting, Kain et al. 2001), LSLEVAEEIARLEAEV (SEQ ID No 61) (Ting, Kain et al. 2001), GTPTPTPTPTGEF (SEQ ID No 62) (Gustaysson, Lehtio et al. 2001), GSTSGSGKPGSGEGSTKG (SEQ ID No 63) (Whitlow, Bell et al. 1993) and GSHSGSGKP (SEQ ID No 64) (Ting, Kain et al. 2001) as described previously in the patent application WO 2011/128444.


Said RNA-binding domain of a protein-RNA tethering system and said catalytic domains can also be assembled by specific protein elements, like leucine zippers.


The C-terminal end or the N-terminal end of said RNA-binding domain can be linked to the N-terminal end or the C-terminal end of one of said catalytic domain of the chimeric enzyme of the invention, respectively.


Preferably, the C-terminal end of said RNA binding domain is linked by covalent (directly or indirectly by a linking peptide preferably of formula Gly4, (Gly4Ser)1 (Gly4Ser)2 or (Gly4Ser)4, even more preferably of formula (Gly4Ser)1 or a Gly4) linkage to the N-terminal end of one of the catalytic domain selected in the group consisting of:

    • said catalytic domain of a capping enzyme, in particular said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, said catalytic domain of a N7-guanine methyltransferase;
    • said catalytic domain of a poly(A) polymerase,
    • said domain which enhances the activity of at least one catalytic domain of the chimeric enzyme of the invention, preferably of at least one catalytic domain selected in the group consisting of a catalytic domain of a RNA triphosphatase, a catalytic domain of a guanylyltransferase, a catalytic domain of a N7-guanine methyltransferase, particularly of a N7-guanine methyltransferase; and
    • said catalytic domain of a 5′end RNA processing enzyme other than cap-0, cap-1 and cap-2 capping enzymes.


In one embodiment, the C-terminal end of said RNA binding domain is linked by covalent (directly or indirectly by a linking peptide preferably of formula Gly4, (Gly4Ser)1 (Gly4Ser)2 or (Gly4Ser)4, even more preferably of formula (Gly4Ser)1 or a Gly4) linkage to the N-terminal end of one of the catalytic domain selected in the group consisting of:

    • said catalytic domain of a capping enzyme, in particular said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, said catalytic domain of a N7-guanine methyltransferase; and
    • said catalytic domain of a poly(A) polymerase.


In one embodiment, the chimeric enzyme of the invention is a fusion protein.


As used herein, the term “fusion protein” relates to artificial proteins created through the joining of two or more proteins or protein domains that originally coded for separate proteins. Translation of this fusion gene results in a single or multiple polypeptides with functional properties derived from each of the original proteins.


In one embodiment, the chimeric enzyme of the invention is a fusion protein, wherein:

    • the C-terminal end of said RNA binding domain is linked by covalent linkage to the N-terminal end of said catalytic domain of a poly(A) polymerase,
    • the C-terminal end of said catalytic domain of a poly(A) polymerase is linked by covalent linkage, in particular by a linking peptide, to the N-terminal end of one of the catalytic domain of a capping enzyme; and
    • the N-terminal end of said catalytic domain of a DNA-dependent RNA polymerase is linked by covalent linkage, in particular by a linking peptide, to the C-terminal end of one of the catalytic domain of a capping enzyme.


In one embodiment, the chimeric enzyme of the invention is a fusion protein, wherein:

    • the C-terminal end of said RNA binding domain is linked by covalent linkage to the N-terminal end of said catalytic domain of a poly(A) polymerase,
    • the C-terminal end of said catalytic domain of a poly(A) polymerase is linked by covalent linkage to the N-terminal end of one of the catalytic domain selected in the group consisting of:
      • said catalytic domain of a RNA triphosphatase,
      • said catalytic domain of a guanylyltransferase, and
      • said catalytic domain of a N7-guanine methyltransferase,


        in particular, of said catalytic domain of a RNA triphosphatase; and
    • the N-terminal end of said catalytic domain of a DNA-dependent RNA polymerase is linked by covalent linkage, in particular by a linking peptide, to the C-terminal end of one of the catalytic domain selected in the group consisting of:
      • said catalytic domain of a RNA triphosphatase,
      • said catalytic domain of a guanylyltransferase, and
      • said catalytic domain of a N7-guanine methyltransferase


        in particular, of said catalytic domain of a N7-guanine methyltransferase


In one embodiment of the chimeric enzyme according to the invention, at least two, in particularly at least three, at least four and more particularly the whole catalytic domains can be assembled, fused, or bound directly or indirectly by a linking peptide (particularly by a linking peptide of formula Gly4, (Gly4Ser)1, (Gly4Ser)2 or (Gly4Ser)4, more particularly of formula (Gly4Ser)2.


In particular, at least two, particularly at least three and more particularly the whole catalytic domains selected in the group consisting of:

    • a catalytic domain of a capping enzyme, in particular selected in the group consisting of a catalytic domain of a RNA triphosphatase, a catalytic domain of a guanylyltransferase and a catalytic domain of a N7-guanine methyltransferase,
    • a catalytic domain of a DNA-dependent RNA polymerase, and a
    • a catalytic domain of a poly(A) polymerase;


      particularly consisting of:
    • a catalytic domain of a RNA triphosphatase,
    • a catalytic domain of a guanylyltransferase,
    • a catalytic domain of a N7-guanine methyltransferase, and
    • a catalytic domain of a DNA-dependent RNA polymerase


      are bound directly or by a linking peptide, particularly selected in the group consisting of linking peptide of formula Gly4, (Gly4Ser)1, (Gly4Ser)2 and (Gly4Ser)4, more particularly of formula (Gly4Ser)2.


Preferably, at least said catalytic domain of a DNA-dependent RNA polymerase is bound by a linking peptide (particularly selected in the group consisting of linking peptide of formula Gly4, (Gly4Ser)1, (Gly4Ser)2 and (Gly4Ser)4, more particularly of formula (Gly4Ser)2) to at least one of the catalytic domain of a capping enzyme in particular selected in the group consisting of:

    • said catalytic domain of a RNA triphosphatase;
    • said catalytic domain of a guanylyltransferase; and
    • said catalytic domain of a N7-guanine methyltransferase;


      more particularly said catalytic domain of a N7-guanine methyltransferase.


Particularly, the linking peptide can be located N-terminally with respect to said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase selected in the group consisting of T7, T3, SP6, K1-5 and K1E RNA polymerases, and C-terminally with respect to said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, and said catalytic domain of a N7-guanine methyltransferase.


In particular, the N-terminal end of said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase selected in the group consisting of T7, T3, SP6, K1-5 and K1E-RNA polymerases, is linked by covalent linkage, in particular by a linking peptide, to the C-terminal end of one of the catalytic domain selected in the group consisting of:

    • said catalytic domain of a RNA triphosphatase,
    • said catalytic domain of a guanylyltransferase, and
    • said catalytic domain of a N7-guanine methyltransferase;


      particularly said catalytic domain of a N7-guanine methyltransferase.


Preferably, at least said catalytic domain of a poly(A) polymerase is bound by a linking peptide to at least one of the catalytic domain of a capping enzyme in particular selected in the group consisting of:

    • said catalytic domain of a RNA triphosphatase;
    • said catalytic domain of a guanylyltransferase; and
    • said catalytic domain of a N7-guanine methyltransferase;


      more particularly said catalytic domain of a RNA triphosphatase.


Particularly, the linking peptide can be located C-terminally with respect to said catalytic domain of a poly(A) polymerase and N-terminally with respect to said catalytic domain of a capping enzyme, in particular said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase, and said catalytic domain of a N7-guanine methyltransferase, more particularly said catalytic domain of a RNA triphosphatase.


Said catalytic domains of a capping enzyme (in particular of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase), of a DNA-dependent RNA polymerase and of a poly(A) polymerase can also be assembled by specific protein elements, like leucine zippers, like biotinylation domain to one of the catalytic domain (e.g. Avi-tag II (Cronan 1990) or PFB-tag (Wu, Yeung et al. 2002)) and a biotin binding domain to one of the other catalytic domain (e.g. Strep-tag II (Schmidt and Skerra 1993) or Nano-tag (Lamla and Erdmann 2004)) in the chimeric enzyme according to the invention.


In one embodiment of the chimeric enzyme according to the invention, at least two of said catalytic domains can be assembled, by non-covalent linkage, in particular by leucine zippers.


Preferably, at least said catalytic domain of a DNA-dependent RNA polymerase or of a poly(A) polymerase is assembled by non-covalent linkage, in particular by leucine zippers, to at least one of the catalytic domain of a capping enzyme, preferably to at least one of the catalytic domain selected in the group consisting of:

    • said catalytic domain of a RNA triphosphatase;
    • said catalytic domain of a guanylyltransferase; and
    • said catalytic domain of a N7-guanine methyltransferase.


In one embodiment, at least said catalytic domain of a poly(A) polymerase is assembled by non-covalent linkage, in particular by leucine zippers, preferably at its C-terminal end, to at least one of the catalytic domain of a capping enzyme, particularly to at least one of the catalytic domain selected in the group consisting of:

    • said catalytic domain of a RNA triphosphatase;
    • said catalytic domain of a guanylyltransferase; and
    • said catalytic domain of a N7-guanine methyltransferase;


      and more particularly to said catalytic domain of a RNA triphosphatase.


The leucine zippers, which are dimeric coiled-coil protein structures composed of two amphipathic α-helices that interact with each other, are commonly used to homo- or hetero-dimerize proteins (O'Shea, Klemm et al. 1991). Each helices consist of repeats of seven amino acids, in which the first amino-acid (residue a) is hydrophobic, the fourth (residue d) is usually a leucine, while the other residues are polar. The leucine zippers VELCRO ACID-p1 and BASE-p1, which form a parallel heterodimeric two-stranded coiled coil structures, have high propensity to form parallel protein hetero-dimers (O'Shea, Lumb et al. 1993). They have been used to heterodimerize membrane proteins (Chang, Bao et al. 1994, Pashine, Busch et al. 2003), as well as several soluble proteins (Busch, Reich et al. 1998, Busch, Pashine et al. 2002).


Other types of oligomerisation peptide domains can be also considered to generate chimeric enzyme according to the invention, to assemble at least two of said catalytic domains of the chimeric enzyme according to the invention, especially leucine zippers that form antiparallel heteromeric structures, such as the ACID-a1/BASE-a1 (Oakley and Kim 1998), ACID-Kg/BASE-Eg (McClain, Woods et al. 2001), NZ/CZ (Ghosh, Hamilton et al. 2000), ACID-pLL/BASE-pLL (Lumb and Kim 1995), and EE1234L and RR1234L (Moll, Ruvinov et al. 2001) leucine zippers. Disulfide-linked versions of leucine zippers can be also used to generate disulfide coiled coil-bound heterodimeric chimeric enzyme according to the invention (O'Shea, Lumb et al. 1993), as well as interchain disulfide bridges between cysteine residues under oxidizing conditions (Wells and Powers 1986).


At least two of said catalytic domains of a poly(A) polymerase, of a capping enzyme (in particular of a RNA triphosphatase, of a guanylyltransferase, of a N7-guanine methyltransferase), and of a DNA-dependent RNA polymerase can thus be assembled by leucine zippers, in particular leucine zippers that form antiparallel heteromeric structures, such as the ACID-a1/BASE-a1 (Oakley and Kim 1998), ACID-Kg/BASE-Eg (McClain, Woods et al. 2001), NZ/CZ (Ghosh, Hamilton et al. 2000), and ACID-pLL/BASE-pLL leucine zippers, disulfide coiled coil-bound (O'Shea, Lumb et al. 1993), as well as disulfide bridges between cysteine residues (Wells and Powers 1986).


In one embodiment, the chimeric enzyme according to the invention comprises:

    • a RNA binding domain of the wild type lambda N antitermination protein fused to
    • the wild type poly(A) polymerase of the mammalian PAPOLB, vaccinia virus VP55, African Swine Fever Virus C475L, Acanthamoeba polyphaga mimivirus R341, Megavirus chilensis MG561, Saccharomyces cerevisiae, Candida albicans, Pneumocystis carinii, mutant PAPOLA, or a mutant or a derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules; or to
    • the wild type Saccharomyces cerevisiae PAP1 poly(A) polymerase, cytoplasmic mutant of the Saccharomyces cerevisiae PAP1 poly(A) polymerase (wherein the nuclear localization signal is non-functional or deleted) or a mutant or derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules; or to
    • the wild type Schizosaccharomyces pombe PLA1 poly(A) polymerase, cytoplasmic mutant of the Schizosaccharomyces pombe PLA1 poly(A) polymerases (wherein the nuclear localization signal is non-functional or deleted) or a mutant or derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules;
    • the wild type mammalian PAPOLA, cytoplasmic mutant of the mammalian PAPOLA poly(A) polymerase (wherein the nuclear localization signal is non-functional or deleted) or a mutant or derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules


      and, fused to, in particular fused to the amino-terminal end of,
    • the wild type mRNA capping enzyme of the NP868R African Swine Fever virus or a mutant or a derivative thereof, which is able to add a m7GpppN cap at the 5′-terminal end of RNA molecules, in particular the wild type NP868R African swine fever virus capping enzyme, fused to, in particular fused to the amino-terminal end of,
    • the amino-terminal end of, the wild type T7, T3, SP6, K1-5, K1E RNA polymerase or mutant or derivative thereof which is able to synthesize single-stranded RNA complementary in sequence to the double-stranded template DNA in the 5′→3′ direction including the R551 S K1E RNA polymerase mutant,


      in particular via a linker, preferably selected in the group consisting of linking peptide of formula Gly4, (Gly4Ser)1, (Gly4Ser)2 and (Gly4Ser)4, more preferably of formula Gly4, (Gly4Ser)1 or (Gly4Ser)2.


In another embodiment, the chimeric enzyme according to the invention comprises:

    • a RNA binding domain of the wild type lambda N antitermination protein fused to
    • the wild type poly(A) polymerase of the R341 virus or a mutant or a derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules; or
    • the wild type Saccharomyces cerevisiae PAP1 poly(A) polymerase or a cytoplasmic mutant of the Saccharomyces cerevisiae PAP1 poly(A) polymerase (wherein the nuclear localization signal is non-functional) or a mutant or derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules; or
    • the wild type Schizosaccharomyces pombe PLA1 poly(A) polymerase, cytoplasmic mutant of the Schizosaccharomyces pombe PLA1 poly(A) polymerases (wherein the nuclear localization signal is non-functional or deleted) or a mutant or derivative thereof, which is able to catalyze the non-templated addition of adenosine residues from ATP onto the 3′ end of RNA molecules;


      and, fused to, in particular fused to the amino-terminal end of,
    • the wild type mRNA capping enzyme of the NP868R African Swine Fever virus or a mutant or a derivative thereof, which is able to add a m7GpppN cap at the 5′-terminal end of RNA molecules, in particular the wild type NP868R African swine fever virus capping enzyme, fused to, in particular fused to the amino-terminal end of,
    • the amino-terminal end of the wild type K1E RNA polymerase or mutant or derivative thereof which is able to synthesize single-stranded RNA complementary in sequence to the double-stranded template DNA in the 5′→3′ direction including the R551 S K1E RNA polymerase mutant,


      in particular via a linker, preferably selected in the group consisting of linking peptide of formula Gly4, (Gly4Ser)1, (Gly4Ser)2 and (Gly4Ser)4, more preferably of formula Gly4 or (Gly4Ser)2).


The invention also relates to an isolated nucleic acid molecule or a group of isolated nucleic acid molecules, said nucleic acid molecule(s) encoding a chimeric enzyme according to the invention or an isolated nucleic acid molecule encoding a chimeric enzyme, characterized in that its sequence comprises a nucleic acid sequence encoding a RNA-binding domain of a protein-RNA tethering system fused in frame, in particular in the order, to:

    • a nucleic acid sequence encoding at least one catalytic domain of a poly(A) polymerase;
    • a nucleic acid sequence encoding at least one catalytic domain of a capping enzyme; and optionally to
    • a nucleic acid sequence encoding at least one catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase; wherein said RNA-binding domain binds specifically to a RNA element of said protein-RNA tethering system, consisting of a specific RNA sequence and/or structure.


Said group of isolated nucleic molecules encoding a chimeric enzyme according to the invention comprises or consists of all the nucleic acid molecules which are necessary and sufficient to obtain a chimeric enzyme according to the invention by their expression.


As used herein, the term “nucleic acid molecule” any molecules composed of linked nucleotides, encompassing DNA and RNA molecules.


In one embodiment, said group of isolated nucleic acid molecules encoding a chimeric enzyme according to the invention comprises or consists of:

    • a nucleic acid molecule encoding said RNA binding domain of a protein-RNA tethering system, and
    • a nucleic acid molecule encoding at least one catalytic domain of a capping enzyme, in particular at least one catalytic domain of a RNA triphosphatase, at least one catalytic domain of a guanylyltransferase and at least one catalytic domain of a N7-guanine methyltransferase;


      and optionally:
    • a nucleic acid molecule encoding at least one catalytic domain of a poly(A) polymerase; and/or
    • a nucleic acid molecule encoding at least one catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase.


In another embodiment, said group of isolated nucleic acid molecules encoding a chimeric enzyme according to the invention comprises or consists of:

    • a nucleic acid molecule encoding said RNA binding domain of a protein-RNA tethering system,
    • a nucleic acid molecule encoding at least one catalytic domain of a RNA triphosphatase,
    • a nucleic acid molecule encoding at least one catalytic domain of a guanylyltransferase,
    • a nucleic acid molecule encoding at least one catalytic domain of a N7-guanine methyltransferase;


      and optionally:
    • a nucleic acid molecule encoding at least one catalytic domain of a poly(A) polymerase; and/or
    • a nucleic acid molecule encoding at least one catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase.


In one embodiment, the isolated nucleic acid molecule of the invention comprises or consists of a nucleic acid sequence encoding said RNA-binding domain of a protein-RNA tethering system fused in frame, in particular in the order, to:

    • a nucleic acid sequence encoding said catalytic domain of a poly(A) polymerase,
    • a nucleic acid sequence encoding said catalytic domain of a capping enzyme, in particular,
      • a nucleic acid sequence encoding said catalytic domain of a RNA triphosphatase,
      • a nucleic acid sequence encoding said catalytic domain of a guanylyltransferase,
      • a nucleic acid sequence encoding said catalytic domain of a N7-guanine methyltransferase,


        and to
    • a nucleic acid sequence encoding said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase.


Such single nucleic acid sequence has the advantage of facilitating the subunit assembly, since there is only a single open-reading frame.


In one embodiment of the isolated nucleic acid molecule of the invention, its sequence comprises at least one nucleic acid sequence encoding a ribosome skipping motif.


As used herein, the term “ribosome skipping motif” relates to alternate mechanism of translation in which a specific viral peptide prevents the ribosome from covalently linking a new inserted amino-acid, and let it continue translation. This results in apparent co-translational cleavage of the polyprotein.


In particular, said ribosome skipping motif is selected in the group consisting of the 2A sequences from the Foot-and-mouth disease virus Aphtovirus (UniProtKB/Swiss-Prot AAT01756), Avisivirua A (UniProtKB/Swiss-Prot M4PJD6), Duck hepatitis A Avihepatovirus (UniProtKB/Swiss-Prot QOZQM1), Encephalomyocarditis Cardiovirus (UniProtKB/Swiss-Prot Q66765), Cosavirus A (UniProtKB/Swiss-Prot B8XTP8), Equine rhinitis B Erbovirus 1 (UniProtKB/Swiss-Prot Q66776), Seneca Valley Erbovirus (UniProtKB/Swiss-Prot Q155Z9), Hunnivirus A (UniProtKB/Swiss-Prot F4YYF3), Kunsagivirus A (UniProtKB/Swiss-Prot S4VD62), Mischivirus A (UniProtKB/Swiss-Prot I3VR62), Mosavirus A2 (UniProtKB/Swiss-Prot X2L6K2), Pasivirus A1 (UniProtKB/Swiss-Prot 16YOK4), Porcine teschovirus 1 (UniProtKB/Swiss-Prot Q9WJ28), Infectious flacherie Iflavirus (UniProtKB/Swiss-Prot Q70710), Thosea asigna Betatetravirus (UniProtKB/Swiss-Prot Q9YK87), Cricket paralysis Cripavirus (UniProtKB/Swiss-Prot Q9IJX4), Human rotavirus C (UniProtKB/Swiss-Prot Q9PY95), and Lymantria dispar cypovirus 1 (UniProtKB/Swiss-Prot 0911D7),


In particular, said nucleic acid sequence encoding a ribosome skipping motif is selected in the group consisting of the 2A sequences from the Foot-and-mouth disease virus Aphtovirus (also designated as “F2A”, UniProtKB/Swiss-Prot AAT01756) or Porcine teschovirus 1 (also designated as “T2A”, UniProtKB/Swiss-Prot Q9WJ28).


Said nucleic acid sequence encoding a ribosome skipping motif can be localized between any of the sequence encoding said catalytic domain of the chimeric enzyme of the invention.


In one embodiment, said nucleic acid sequence encoding a ribosome skipping motif can be localized between the sequence encoding said catalytic domain of a poly(A) polymerase fused in frame with the sequence encoding:

    • said catalytic domain of a capping enzyme, preferably selected in the group consisting of said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase and said catalytic domain of a N7-guanine methyltransferase, or
    • said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase;


      in particular, fused in frame with the sequence encoding said catalytic domain of a capping enzyme, more particularly said catalytic domain of a RNA triphosphatase.


In another embodiment, said nucleic acid sequence encoding a ribosome skipping motif can be localized between the sequence encoding said catalytic domain of a DNA-dependent RNA polymerase fused in frame with the sequence encoding said catalytic domain of a capping enzyme, in particular selected in the group consisting of said catalytic domain of a RNA triphosphatase, said catalytic domain of a guanylyltransferase and said catalytic domain of a N7-guanine methyltransferase, more particularly fused in frame with the sequence encoding said catalytic domain of a N7-guanine methyltransferase.


In one embodiment, the isolated nucleic acid molecule according to the invention is characterized in that its sequence comprises or consists of

    • a nucleic acid sequence encoding a RNA-binding domain of a protein-RNA tethering system fused in frame, in particular in the order, to:
      • a nucleic acid sequence encoding a catalytic domain of a poly(A) polymerase;
      • a nucleic acid sequence encoding a catalytic domain of a capping enzyme, and optionally to
      • a nucleic acid sequence encoding said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase;


        and
    • a nucleic acid sequence encoding a ribosome skipping motif between said nucleic acid sequence encoding a catalytic domain of a poly(A) polymerase and said nucleic acid sequence encoding one of the catalytic domain selected in the group consisting of:
      • said catalytic domain of a capping enzyme, and
      • said catalytic domain of a DNA-dependent RNA polymerase.


In one embodiment, the isolated nucleic acid molecule according to the invention is characterized in that its sequence comprises or consists of

    • a nucleic acid sequence encoding a RNA-binding domain of a protein-RNA tethering system fused in frame, in particular in the order, to:
      • a nucleic acid sequence encoding a catalytic domain of a poly(A) polymerase;
      • a nucleic acid sequence encoding a catalytic domain of a RNA triphosphatase,
      • a nucleic acid sequence encoding a catalytic domain of a guanylyltransferase,
      • a nucleic acid sequence encoding a catalytic domain of a N7-guanine methyltransferase, and optionally to
      • a nucleic acid sequence encoding said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase;


        and
    • a nucleic acid sequence encoding a ribosome skipping motif between said nucleic acid sequence encoding a catalytic domain of a poly(A) polymerase and said nucleic acid sequence encoding one of the catalytic domain selected in the group consisting of:
      • said catalytic domain of a RNA triphosphatase,
      • said catalytic domain of a guanylyltransferase,
      • said catalytic domain of a N7-guanine methyltransferase, and
      • said catalytic domain of a DNA-dependent RNA polymerase,


        preferably said catalytic domain of a RNA triphosphatase.


In one embodiment, the isolated nucleic acid molecule of the invention encoding a chimeric enzyme, is characterized in that its sequence comprises a nucleic acid sequence encoding a RNA-binding domain of a protein-RNA tethering system fused in frame in the order to:

    • a nucleic acid sequence encoding at least one catalytic domain of a poly(A) polymerase;
    • a nucleic acid sequence encoding at least one catalytic domain of a capping enzyme; and optionally to
    • a nucleic acid sequence encoding said catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase;


      and in that its sequence further comprises a nucleic acid sequence encoding a ribosome skipping motif between said nucleic acid sequence encoding a catalytic domain of a poly(A) polymerase and said nucleic acid sequence encoding at least one catalytic domain of a capping enzyme.


In fact, unexpectedly, the inventor has demonstrated (as illustrated in Example 8) that such nucleic acid molecule allows higher expression rate by a DNA-dependent RNA polymerase than the combination of a nucleic acid molecule encoding a chimeric enzyme comprising at least one domain of a capping enzyme and a DNA-dependent RNA polymerase associated with a nucleic acid molecule encoding a RNA-binding domain of a protein-RNA tethering system fused in frame to a nucleic acid sequence encoding at least one catalytic domain of a poly(A) polymerase.


These results are really surprising and one skilled in the art could have expected to obtain the same expression rate since the components are the same.


In particular, the nucleic acid molecule according to the invention can be operatively linked to at least one, preferably the whole promoter(s) selected from the group consisting of:

    • a promoter for an eukaryotic DNA-dependent RNA polymerase, preferably for RNA polymerase II;
    • the promoter for a bacteriophage DNA-dependant RNA polymerase; and
    • a promoter for said catalytic domain of a DNA-dependent RNA polymerase of the chimeric enzyme of the invention.


The link of the nucleic acid to a promoter for a eukaryotic DNA-dependent RNA polymerase, preferably for RNA polymerase II has notably the advantage that when the chimeric enzyme of the invention is expressed in an eukaryotic host cell, the expression of the chimeric enzymes is driven by the eukaryotic RNA polymerase, preferably the RNA polymerase II. These chimeric enzymes, in turn, can initiate transcription of the transgene. If tissue-specific RNA polymerase II promoters are used, the chimeric enzyme of the invention can be selectively expressed in the targeted tissues/cells.


Said promoter can be a constitutive promoter or an inducible promoter well known by one skilled in the art. The promoter can be developmentally regulated, inducible or tissue specific.


The invention also relates to a vector comprising a nucleic acid molecule according to the invention. Said vector can be appropriated for semi-stable or stable expression.


The invention also relates to a group of vectors comprising said group of isolated nucleic acid molecules according to the invention.


Particularly said vector according to the invention is a cloning or an expression vector.


The invention also relates to a host cell comprising a nucleic acid molecule according to the invention or a vector according to the invention or a group of vectors according to the invention.


The host cell according to the invention can be useful for large-scale protein production.


Preferably, said catalytic domains of the DNA-polymerase RNA polymerase chimeric enzyme according to the invention are from different enzymes than those of the host cell to prevent the competition between the endogenous gene transcription and the transgene transcription.


The invention also relates to a genetically engineered non-human eukaryotic organism, which expresses a chimeric enzyme encoded by the nucleic acid molecule or the group of isolated nucleic acid molecules according to the invention, in particular a chimeric enzyme according to the invention. Said non-human eukaryotic organism can be any non-human animals, plants.


The invention also relates to the use, particularly in vitro or ex vivo, of a chimeric enzyme according to the invention, for the production of RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap and preferably with 3′ poly(A) tail and optionally comprising at least one chemical modification.


The invention also relates to the use, particularly in vitro or ex vivo, of a nucleic acid molecule or a group of isolated nucleic acid molecules according to the invention, for the production of RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap and preferably with 3′ poly(A) tail and optionally comprising at least one chemical modification.


Particularly said RNA molecule is synthetized by a bacteriophage DNA-dependant RNA polymerase.


In fact, the chimeric enzyme according to the invention is suitable for synthetizing a capped RNA with at least one chemical modification.


In particular, the invention relates to the use, particularly in vitro or ex vivo, of a chimeric enzyme according to the invention, for the production of RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap and preferably with 3′ poly(A) tail and comprising at least one chemical modification selected from the group consisting of pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, l-taurinomethyl-4-thio-uridine, pseudouridine, 5-methyl-cytidine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-methyl-pseudoisocytidine, 2-aminopurine, 2, 6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6-dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, 2-methoxy-adenine, inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, I-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.


The invention also relates to the in vitro or ex vivo use of a chimeric enzyme according to the invention or an isolated nucleic acid molecule or a group of isolated nucleic acid molecules according to the invention, for the production of protein, in particular protein of therapeutic interest like antibody, particularly in eukaryotic systems, such as in vitro synthesized protein assay or cultured cells.


The invention also relates to an in vitro or ex vivo method for producing a RNA molecule with a 5′-terminal cap, in particular a 5′-terminal m7 GpppN cap and preferably a 3′ poly(A) tail encoded by a DNA sequence, in a host cell, said method comprising the step of expressing in the host cell a nucleic acid molecule or a group of isolated nucleic acid molecules according to the invention, wherein said DNA sequence is covalently linked to at least one sequence encoding the RNA element of said protein-RNA tethering system, which specifically binds to said RNA-binding domain. As used herein the term “the RNA element of a protein-RNA tethering system which specifically binds to said RNA-binding domain” relates to a RNA sequence, usually forming a stem-loop, which is able to bind with high affinity to the corresponding RNA-binding domain of a protein-RNA tethering system.


Particularly, said DNA sequence is operatively linked to the promoter for a bacteriophage DNA-dependant RNA polymerase or to the promoter for said DNA-dependent RNA polymerase of the chimeric of the invention.


In particular, when the RNA-binding domain of a protein-RNA tethering system is the RNA-binding domain of the lambdoid N antitermination protein-RNA tethering systems, the element, which specifically binds to said RNA-binding domain can be a boxBL and/or a boxBR stem loop RNA structure (Das 1993, Greenblatt, Nodwell et al. 1993, Friedman and Court 1995), including the elements encoded by SEQ ID No 7 and SEQ ID No 38.


In particular, when the RNA-binding domain of a protein-RNA tethering system is the RNA-binding domain of the MS2 coat protein-RNA tethering system, the element, which specifically binds to said RNA-binding domain can be the 19 nucleotide or the 21 nucleotide stem-loop sequences, including the element encoded by SEQ ID No 39 (Peabody 1993, Valegard, Murray et al. 1994, LeGuyer, Behlen et al. 1996, Valegard, Murray et al. 1997) (nucleotides 1748-766 from enterobacteriophage MS2 isolate DL52, NCBI accession number J0966307.1), which contains the initiation codon of the gene for the viral replicase (Valegard, Murray et al. 1994, Valegard, Murray et al. 1997).


In particular, when the RNA-binding domain of a protein-RNA tethering system is the RNA-binding domain of the R17 isolate coat protein-RNA tethering system, the element, which specifically binds to said RNA-binding domain can be the 19 nucleotide or the 21 nucleotides stem-loop (nucleotides 1746-764 from enterobacteriophage R17, NCBI accession number EF108465.1), which contains the initiation codon of the gene for the viral replicase (Carey and Uhlenbeck 1983).


In particular, said DNA sequence is operatively linked to the promoter for a bacteriophage DNA-dependant RNA polymerase or to the promoter for said DNA-dependent RNA polymerase of the chimeric of the invention and covalently linked at its 3′ terminal end to at least one, preferably at least two, at least three and more preferably at least four sequences encoding the element which specifically binds to said RNA-binding domain.


In particular, said method according to the invention further comprises the step of contacting said DNA sequence encoding the RNA molecule with the enzyme of the invention.


In particular, said DNA sequence is operatively linked to the promoter for a bacteriophage DNA-dependant RNA polymerase or to the promoter for said DNA-dependent RNA polymerase of the chimeric of the invention and covalently linked at its 3′ terminal end to at least one sequence encoding the element which specifically binds to said RNA-binding domain covalently linked to a poly(A) track sequence consisting of at least 10, in particular at least 20, 30, and more particularly at least 40 deoxyadenosine residues.


In particular, said poly(A) track sequence can be covalently linked at its 3′ terminal end to a self-cleaving RNA sequence and optionally to a transcription stop sequence.


In particular, said self-cleaving RNA sequence can be the self-cleaving RNA sequence from the group comprising the genomic pseudoknot ribozyme of the hepatitis D virus (Genbank accession number AJ000558.1), antigenomic hepatitis-D Virus pseudoknot ribozyme (Genbank accession number AJ000558.1), tobacco Ringspot Virus satellite hairpin ribozyme (Genbank accession number NC_003889.1) or artificial short hairpin RNA (shRNA).


In particular, said transcription stop sequence can be the bacteriophageT7 phi10 transcription stop sequence (Genbank accession number GU071091.1) or E. coli RNA polymerase rrnB t1 stop (Genbank accession number LN832404.1).


In particular, said method according to the invention can further comprise the step of introducing in the host cell said DNA sequence and/or the nucleic acid according to the invention, using well-known methods by one skilled in the art like by transfection using calcium phosphate, by electroporation or by mixing a cationic lipid with DNA to produce liposomes.


In one embodiment, said method according to the invention further comprises the step of inhibiting, in particular silencing, preferably by siRNA (small interfering RNA), miRNA (microRNA) or shRNA, the cellular transcription and post-transcriptional machineries of said host cell.


In one embodiment, said method according to the invention further comprises the step of inhibiting the expression of the endogenous DNA-dependent RNA polymerase and/or the endogenous capping enzyme in said host cell.


As used herein the term “endogenous DNA-dependent RNA polymerase” relates to the endogenous DNA-dependent RNA polymerase of said host cell. When the host cell is a eukaryotic cell, said endogenous DNA-dependent RNA polymerase is the RNA polymerase II.


As used herein the term “endogenous capping enzyme” refers to the endogenous capping enzyme of said host cell.


As used herein the term “inhibiting the expression of a protein” relates to a decrease of at least 20%, particularly at least 35%, at least 50% and more particularly at least 65%, at least 80%, at least 90% of expression of said protein. Inhibition of protein expression can be determined by techniques well known to one skilled in the art, including but not limiting to Northern-Blot, Western-Blot, RT-PCR.


The step of inhibiting the expression of the endogenous DNA-dependent RNA polymerase and/or the endogenous capping enzyme in said host cell can be implemented by any techniques well known to one skilled in the art, including but not limiting to siRNA techniques that target said endogenous DNA-dependent RNA polymerase and/or the endogenous capping enzyme, antisense RNA techniques that target said endogenous DNA-dependent RNA polymerase and/or the endogenous capping enzyme, shRNA techniques that target said endogenous DNA-dependent RNA polymerase and/or the endogenous capping enzyme.


In addition to siRNA (or shRNA), other inhibitory sequences might be also considered for the same purpose including DNA or RNA antisense (Liu and Carmichael 1994, Dias and Stein 2002), hammerhead ribozyme (Salehi-Ashtiani and Szostak 2001), hairpin ribozyme (Lian, De Young et al. 1999) or chimeric snRNA U1-antisense targeting sequence (Fortes, Cuevas et al. 2003). In addition, other cellular target genes might be considered for inhibition, including other genes involved in the cellular transcription (e.g. other subunits of the RNA polymerase II or transcription factors), post-transcriptional processing (e.g. other subunit of the capping enzyme, as well as polyadenylation or spliceosome factors), and mRNA nuclear export pathway.


In one embodiment of the method according to the invention, said RNA molecule can encode a polypeptide of therapeutic interest.


In another embodiment, said RNA molecule can be a non-coding RNA molecule selected in the group comprising siRNA, ribozyme, shRNA and antisense RNA. In particular, said DNA sequence can encode a RNA molecule selected in the group consisting of mRNA, non-coding RNA, particularly siRNA, ribozyme, shRNA and antisense RNA.


The invention also relates to the use of a chimeric enzyme according to the invention as a capping enzyme and preferably a pol(A) polymerase and a DNA-dependent RNA polymerase.


The invention also relates to a kit for the production of a RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap, comprising at least one chimeric enzyme according to the invention as defined above, and/or an isolated nucleic acid molecule and/or a group of nucleic acid molecule according to the invention as defined above, and/or a vector according to the invention as defined above, or


a chimeric enzyme, in particular a cytoplasmic chimeric enzyme comprising at least one catalytic domain of a RNA triphosphatase, at least one catalytic domain of a guanylyltransferase, at least one catalytic domain of a N7-guanine methyltransferase, and at least one catalytic domain of a DNA-dependent RNA polymerase and/or an isolated nucleic acid molecule and/or a group of isolated nucleic acid molecules encoding said chimeric enzyme and a poly(A) polymerase, in particular a cytoplasmic poly(A) polymerase, comprising at least one RNA-binding domain of a protein-RNA tethering system linked to at least one catalytic domain of said poly(A) polymerase and/or an isolated nucleic acid molecule encoding said poly(A) polymerase;


and optionally a DNA sequence encoded said RNA molecule, which is covalently linked to at least one sequence encoding the RNA element of said protein-RNA tethering system, which specifically binds to said RNA-binding domain.


Particularly, said kit for the production of an RNA molecule with a 5′-terminal cap, comprises a DNA sequence encoded said RNA molecule, which is operatively linked to the promoter for a bacteriophage DNA-dependant RNA polymerase or to the promoter for said DNA-dependent RNA polymerase of the chimeric enzyme of the invention.


In particular, said kit for the production of an RNA molecule with a 5′-terminal cap, comprises at least one chimeric enzyme according to the invention, and/or an isolated nucleic acid molecule and/or a group of isolated nucleic acid molecules according to the invention, and/or a vector according to the invention and optionally a DNA sequence encoded said RNA molecule, which is covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain and particularly which is operatively linked to the promoter for a bacteriophage DNA-dependant RNA polymerase or to the promoter for said DNA-dependent RNA polymerase of the chimeric enzyme of the invention.


In particular, said kit for the production of an RNA molecule with a 5′-terminal cap, further comprises an isolated nucleic acid molecule encoding at least one catalytic domain of a DNA-dependent RNA polymerase, and/or at least one catalytic domain of a DNA-dependent RNA polymerase, in particular of a bacteriophage DNA-dependent RNA polymerase.


Particularly, said kit further comprises its instructions of use. The invention also relates to a composition (in particular a kit or a pharmaceutical composition) comprising:

    • a chimeric enzyme, in particular a cytoplasmic chimeric enzyme, comprising at least one catalytic domain of capping enzyme and at least one catalytic domain of a DNA-dependent RNA polymerase, particularly of a bacteriophage DNA-dependent RNA polymerase and/or an isolated nucleic acid molecule or a group of isolated nucleic acid molecules encoding said chimeric enzyme; and
    • a poly(A) polymerase, in particular a cytoplasmic poly(A) polymerase, comprising at least one RNA-binding domain of a protein-RNA tethering system, particularly of a bacteriophage protein-RNA tethering system, linked to at least one catalytic domain of said poly(A) polymerase and/or an isolated nucleic acid molecule encoding said poly(A) polymerase; and optionally
    • a DNA sequence, which is operatively linked to the promoter for said DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element interacting with high affinity with said RNA-binding domain;


      said composition being useful for the production of a RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap.


In particular, said composition (in particular a kit or a pharmaceutical composition) comprising:

    • a chimeric enzyme, in particular a cytoplasmic chimeric enzyme, comprising at least one catalytic domain of a RNA triphosphatase, at least one catalytic domain of a guanylyltransferase, at least one catalytic domain of a N7-guanine methyltransferase, and at least one catalytic domain of a DNA-dependent RNA polymerase and/or an isolated nucleic acid molecule or a group of isolated nucleic acid molecules encoding said chimeric enzyme; and
    • a poly(A) polymerase, in particular a cytoplasmic poly(A) polymerase, comprising at least one RNA-binding domain of a protein-RNA tethering system linked to at least one catalytic domain of said poly(A) polymerase and/or an isolated nucleic acid molecule encoding said poly(A) polymerase; and optionally
    • a DNA sequence, which is operatively linked to the promoter for said DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element interacting with high affinity with said RNA-binding domain;


      said composition being useful for the production of a RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap.


More particularly, said composition (in particular a kit or a pharmaceutical composition) comprising:

    • a chimeric enzyme, in particular a cytoplasmic chimeric enzyme, comprising the NP868R capping enzyme, and the K1E DNA-dependent RNA polymerase, particularly linked by the (Gly4Ser)2 linker and/or an isolated nucleic acid molecule or a group of isolated nucleic acid molecules encoding said chimeric enzyme; and
    • a poly(A) polymerase, in particular a cytoplasmic poly(A) polymerase comprising at least one catalytic domain of a poly(A) polymerase selected in the group consisting of PAP1, PAPOLA, PAPOLB, VP55, C475L, R341 and MG561 poly(A) polymerase and comprising at least one RNA-binding domain of a protein-RNA tethering system linked to at least one catalytic domain of said poly(A) polymerase and/or an isolated nucleic acid molecule encoding said poly(A) polymerase; and optionally
    • a DNA sequence, which is operatively linked to the promoter for said DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element interacting with high affinity with said RNA-binding domain;


      said composition being useful for the production of a RNA molecule with 5′-terminal cap, in particular 5′-terminal m7GpppN cap.


Advantageously, the kit or the compositions of the invention can be used as an orthogonal gene expression system. As used herein, the term “orthogonal” designate biological systems whose basic structures are independent and generally originates from different species.


The invention also relates to a chimeric enzyme according to the invention, an isolated nucleic acid molecule according to the invention, a group of nucleic acid molecule according to the invention or a vector according to the invention, for its use as a medicament, in particular for the prevention and/or treatment of human or animal pathologies, preferably by means of gene therapy.


The invention also relates to a pharmaceutical composition comprising a chimeric enzyme according to the invention, and/or an isolated nucleic acid molecule according to the invention and/or a group of nucleic acid molecule according to the invention, and/or a vector according to the invention. Preferably, said pharmaceutical composition according to the invention is formulated in a pharmaceutical acceptable carrier.


Pharmaceutical acceptable carriers are well known by one skilled in the art.


The pharmaceutical composition according to the invention can further comprise at least one DNA sequence of interest, wherein said DNA sequence is operatively linked to a promoter for said catalytic domain of a DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain.


Such components (in particular selected in the group consisting of a chimeric enzyme according to the invention, an isolated nucleic acid molecule according to the invention, a vector according to the invention and at least one DNA sequence of interest) can be present in the pharmaceutical composition or medicament according to the invention in a therapeutically amount (active and non-toxic amount).


Such therapeutically amount can be determined by one skilled in the art by routine tests including assessment of the effect of administration of said components on the pathologies and/or disorders which are sought to be prevent and/or to be treated by the administration of said pharmaceutical composition or medicament according to the invention.


For example, such tests can be implemented by analyzing both quantitative and qualitative effect of the administration of different amounts of said aforementioned components (in particular selected in the group consisting of a chimeric enzyme according to the invention, an isolated nucleic acid molecule according to the invention, a vector according to the invention and at least one DNA sequence of interest) on a set of markers (biological and/or clinical) characteristics of said pathologies and/or of said disorders, in particular from a biological sample of a subject.


The invention also relates to a therapeutic method comprising the administration of a chimeric enzyme according to the invention, and/or an isolated nucleic acid molecule according to the invention, and/or a group of nucleic acid molecule according to the invention and/or a vector according to the invention in a therapeutically amount to a subject in need thereof. The therapeutic method according to the invention can further comprise the administration of at least one DNA sequence of interest, wherein said DNA sequence is operatively linked to a promoter for said catalytic domain of a DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain, in a therapeutically amount to a subject in need thereof.


Said chimeric enzyme, nucleic acid molecule and/or said vector according to the invention can be administrated simultaneously, separately or sequentially of said DNA sequence of interest, in particular before said DNA sequence of interest.


The invention also relates to a pharmaceutical composition according to the invention for its use for the prevention and/or treatment of human or animal pathologies, in particular by means of gene therapy.


Said pathologies can be selected from the group consisting of pathologies, which can be improved by the administration of said at least one DNA sequence of interest.


The invention also relates to the use of a chimeric enzyme according to the invention, and/or an isolated nucleic acid molecule according to the invention, and/or a group of nucleic acid molecule according to the invention and/or a vector according to the invention, for the preparation of a medicament for the prevention and/or treatment of human or animal pathologies, in particular by means of gene therapy.


The invention also relates to a first combination product, which comprises as active ingredients:

    • at least one chimeric enzyme according to the invention and/or at least one nucleic acid molecule according to the invention and/or a group of nucleic acid molecule according to the invention and/or a at least one vector comprising and/or expressing a nucleic acid molecule according to the invention; and
    • at least one DNA sequence of interest, wherein said DNA sequence is operatively linked to a promoter for said catalytic domain of a DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain;


      for its use as a medicament, wherein said active ingredients are formulated for separate, simultaneous or sequential administration.


The invention also relates to a second combination product, which comprises as active ingredients:

    • a chimeric enzyme, in particular a cytoplasmic chimeric enzyme, comprising at least one catalytic domain of a RNA triphosphatase, at least one catalytic domain of a guanylyltransferase, at least one catalytic domain of a N7-guanine methyltransferase, and at least one catalytic domain of a DNA-dependent RNA polymerase and/or an isolated nucleic acid molecule or a group of isolated nucleic acid molecules encoding said chimeric enzyme; and
    • a poly(A) polymerase, in particular a cytoplasmic poly(A) polymerase, comprising at least one RNA-binding domain of a protein-RNA tethering system linked to at least one catalytic domain of said poly(A) polymerase and/or an isolated nucleic acid molecule encoding said poly(A) polymerase; and
    • a DNA sequence, which is operatively linked to the promoter for said DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain;


for its use as a medicament, wherein said active ingredients are formulated for separate, simultaneous or sequential administration.


Said DNA sequence of interest can be an anti-oncogene (a tumor suppressor gene).


Said DNA sequence of interest can encode a polypeptide of therapeutic interest or a non-coding RNA selected in the group comprising siRNA, ribozyme, shRNA and antisense RNA.


Said polypeptide of therapeutic interest can be selected from, a monoclonal antibody or its fragments, a growth factor, a cytokine, a cell or nuclear receptor, a ligand, a coagulation factor, the CFTR protein, insulin, dystrophin, a hormone, an enzyme, an enzyme inhibitor, a polypeptide which has an antineoplastic effect, a polypeptide which is capable of inhibiting a bacterial, parasitic or viral, in particular HIV, infection, an antibody, a toxin, an immunotoxin.


Preferably, the combination product according to the invention can be formulated in a pharmaceutical acceptable carrier.


In one embodiment of the combination product according to the invention, said vector is administrated before said DNA sequence of interest.


The invention also relates to a combination product according to the invention for its use as a medicament in the prevention and/or treatment of human or animal pathologies, particularly by means of gene therapy.


Said pathologies can be selected from the group consisting of pathologies, which can be improved by the administration of at least one DNA sequence of interest, as described above.


For example, said pathologies, as well as their clinical, biological or genetic subtypes, can be selected from the group comprising liver disorders (e.g. acute liver failure due to acetaminophen intoxication or other causes, prevention of liver failure post-hepatectomy, liver primary cancers including hepatoma or cholangiocarcinoma, nonalcoholic steatohepatitis, as well as liver monogenic disorders such as hemochromatosis, ornithine transarbamylase deficiency, argininosuccinatelyase deficiency, argininosuccinate synthetase 1, hemochromatosis or Wilson's disease), disorders due or associated to deficiencies of secreted proteins (e.g. lysosomal storage diseases such as Gaucher's disease, Niemann-Pick disease, Tay-Sacks or Sandhoff disease, Hunter syndrome, or Hurler disease; deficiencies of coagulation factors including factors VIIIc, IX, Von Willebrand, fibrinogen or other coagulation proteins, as well as colony stimulating factors including erythropoietin, granulocyte colony stimulating factor and thrombopoietin), cancers and their predisposition (e.g. breast, colorectal, pancreas, gastric, esophageal and lung cancers, as well as melanoma), malignant hemopathies (e.g. leukemias, Hodgkin's and non-Hodgkin's lymphomas, myeloma), hemoglobinopathies (e.g. sickle cell anemia, glucose-6-phosphate dehydrogenase deficiency) and thalassemias, autoimmune disorders (e.g. systemic lupus erythematosus, scleroderma, autoimmune hepatitis), cardiovascular disorders (e.g. cardiac rhythm and conduction disorders, hypertrophic cardiomyopathy, cardiovascular disease, or chronic cardiac failure), metabolic disorders (e.g. type I and type II diabetes mellitus and their complications, dyslipidemia, atherosclerosis and their complications), infectious disorders (e.g. AIDS, viral hepatitis B, viral hepatitis C, influenza flu, Zika, Ebola and other viral diseases; botulism, tetanus and other bacterial disorders; malaria and other parasitic disorders), muscular disorders (e.g. Duchenne muscular dystrophy and Steinert myotonic muscular dystrophy), respiratory diseases (e.g. cystic fibrosis, alpha-1 antitrypsin deficiency, acute respiratory distress syndrome, pulmonary arterial hypertension, pulmonary veno-occlusive disease), renal diseases (e.g. polycystic kidney disease, glomerulopathy), colorectal disorders (e.g. Crohn's disease and ulcerative colitis), ocular disorders especially retinal diseases (e.g. Leber's amaurosis, retinitis pigmentosa, age related macular degeneration), central nervous system disorders (e.g. Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, Huntington's disease, neurofibromatosis, adrenoleukodystrophy, bipolar disease, schizophrenia and autism), bone and joint disorders (e.g. rheumatoid arthritis, ankylosing spondylitis, osteoarthritis) and skin and connective tissue disorders (e.g. neurofibromatosis and psoriasis).


In one embodiment, the combination product of the invention comprises:

    • at least one vector comprising and expressing a nucleic acid molecule according to the invention, wherein said catalytic domain of a DNA-dependent RNA polymerase is a catalytic domain of a bacteriophage DNA-dependent RNA polymerase; and
    • at least one DNA sequence of interest, wherein said DNA sequence is operatively linked to a promoter for said catalytic domain of a bacteriophage DNA-dependent RNA polymerase and covalently linked to at least one sequence encoding the element which specifically binds to said RNA-binding domain.


The invention also relates to a method for producing the chimeric enzyme according to the invention comprising the step of expressing in at least one host cell said nucleic acid molecule or said group of nucleic acid molecules encoding the chimeric enzyme of the invention in conditions allowing the expression of said nucleic acid molecule(s) in said host cell.


The invention also relates to a method for producing the chimeric enzyme according to the invention comprising the steps of:

    • expressing a part of said group of nucleic acid molecules encoding a chimeric enzyme of the invention in a first host cell in conditions allowing the expression of said nucleic acid molecules in said host cell, to obtain a first part of the chimeric enzyme of the invention;
    • expressing the other part of said group of nucleic acid molecules encoding the chimeric enzyme of the invention in a second host cell in conditions allowing the expression of said nucleic acid molecules in said host cell to obtain a second part of the chimeric enzyme of the invention; and
    • assembling said first part and said second part to obtain the chimeric enzyme of the invention.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1: maps of the pK1ERNAP and pT3RNAP plasmids.



FIG. 2: map of the test tethered pNλ-D12L plasmid. Other test tethered test plasmids have the same general design, except ORFs which were substituted by endonuclease restriction digestion and ligation.



FIG. 3: map of the test pD12L plasmid encoding untethering vaccinia virus capping enzyme D12L subunit. Other untethering test plasmids have the same design, except ORFs which were substituted by endonuclease restriction digestion and ligation.



FIG. 4: maps of untethered pK1Ep-Luciferase and pT3p-Luciferase reporter plasmids without 4xλBoxBr repeat.



FIG. 5: maps of NA-tethered pK1Ep-Luciferase-4xλBoxBr and pT3p-Luciferase-4xλBoxBr reporter plasmids with 4xλBoxBr repeat.



FIG. 6: K1ERNAP or T3RNAP-driven expression systems used to test the activity of NA-tethering system coupled to the vaccinia virus capping enzyme on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA.



FIG. 7: K1ERNAP or T3RNAP-driven expression systems used to test the activity of NA-tethering system coupled to the African swine fever virus capping enzyme on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA.



FIG. 8: K1ERNAP-driven expression system used to assay the activity of various protein:RNA binding systems coupled to the African swine fever virus capping enzyme on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA.



FIG. 9: K1ERNAP-driven expression system used to assay the activity of various protein:RNA binding systems coupled to the vaccinia virus capping enzyme on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA.



FIG. 10: structure of constructions resulting of Nλ-tethering domain coupled to the African swine fever virus capping enzyme fused to poly(A) polymerases. (A) Nλ-poly(A) polymerase-G4-NP868R monomer, (B) Nλ-NP868R-G4-poly(A) polymerase monomer.



FIG. 11: K1ERNAP-driven expression system used to assay the activity of Nλ-tethering system coupled to the African swine fever virus capping enzyme fused to poly(A) polymerases on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA.



FIG. 12: structure of constructions resulting of Nλ-tethering domain coupled to the vaccinia virus capping enzyme fused to poly(A) polymerases. Arrow indicates D12L-D1R binding. (A) Nλ-poly(A) polymerase-G4-D12L/D1R heterodimer, (B) Nλ-D12L-G4-poly(A) polymerase/D1R heterodimer.



FIG. 13: K1ERNAP-driven expression system used to assay the activity of Nλ-tethering system coupled to the vaccinia virus capping enzyme fused to poly(A) polymerases on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA



FIG. 14 structure of Nλ-tethering system coupled to the vaccinia virus capping enzyme bound to poly(A) polymerases through complementary leucine zippers. Arrow indicates complementary leucines zippers that forms heterodimers. (A) Nλ-R341-EE1234L/RR1234L-NP868R heterodimer, and (B) Nλ-NP868R-EE1234L/RR1234L-R341 heterodimer.



FIG. 15: K1ERNAP-driven expression system used to assay the activity of Nλ-tethering system coupled to the vaccinia virus capping enzyme fused to poly(A) polymerases on expression of uncapped 4xλBoxBr-tethered Firefly Luciferase mRNA. Arrow indicates D12L-D1R binding.



FIGS. 16A-D: structures of tethering complexes between the Acanthamoeba polyphaga mimivirus poly(A) polymerase R341, the African swine fever virus NP868R capping enzyme and the phage K1E RNA polymerase on expression of polyadenylated 4xλBoxBl-tethered Firefly Luciferase mRNA. [X1] and [X2] designate variable domains, where G4 and (G4S)2 is a flexible linker, T2A and F2A are ribosomal skipping sequences from the porcine teschovirus-1 and picornavirus Foot-and-mouth disease aphtovirus, respectively.



FIG. 17: expression system used to assay the activity of tethering complexes between the Acanthamoeba polyphaga mimivirus poly(A) polymerase R341, the African swine fever virus NP868R capping enzyme and the phage K1E RNA polymerase on expression of 4xλBoxBl-tethered Firefly Luciferase mRNA.





The present invention will be explained in detail with examples in the following, but the technical scope of the present invention is not limited to these examples.


Example 1: D1R/D12L, the Vaccinia Virus Capping Enzyme Tethered to Luciferase Reporter mRNA Increases its Expression

1. Objectives


The objective of this experiment was to determine if the heterodimeric vaccinia virus capping enzyme appropriately tethered to Firefly Luciferase reporter mRNA synthesized in cellulo increases its expression.


The vaccinia virus capping enzyme consist of two subunits, which form a heterodimer: (i) a 95 kDa subunit encoded by the vaccinia virus D1R gene (genomic sequence AY243312.1; UniProtKB/Swiss-Prot accession number P04298), designated hereafter as D1R, which has RNA-triphosphatase, RNA guanylyltransferase and RNA N7-guanine methyltransferase enzymatic activities (Cong and Shuman 1993, Niles and Christen 1993, Higman and Niles 1994, Mao and Shuman 1994, Gong and Shuman 2003), (ii) and a 31-kDa subunit encoded by the vaccinia virus D12L gene (genomic sequence AY243312.1; UniProtKB/Swiss-Prot accession number P04318), designated hereafter as D12L, which has no intrinsic enzymatic activity, but enhances the RNA N7-guanine methyltransferase activity of the D1R subunit (Higman, Bourgeois et al. 1992, Higman, Christen et al. 1994, Mao and Shuman 1994). Cotransfection of plasmids encoding these two subunits therefore generate in cellulo the heterodimer D1R/D12L capping enzyme, which can eventually fused to a protein tethering domain.


2. Methods


a. Plasmids


The coding sequences of the following plasmids were optimized for expression in human cells with respect to codon adaptation index using the GeneOptimizer algorithm (Raab, Graf et al. 2010). All gene sequences were artificially synthesized and assembled from stepwise PCR using oligonucleotides, cloned and fully sequenced.


For all the following examples, the conditions tested consist of a variable combination of several plasmids. In the present example, the pK1ERNAP/pT3RNAP plasmids together with Firefly luciferase reporter plasmids were used to generate in cellulo the Firefly Luciferase mRNA with or without tethering domain, which then can be specifically modified by enzyme produced by the test plasmid appropriately tethered to the Firefly Luciferase mRNA.


The expression plasmids consisted of the phage T3 and K1E RNA polymerase open reading-frames (ORFs), which were subcloned in the pCMVScript plasmid backbone (Stratagene, La Jolla, Calif.), following the removal of the T7 φ10 promoter sequence. These corresponding plasmids, designated as p-followed by the name of the ORF, have the following design (i.e. pK1ERNAP or pT3RNAP; FIG. 1): IE1 promoter/enhancer from the human cytomegalovirus (CMV), 5′-untranslated region (5′-UTR), Kozak consensus sequence, ORFs, 3′-untranslated region (3′-UTR), and SV40 polyadenylation signal. The corresponding ORFs were subcloned by digestion at endonuclease restriction enzyme sites immediately upstream to the Kozak sequence and downstream to stop codon.


The test plasmids contained the coding sequence of the capping enzymes under investigation with (FIG. 2 for pNλ-D12L) or without peptide tethering domain (FIG. 3 for pD12L). Peptide tethering domain consist of the 22 amino-acids of antitermination N proteins from the lambda bacteriophage (Nλ; amino-acids 1-22 from Enterobacteria phage lambda nucleocapsid protein AAA32249; SEQ ID No 1 and SEQ ID No 2 corresponding to the nucleotide and amino acid sequences of N-terminal tethering domain from antitermination N protein from A bacteriophage, respectively) was fused to the amino-terminal ends of the D12L protein through a flexible G4 linker. The corresponding ORFs were subcloned by digestion at endonuclease restriction enzyme sites immediately upstream to Kozak sequence or downstream to Nλ-G4 motif, and downstream to stop codon. Two pairs of plasmids were used to encode the tethered or untethered heterodimeric vaccinia virus capping enzyme. Firstly, plasmids containing the D12L coding sequences with the NA tethering domain (pNλ-D12L; SEQ ID No 3 and SEQ ID No 4 corresponding to the nucleotide and amino acid sequences of D12L subunit of vaccinia virus capping enzyme, respectively) and wild-type D1R coding sequence (pD1R; SEQ ID No 5 and SEQ ID No 6 corresponding to the nucleotide and amino acid sequences of D1R subunit of vaccinia virus capping enzyme, respectively), which generate the tethered D1R/Nλ-D12L heterodimer. Secondly, plasmids containing the wild-type D12L coding sequences without the NA tethering domain (pD12L) and wild-type D1R coding sequence (pD1R), which form the untethered D1R/D12L heterodimer.


The Firefly Luciferase reporter plasmids containing the Firefly Luciferase gene under control of the K1E or T3 RNA polymerase promoters, contained a 5′-UTR sequence, Kozak consensus sequence followed by the ORF of Luciferase gene from Photinus pyralis and stop codon, RNA tethering domain consisting of four BoxBr in tandem from λ virus (optional, lacking in the untethered version; nucleotides 38312-38298 of genomic sequence of Enterobacteria phage lambda KT232076.1; SEQ ID No 7 corresponding to the nucleotide sequence of the BoxBr RNA stem-loops from A bacteriophage), poly(A) track of 40 adenosine residues, followed by a self-cleaving RNA sequence from the genomic ribozyme of the hepatitis D virus, and terminated by the bacteriophage T7 φ10 transcription stop. These plasmids were designated either pK1Ep-Luciferase/pT3p-Luciferase in their untethered versions (FIG. 4), or pK1Ep-Luciferase-4xλBoxBr/pT3p-Luciferase-4xλBoxBr (FIG. 5) in their 4xBoxBr RNA tethered versions. The RNA molecules produced by this system are therefore uncapped and have a short 3′-end polyadenylation track encoded by 40 adenosine residues in the template Firefly Luciferase reporter plasmids.


b. Cell Culture and Transfection


For standard experiments, the Human Embryonic Kidney 293 (HEK-293, ATCC CRL 1573) were routinely grown at 37° C. in 5% CO2 atmosphere at 100% relative humidity. Cells were maintained in Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 4 mM L-alanyl-L-glutamine, 10% fetal bovine serum (FBS), 1% non-essential amino-acids, 1% sodium pyruvate, 1% penicillin and streptomycin, and 0.25% fungizone.


Cells were routinely plated in 24-well plates at 1×105 cells per well the day before transfection and transfected at 80% cell confluence. Transient transfection was performed with Lipofectamine 2000 reagent (Invitrogen, Carlsbad, Calif.) according to manufacturer's recommendations. Except otherwise stated, cells were transfected with 2 μl of Lipofectamine 2000 and 0.8 μg of total plasmid DNA. For standard luciferase and hSEAP gene reporter expression assays, cells were analyzed 48 hours after transfection, except otherwise stated.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Luciferase luminescence was assayed by the Luciferase Assay System (Promega, Madison, Wis.) according to the manufacturer's recommendations. In brief, cells were lysed in Cell Culture Lysis Reagent buffer (CLR), and then centrifuged at 12,000×g for two minutes at 4° C. Luciferase Assay Reagent (Promega; 100 μl/well) diluted at 1:10 was added to supernatant (20 μl/well). Luminescence readout was taken on a Tristar 2 microplate reader (Berthold, Bad Wildbad, Germany) with a read time of one second per well.


In order to normalize for transfection efficacy, cells were transfected with the pORF-eSEAP plasmid (InvivoGen, San Diego, Calif.), which encodes for the human secreted embryonic alkaline phosphatase (hSEAP) driven by the EF-1α/HTLV composite promoter. Enzymatic activity was assayed in cell culture medium using the Quanti-Blue colorimetric enzyme assay kit (InvivoGen). Gene reporter expression was expressed as the ratio of luciferase luminescence (RLU, relative light units) to eSEAP absorbance (OD, optic density).


d. Statistical Analysis


Statistical analyses were performed with paired two-tailed Student's t-test. Results are means (n≥4)±standard deviation. P-value<0.05 was considered statistically significant.


3. Results


In this set of experiments, the Firefly Luciferase mRNA was produced by the phage K1E or T3 RNA polymerases by cotransfection of pK1ERNAP/pT3RNAP and pK1Ep-Luciferase-4xλBoxBr/pT3p-Luciferase-4xλBoxBr for the tethered version of the Firefly Luciferase reporter plasmids or pK1Ep-Luciferase-4xλBoxBr/pT3p-Luciferase-4xλBoxBr for their untethered version. The test plasmids contain the coding sequence of the D1R/D12L vaccinia virus with or without tethering domain were co-transfected. The translatability of the resulting transcripts, which is expected to increase in case of proficient capping, is measured by the Firefly Luciferase assay. A general depiction of the assay is shown FIG. 6.


Results of the first set of experiments with the K1E-driven system are shown in the table below:














Plasmids
mean
SEM







(1) pK1ERNAP, pK1Ep-Luciferase
35 329
3 113


(2) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBr
34 784
4 388


(3) pK1ERNAP, pD1R, pK1Ep-Luciferase-
315 159 
36 188 


4xλBoxBr




(4) pK1ERNAP, pD12L, pK1Ep-Luciferase-
60 212
2 219


4xλBoxBr




(5) pK1ERNAP, pD1R, pD12L, pK1Ep-
851 056 
144 590 


Luciferase-4xλBoxBr




(6) pK1ERNAP, pD1R, pNλ-pD12L, pK1Ep-
2 237 689  
92 709 


Luciferase-4xλBoxBr




(7) Baseline
14 537
3 145









As expected when capping is lacking, Firefly Luciferase mRNA generated by pK1Ep-Luciferase and pK1Ep-Luciferase-4xλBoxBr cotransfected with the K1ERNAP plasmid alone (designated pK1ERNAP) was poorly expressed (row 1 and 2). Cotransfection of the untethered D1R plasmid (designated pD1R) together with pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr increased the expression by approximately 9-fold in comparison to cotransfection of pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr plasmids only (row 3 vs. 1 or 2, p<0.05, two-way Student t-test), whereas the transfection of the untethered pD12L plasmid with pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr had virtually no effect on Firefly Luciferase expression (row 4 vs. 1 or 2, p=NS, two-way Student t-test). The cotransfection of the untethered pD12L and pD1R plasmids together with pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr, which result in the vaccinia virus D1R/D12L capping enzyme heterodimer without tethering domain, significantly increased the expression of Firefly Luciferase in comparison to previous conditions, therefore confirming that mRNA capping is requested for mRNA translation (row 5 vs. 1 to 4, p=NS, two-way Student t-test p<0.05, two-way Student t-test). Finally, cotransfection of the tethered D12L plasmid (pNλ-D12L) and D1R together with pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr, which produces the tethering vaccinia virus capping enzyme D1R/Nλ-D12L, increased drastically the expression of the Luciferase mRNA by 63.3 (row 6 vs. 2) and 2.6-fold (row 6 vs. 5) in comparison to no vaccinia virus capping enzyme (pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr alone), or untethering vaccinia virus capping enzyme (pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr/pD1R/pD12L), respectively.


In a second set of experiments, the Firefly Luciferase mRNA was produced by the phage T3 RNA polymerase and tethered to the vaccinia virus capping enzyme. Results of the second set of experiments with the T3-driven system are shown in the table below:














Plasmids
mean
SEM







(1) pT3RNAP, pT3p-Luciferase
40 387
5 041


(2) pT3RNAP, pT3p-Luciferase-4xλBoxBr
43 682
2 072


(3) pT3RNAP, pD1R, pT3p-Luciferase-4xλBoxBr
82 445
6 913


(4) pT3RNAP, pD12L, pT3p-Luciferase-4xλBoxBr
47 167
1 342


(5) pT3RNAP, pD1R, pD12L, pT3p-Luciferase-
110 371 
5 390


4xλBoxBr




(6) pT3RNAP, pD1R, pNA-pD12L, pT3p-Luciferase-
353 096 
12 560 


4xλBoxBr




(7) Baseline
 7 269
1 901









This second set of experiments gave very similar results with cotransfection results in the following order: RNA with no 4xλBoxBr and no capping enzyme (row 1, pT3RNAP/pT3p-Luciferase) 4xλBoxBr-RNA and no capping enzyme (row 2, pT3RNAP/pT3p-Luciferase-4xλBoxBr) 4xλBoxBr-RNA with D12L subunit alone (row 4, pT3RNAP/pD12L/pT3p-Luciferase-4xλBoxBr)<4xλBoxBr-RNA with D1R (row 3, pT3RNAP/pD1R/pT3p-Luciferase-4xλBoxBr)<4xλBoxBr-RNA with untethered D1R/D12L capping enzyme (row 5, pT3RNAP/pD1R/pD12L/UpT3p-Luciferase-4xλBoxBr)<<4xλBoxBr-RNA with tethered D1R/D12L capping enzyme (row 6, pT3RNAP/pD1R/pNλ-D12L/UpT3p-Luciferase-4xλBoxBr). The expression levels of this latter condition was statistically greater than all other conditions, especially 3.2 fold higher than with the untethered D1R/D12L capping enzyme, therefore demonstrating the importance of guiding the D1R/D12L capping enzyme to the target reporter mRNA by tethering domain (p<0.05, two-way Student t-test).


4. Conclusions


These experiments show that the vaccinia virus capping enzyme, which contains no known or demonstrated binding domain for a specific RNA sequence, drastically increases gene Firefly Luciferase reporter expression when appropriately tethered to uncapped and polyadenylated Firefly Luciferase reporter mRNA by the Nλ-BoxBr tethering system.


Example 2: NP868R, the African Swine Fever Virus Capping Enzyme Tethered to Luciferase Reporter mRNA Increases its Expression

1. Objectives


The objective of this set of experiments was to demonstrate if NP868R, the African swine fever virus capping enzyme, appropriately tethered to polyadenylated Firefly Luciferase reporter mRNA increases its expression in cellulo. NP868R (also named G4R) is a single-unit 868 amino-acids protein, which all enzymatic activities required for cap-0 formation demonstrated in vitro, i.e. RNA-triphosphatase, RNA guanylyltransferase and RNA N7-guanine methyltransferase (Pena, Yanez et al. 1993, Jais 2011, Dixon, Chapman et al. 2013, Jais, Decroly et al. 2018).


2. Methods


a. Plasmids


The expression (pK1ERNAP or pT3RNAP), as well as the Firefly Luciferase reporter plasmids in their tethered versions (pK1Ep-Luciferase and pT3p-Luciferase) or untethered versions (pK1Ep-Luciferase-4xλBoxBr and pT3p-Luciferase-4xλBoxBr) were the same as described above.


The test plasmid consisted of the coding sequence from the African swine fever virus NP868R capping enzyme (NCBI ASFV genomic sequence strain BA71V NC_001659; UniProtKB/Swiss-Prot accession number P32094; SEQ ID No 8 and SEQ ID No 9 corresponding to the nucleotide and amino acid sequences of African swine fever virus NP868R capping enzyme, respectively) with (pNλ-NP868R) or without the NA tethering domain (pNP868R) subcloned in the pCMVScript plasmid backbone as described above.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


The design of the assay was very similar to Example 1, except that the single subunit capping enzyme NP868R was used instead of the heterodimeric D1R/D12L capping enzyme. In brief, uncapped but polyadenylated Firefly Luciferase mRNA was synthesized in cellulo by the phage K1E or T3 RNA polymerases and its expression in presence of NP868R was assayed (FIG. 7).


Results of the first set of experiment with the K1E-driven system are shown in the table below:














Plasmids
mean
SEM







(1) pK1ERNAP, pK1Ep-Luciferase
  109 401
7 601


(2) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBr
  147 494
6 310


(3) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
2 433 576
226 803 


4xλBoxBr




(4) pK1ERNAP, pNλ-pNP868R, pK1Ep-
4 099 936
465 513 


Luciferase-4xλBoxBr




(5) Baseline
  10 393
2 302









In this first set of experiments, cells were cotransfected with pK1ERNAP/pK1Ep-Luciferase or pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr plasmids, showed low levels of Firefly Luciferase reporter expression (row 1 and 2). Cotransfection of the pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr with the untethered pNP868R plasmid (pNP868R) increased the expression by approximately 29-fold in comparison to pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr alone (row 3 vs. 1 or 2 respectively, p<0.05, two-way Student t-test), therefore confirming that mRNA capping is requested for mRNA translation. Finally, cotransfection of pK1ERNAP/pK1Ep-Luciferase-4xλBoxBr with tethered NP868R (pNλ-NP868R) even increased by 2.1-fold the expression of Firefly Luciferase in comparison to untethered NP868R condition, demonstrating the importance of guiding the enzyme to the target mRNA by tethering domains for proficient mRNA capping (raw 4 vs. 3, p<0.05, two-way Student t-test).


Results of the second set of experiments with the T3-driven system are shown in the table below:














Plasmids
mean
SEM







(1) pT3RNAP, pT3p-Luciferase
75 065
7 575


(2) pT3RNAP, pT3p-Luciferase-4xλBoxBr
62 220
5 781


(3) pT3RNAP, pNP868R, pT3p-Luciferase-
122 957 
24 166 


4xλBoxBr




(4) pT3RNAP, pNλ-NP868R, pT3p-Luciferase-
352 978 
16 813 


4xλBoxBr




(5) Baseline
23 464
6 302









In this second set of experiments, the Firefly Luciferase mRNA was produced by the phage T3 RNA polymerase and tethered to the African swine fever virus capping enzyme. This second set of experiments gave very similar results with cotransfection results in the following order: RNA with no 4xλBoxBr and no capping enzyme (row 1, pT3RNAP/pT3p-Luciferase) 4xλBoxBr-RNA without capping enzyme (row 2, pT3RNAP/pT3p-Luciferase-4xλBoxBr)<4xλBoxBr-RNA with untethered NP868R capping enzyme (row 3, pT3RNAP/pNP868R/pT3p-Luciferase-4xλBoxBr)<<4xλBoxBr-RNA with tethered NP868R capping enzyme (row 4, pT3RNAP/pNλ-NP868R/pT3p-Luciferase-4xλBoxBr). The expression levels of this last condition was statistically greater than all other conditions, especially 2.9 fold higher than with the untethered NP868R capping enzyme, therefore demonstrating the importance of tethering NP868R to the target mRNA by tethering domains for proficient mRNA capping (row 4 vs.3, p<0.05, two-way Student t-test).


4. Conclusions


These experiments show that another capping enzyme, NP868R from the African swine fever virus, which contains no known or predicted binding domain for a specific RNA sequence, increases Firefly Luciferase reporter expression when appropriately tethered to uncapped and polyadenylated reporter mRNA by the Nλ-BoxBr tethering system.


Example 3: Various Protein:RNA Tethering Systems, Coupled to African Swine Fever Virus Capping Enzyme NP868R can Increase the Expression of Luciferase Reporter mRNA Produced by K1E Phage RNA Polymerase in Host-Cell Cytoplasm

1. Objectives


The objectives of the present experiments were to investigate if other protein:RNA tethering systems than the Nλ-4xBoxBr system can guide the African swine fever virus capping enzyme NP868R in order to increase the expression of appropriately tethered Luciferase reporter mRNA produced by the phage K1E RNA polymerase.


The following tethering systems were presently tested: i) MS2 protein and the RNA stem loop tethered sequence from MS2 virus (Valegard, Murray et al. 1994, Valegard, Murray et al. 1997), ii) NA peptide from the lambda virus and its BoxBl RNA tethered sequence (Das 1993, Greenblatt, Nodwell et al. 1993, Friedman and Court 1995), iii) NA peptide from the P22 lamboid virus and its BoxBr RNA tethered sequences (Das 1993, Greenblatt, Nodwell et al. 1993, Friedman and Court 1995), iv) NA peptide from the ϕ21 lamboid virus and its BoxBr RNA tethered sequence (Das 1993, Greenblatt, Nodwell et al. 1993, Friedman and Court 1995), v) TAT binding domain from the Human immunodeficiency virus-1 (HIV-1), which contains a biologically validated nuclear localization signal (Duconge and Toulme 1999), and TAR RNA tethered sequence (Dingwall, Ernberg et al. 1990, Weeks, Ampe et al. 1990, Karn, Dingwall et al. 1991, Puglisi, Tan et al. 1992, Frankel and Young 1998), and vi) the human small nuclear ribonucleoprotein U1 subunit 70 (SNRNP70) protein tethering sequence (Romac, Graff et al. 1994), which contains a biologically validated nuclear localization signal (Keene, Query et al. 1999), and its U1snRNA-stem loop tethered sequence.


2. Methods


a. Plasmids


The pK1ERNAP expression plasmid was described in Example 1.


The test plasmids consisted of the coding sequence of the African swine fever virus NP868R capping enzyme fused at its amino-terminal end to: i) bacteriophage N-antitermination protein the N-terminus of the entire MS2 protein (pMS2-NP868R, NCBI accession number NC_001417.2, UniProtKB/Swiss-Prot P03612; SEQ ID No 36 and SEQ ID No 37 corresponding to the nucleotide and amino-acid sequences, respectively), ii) N-terminal peptide from lambda bacteriophage previously described, iii)N-terminal peptide from P22 bacteriophage N-antitermination protein (pP22N-NP868R, UniProtKB/Swiss-Prot P04891), iv)N-terminal peptide from ϕ21 bacteriophage N-antitermination protein (pNϕ21-NP868R, UniProtKB/Swiss-Prot P07243), v) the TAT protein binding domain from HIV-1 isolate HXB2 (pTAT-NP868R, NCBI reference sequence: AAB50256.1), and vi) human small nuclear ribonucleoprotein U1 subunit 70 (SNRNP70) RNA-binding protein sequence (pSNRNP70-NP868R, amino-acid 92-202, NCBI accession number NM_003089.5).


The RNA tethering domains of the Firefly Luciferase reporter plasmids substituted by four tandem repeats of: i) MS2 RNA stem-loops (pK1Ep-Luciferase-4xMS2sl plasmid; nucleotides 1748-766 from Enterobacteriophage MS2 isolate DL52, NCBI accession number J0966307.1; SEQ ID No 38), ii) BoxBl RNA sequence from A virus (pK1Ep-Luciferase-4xABoxBl; NCBI accession number J02459.1 nucleotides 35518-35534), iii) λBoxBr RNA sequence from P22 lamboid virus (pK1Ep-Luciferase-4xP22BoxBr; NCBI accession number NC_002371.2, nucleotides 31,953-31,971), iv) λBoxBr RNA sequence from ϕ21 lamboid virus (pK1Ep-Luciferase-4xϕ21BoxBr; NCBI accession number AH007390.1, nucleotides 866-883), v) TAR RNA sequence from Human immunodeficiency virus type 1, isolate HXB2 (pK1Ep-Luciferase-4xTAR; NCBI accession number K03455.1, nucleotides 471-497) and vi) U1snRNA RNA stem-loop (pK1Ep-Luciferase-4xU1snRNA; NCBI accession number M28013.1, nucleotides 123-155).


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


The design of the assay was very similar to Example 2, except that various protein:RNA tethering systems were tested in replacement to the Nλ:BoxBr system. In brief, uncapped Firefly Luciferase mRNA with a short polyadenylation tail of 40 adenosine residues was synthesized in cellulo by the phage K1E RNA polymerase and its expression in presence of NP868R tethered by various systems was assayed (FIG. 8).


Results of these experiments are shown in the table below:














Plasmids
mean
SEM















MS2-4xMS2sl tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
143 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
1 033 076
113 402


4xMS2sl




(3) pK1ERNAP, pMS2-NP868R, pK1Ep-
1 083 076
232 757


Luciferase




(4) pK1ERNAP, pMS2-NP868R, pK1Ep-
4 999 936
232 757


Luciferase-4xMS2sl




(5) Baseline
   162
   150







Nλ-4xλBoxBl tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
113 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
1 433 576
113 402


4xλBoxBl




(3) pK1ERNAP, pNλ-NP868R, pK1Ep-
1 430 576
232 757


Luciferase




(4) pK1ERNAP, pNλ-NP868R, pK1Ep-
5 699 936
332 757


Luciferase-4xλBoxBl




(5) Baseline
   162
   150







NP22-4xP22BoxBr tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
113 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
  913 576
113 402


4xP22BoxBr




(3) pK1ERNAP, pNP22-NP868R, pK1Ep-
  813 576
122 757


Luciferase




(4) pK1ERNAP, pNP22-NP868R, pK1Ep-
4 699 936
232 757


Luciferase-4xP22BoxBr




(5) Baseline
   162
   150







NΦ21-4xΦ21BoxBr tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
113 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
1 313 576
113 402


4xΦ21BoxBr




(3) pK1ERNAP, pNΦ21-NP868P, pK1Ep-
1 115 600
232 757


Luciferase




(4) pK1ERNAP, pNΦ21-NP868P, pK1Ep-
4 919 936
232 757


Luciferase-4xΦ21BoxBr




(5) Baseline
   162
   150







TAT-4xTAR tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
113 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
1 333 076
113 402


4xTAR




(3) pK1ERNAP, pTAT-NP868R, pK1Ep-
1 153 076
232 757


Luciferase




(4) pK1ERNAP, pTAT-NP868R, pK1Ep-
1 583 076
232 757


Luciferase-4xTAR




(5) Baseline
   162
   150







SNRNP70-4xU1snRNA tethering system









(1) pK1ERNAP, pNP868R, pK1Ep-Luciferase
1 233 076
113 402


(2) pK1ERNAP, pNP868R, pK1Ep-Luciferase-
1 331 222
 38 402


4xU1snRNA




(3) pK1ERNAP, pSNRNP70-NP868R, pK1Ep-
1 153 076
232 757


Luciferase




(4) pK1ERNAP, pSNRNP70-NP868R, pK1Ep-
1 423 076
157 757


Luciferase-4xU1snRNA




(5) Baseline
   162
   150









The cotransfection of pK1ERNAP with plasmids having only one component of the tethering system, i.e. the protein domains fused to the NP868R capping enzyme of the test plasmid or Firefly Luciferase reporter plasmids with four tandem RNA tethered repeats introduced in their 3′UTR, had no significant effects on the expression of the Firefly Luciferase reporter mRNA with any system when compared to no tethering system (row 2 or 3 vs. 1; p=NS for all comparisons, two-way Student t-test). The cotransfection of pK1ERNAP with plasmids encoding for the components of the MS2-4xMS2sl (i.e. pMS2-NP868R/pK1Ep-Luciferase-4xMS2sl), Nλ-NP868R-4xABoxBl (i.e. pNλ-NP868R/pK1Ep-Luciferase-4xλBoxBl), NP22-NP868R-4xP22BoxBr (i.e. pNP22-NP868R/pK1Ep-Luciferase-4xP22BoxBr), Nϕ21-4xϕ21BoxBr (i.e. pNϕ21-NP868R/pK1Ep-Luciferase-4xϕ21BoxBr), tethering system increased significantly by 3.8- to 4.6-fold the expression levels of firefly luciferase reporter in comparison to conditions with the untethering capping enzyme and/or untethered Firefly Luciferase plasmids (row 4 vs. 1-3; p<0.05 for all comparisons, two-way Student t-test). In contrast, the cotransfection of pK1ERNAP with either the TAT/4xTAR tethering system (i.e. pTAT-NP868R/pK1Ep-Luciferase-4xTAR) or the SNRNP70/4xU1snRNA tethering system (i.e. pSNRNP70-NP868R/pK1Ep-Luciferase-4xU1snRNA) shows very low change of Firefly Luciferase in comparison to conditions with the untethering capping enzyme and/or untethered Firefly Luciferase plasmids (row 4 vs. 1-3; p=NS for all comparisons, two-way Student t-test).


In conclusion, the best performances were obtained with the Nλ-4xλBoxBl tethering expression system (i.e. pNλ-NP868R/pK1Ep-Luciferase-4xλBoxBl), with performances of other tethering systems ranging in the following order (i.e. ratio of condition 4 vs.1): Nλ-4xλBoxBl>MS2-4xMS2sl>Nϕ21-4xϕ21BoxBr>NP22-4xP22BoxBr>>TAT-4xTAR>SNRNP70-4xU1snRNA.


4. Conclusions


The present experiments show that the African swine fever virus capping enzyme NP868R can increase the expression of Firefly Luciferase mRNA produced by the K1E phage RNA polymerase when appropriately tethered to the mRNA by bacteriophage protein-RNA tethering systems.


Example 4: Bacteriophage Protein:RNA Tethering Systems, Coupled to the D12L Subunit of the Vaccinia Virus Capping can Increase the Expression of Luciferase Reporter mRNA Produced by K1E Phage RNA Polymerase in Host-Cell Cytoplasm

1. Objectives


The objectives of the present experiments were to investigate if other protein:RNA tethering systems than the Nλ-4xBoxBr system can be used to guide the heterodimeric capping enzyme from the vaccinia virus to the a target mRNA, and thereby increase its expression.


The protein:RNA tethering systems tested hereinafter are the same as described in the previous example.


2. Methods


a. Plasmids


The pK1ERNAP expression plasmid was described in Example 1.


The test plasmid consisted of the coding sequence of the D12L subunit from the vaccinia virus capping enzyme fused at its amino-terminal end with the tethering protein sequences described above. The D1R plasmid was the same as previously described.


The Firefly Luciferase reporter plasmids containing the various tethered RNA sequences were the same as described in the previous example.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


The design of the assay was very similar to Example 1, except that various protein:RNA tethering systems were tested in replacement to the Nλ:BoxBr system (FIG. 9).


Results of these experiments are shown in the table below:














Plasmids
mean
SEM















MS2-4xMS2sl tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
573 812
41312


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-4xMS2sl
564 522
92952


(3) pK1ERNAP, pMS2-D12L, pD1R, pK1Ep-Luciferase
687 767
84793


(4) pK1ERNAP, pMS2-D12L, pD1R, pK1Ep-Luciferase-
1 147 529  
181568


4xMS2sl




(5) Baseline
   187
173







Nλ-4xλBoxBl tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
573 812
41312


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
566 750
60984


(3) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase
521 157
161568


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-
1 247 165  
141568


4xλBoxBl




(5) Baseline
   187
173







NP22-4xP22BoxBr tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
573 812
41312


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-
748 833
41312


4xP22BoxBr




(3) pK1ERNAP, pNP22-D12L, pD1R, pK1Ep-Luciferase
548 192
221568


(4) pK1ERNAP, pNP22-D12L, pD1R, pK1Ep-Luciferase-
1 306 542  
127189


4xP22BoxBr




(5) Baseline
   187
173







NΦ21-4xΦ21BoxBr tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
573 812
41312


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-
576 702
123936


4xΦ21BoxBr




(3) pK1ERNAP, pNΦ21-D12L, pD1R, pK1Ep-Luciferase
704 808
127189


(4) pK1ERNAP, pNΦ21-D12L, pD1R, pK1Ep-Luciferase-
1 432 734  
127189


4xΦ21BoxBr




(5) Baseline
   187
173







TAT-4xTAR tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
573 812
41312


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-4xTAR
489 422
81968


(3) pK1ERNAP, pTAT-D12L, pD1R, pK1Ep-Luciferase
420 064
190784


(4) pK1ERNAP, pTAT-D12L, pD1R, pK1Ep-Luciferase-
680 137
83594


4xTAR




(5) Baseline
   187
173







SNRNP70-4xU1snRNA tethering system









(1) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase
510 718
61968


(2) pK1ERNAP, pD12L, pD1R, pK1Ep-Luciferase-
582 331
6995


4xU1snRNA




(3) pK1ERNAP, pSNRNP70-D12L, pD1R, pK1Ep-Luciferase
630 096
190784


(4) pK1ERNAP, pSNRNP70-D12L, pD1R, pK1Ep-Luciferase-
618 425
129309


4xU1snRNA




(5) Baseline
   187
173









The cotransfection of pK1ERNAP with plasmids having only one out of the two components of the tethering system, i.e. the protein domains fused to the D12L subunit of the vaccinia virus capping of the test plasmid or the Firefly Luciferase reporter plasmids with four tandem RNA tethered repeats introduced in their 3′UTR, had no significant effects on the expression of the Firefly Luciferase reporter mRNA with any system when compared to no tethering system (row 2 or 3 vs. 1; p=NS for all comparisons, two-way Student t-test). Similarly to previous findings, the cotransfection of pK1ERNAP with plasmids with all the components of the MS2-4xMS2sl and D1R subunit of the vaccinia virus capping enzyme (i.e. pMS2-D12L/pD1R/pK1Ep-Luciferase-4xMS2sl), Nλ-D12L-4xABoxBl (i.e. pNλ-D12L/pD1R/pK1Ep-Luciferase-4xABoxBl), NP22-D12L-4xP22BoxBr (i.e. pNP22-D12L/pD1R/pK1Ep-Luciferase-4xP22BoxBr), Nϕ21-4xϕ21BoxBr (i.e. pNϕ21-D12L/pD1R/pK1Ep-Luciferase-4xϕ21BoxBr), tethering system increased significantly by 2- to 2.5-fold the expression levels of firefly luciferase reporter in comparison to conditions with the untethering capping enzyme and/or untethered Firefly Luciferase plasmids (row 4 vs. 1-3; p<0.05 for all comparisons, two-way Student t-test). In contrast, the cotransfection of pK1ERNAP with either the TAT/4xTAR tethering system (i.e. pTAT-D12L/pD1R/pK1Ep-Luciferase-4xTAR) or the SNRNP70/4xU1snRNA tethering system (i.e. pSNRNP70-D12L/pD1R/pK1Ep-Luciferase-4xU1snRNA) shows very low change of Firefly Luciferase in comparison to conditions with the untethering capping enzyme and/or untethered Firefly Luciferase plasmids (p=NS for all comparisons, two-way Student t-test).


Finally, the performances of the tethering systems ranged in a different order than in the previous example (i.e. ratio of condition 4 vs.1): Nϕ21-4xϕ21BoxBr>NP22-4xP22BoxBr>Nλ-4xλBoxBl>MS2-4xMS2sl>>TAT-4xTAR>SNRNP70-4xU1snRNA.


4. Conclusions


The present experiments show that the heterodimeric D1R/D12L capping enzyme from the vaccinia virus can also increase the expression of Firefly Luciferase mRNA produced by the K1E phage RNA polymerase when appropriately tethered to the mRNA by bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system.


Example 5: Fusion Between Poly(A) Polymerases and African Swine Fever Virus NP868R Capping Enzyme Increase the Expression of Luciferase Reporter mRNA Produced by K1E Phage RNA Polymerase when Appropriately Tethered to the Target Transcript

1. Objectives


The present experiments aimed to determine if poly(A) polymerases fused to NP868R African Swine Fever virus capping enzyme can increase the expression of Firefly Luciferase reporter mRNA produced by the K1E phage RNA polymerase when appropriately tethered to the target transcript by the Nλ-BoxBl the thering system.


2. Methods


a. Plasmids


The ORFs of the following poly(A) polymerases were synthesized: i) PAP1 poly(A) polymerase from Saccharomyces cerevisiae sorted to the cytoplasm by deletion of the 42 carboxyl-terminal amino-acids that contains a nuclear localization signal (Zhelkovsky, Helmling et al. 1998) (NCBI accession number: P29468); the vaccinia virus VP55 poly(A) polymerase (UniProtKB/Swiss-Prot accession number P23371 corresponding to the nucleotide and amino-acid sequences, respectively), the viral R341 poly(A) polymerase from Acanthamoeba polyphaga mimivirus (UniProtKB/Swiss-Prot accession number: E3VZZ8), iv) the viral MG561 poly(A) polymerase from Megavirus chilensis (NCBI Accession number: YP_004894612), v) the viral C475L poly(A) polymerase from the African swine fever virus (UniProtKB/Swiss-Prot accession number: A0A0A1E081), vi) mutant PAPOLA (K656R-K657R mutation of the human PAPOLA, UniProtKB/Swiss-Prot accession number P51003) mutated at its the nuclear localization signal (Raabe, Murthy et al. 1994, Vethantham, Rao et al. 2008), vii) the wild-type canonical Mus musculus testis specific PAPOLB (UniProtKB/Swiss-Prot Q9WVP6).


Four types of test plasmids were generated by in-frame subcloning of the poly(A) polymerases ORFs: i) in the pCMV-Script backbone only (e.g. pPAP1), ii) downstream to the NA tethering domain (e.g. pNλ-PAP1), iii) downstream to the Nλ-NP868R protein through a G4 flexible linker (e.g. pNλ-NP868R-G4-PAP1) resulting in the expression of monomeric protein, or iv) between the N protein tethering domain from the lambda bacteriophage and NP868R through a G4 flexible linker (e.g. pNλ-PAP1-G4-NP868R) also resulting in the expression of monomeric protein. The design of these two latter constructions is shown FIG. 10.


The Firefly Luciferase reporter plasmids in their untethered (pK1Ep-Luciferase) or tethered version (pK1Ep-Luciferase-4xλBoxBl) were the same as described above.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


The design of the assay was very similar to Example 2, except that the poly(A) polymerases were fused to African Swine Fever virus NP868R capping enzyme (FIG. 11).


Results of these experiments are shown in the table below:














Plasmids
mean
SEM















C475L poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, C475L, pK1Ep-Luciferase-4xλBoxBl
  240 907
386 848


(3) pK1ERNAP, pNλ-C475L, pK1Ep-Luciferase-4xλBoxBl
  301 134
499 738


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pC475L, pNλ-NP868R, pK1Ep-Luciferase-
4 246 290
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-C475L, pNλ-NP868R, pK1Ep-
6 794 064
364 911


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-C475L-G4-NP868R, pK1Ep-Luciferase-
8 685 660
414 672


4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-C475L, pK1Ep-Luciferase-
8 385 560
393 938


4xλBoxBl




(9) Baseline
  22 217
 14 176







MG561 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, MG561, pK1Ep-Luciferase-4xλBoxBl
  316 191
360 922


(3) pK1ERNAP, pNλ-MG561, pK1Ep-Luciferase-4xλBoxBl
  421 588
832 896


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pMG561, pNλ-NP868R, pK1Ep-Luciferase-
4 246 290
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-MG561, pNλ-NP868R, pK1Ep-
6 199 583
364 911


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-MG561-G4-NP868R, pK1Ep-Luciferase-
8 638 575
734 672


4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-MG561, pK1Ep-Luciferase-
9 070 504
697 938


4xλBoxBl




(9) Baseline
  22 217
 14 176







PAP1 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, PAP1, pK1Ep-Luciferase-4xλBoxBl
  225 851
277 632


(3) pK1ERNAP, pNλ-PAP1, pK1Ep-Luciferase-4xλBoxBl
  361 361
694 080


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pPAP1, pNλ-NP868R, pK1Ep-Luciferase-
4 352 447
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-PAP1, pNλ-NP868R, pK1Ep-Luciferase-
6 659 244
364 911


4xλBoxBl




(7) pK1ERNAP, pNλ-PAP1-G4-NP868R, pK1Ep-Luciferase-
10 638 575 
734 688


4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-PAP1, pK1Ep-Luciferase-
12 538 575 
404 078


4xλBoxBl




(9) Baseline
  22 217
 14 176







R341 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, R341, pK1Ep-Luciferase-4xλBoxBl
  271 021
249 869


(3) pK1ERNAP, pNλ-R341, pK1Ep-Luciferase-4xλBoxBl
  338 776
430 330


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pR341, pNλ-NP868R, pK1Ep-Luciferase-
4 069 361
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-R341, pNλ-NP868R, pK1Ep-Luciferase-
6 673 752
364 911


4xλBoxBl




(7) pK1ERNAP, pNλ-R341-G4-NP868R, pK1Ep-Luciferase-
8 992 399
734 688


4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-R341, pK1Ep-Luciferase-
8 522 353
881 625


4xλBoxBl




(9) Baseline
  22 217
 14 176







VP55 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, VP55, pK1Ep-Luciferase-4xλBoxBl
  271 021
388 685


(3) pK1ERNAP, pNλ-VP55, pK1Ep-Luciferase-4xλBoxBl
  331 247
291 514


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pVP55, pNλ-NP868R, pK1Ep-Luciferase-
4 493 990
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-VP55, pNλ-NP868R, pK1Ep-Luciferase-
6 673 576
364 911


4xλBoxBl




(7) pK1ERNAP, pNλ-VP55-G4-NP868R, pK1Ep-Luciferase-
8 638 575
134 687


4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-VP55, pK1Ep-Luciferase-
10 107 133 
220 406


4xλBoxBl




(9) Baseline
  22 217
 14 176







PAPOLA poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  124 567
138 816


(2) pK1ERNAP, PAPOLA, pK1Ep-Luciferase-4xλBoxBl
  267 819
388 685


(3) pK1ERNAP, pNλ-PAPOLA, pK1Ep-Luciferase-4xλBoxBl
  274 047
291 514


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pPAPOLA, pNλ-NP868R, pK1Ep-Luciferase-
4 635 533
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-PAPOLA, pNλ-NP868R, pK1Ep-
7 231 432
364 911


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-PAPOLA-G4-NP868R, pK1Ep-
12 203 575 
734 688







Luciferase-4xλBoxBl









(8) pK1ERNAP, pNλ-NP868R-G4-PAPOLA, pK1Ep-
11 262 433 
220 406


Luciferase-4xλBoxBl




(9) Baseline
  22 217
 14 176







PAPOLB polu(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  150 567
138 816


(2) pK1ERNAP, PAPOLB, pK1Ep-Luciferase-4xλBoxBl
  251 447
360 922


(3) pK1ERNAP, pNλ-PAPOLB, pK1Ep-Luciferase-4xλBoxBl
  337 270
832 896


(4) pK1ERNAP, pNλ-NP868R, pK1Ep-Luciferase-4xλBoxBl
3 538 575
414 672


(5) pK1ERNAP, pPAPOLB, pNλ-NP868R, pK1Ep-Luciferase-
5 874 035
456 139


4xλBoxBl




(6) pK1ERNAP, pNλ-PAPOLB, pNλ-NP868R, pK1Ep-
7 753 726
364 911


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-PAPOLB-G4-NP868R, pK1Ep-
9 338 575
734 688


Luciferase-4xλBoxBl




(8) pK1ERNAP, pNλ-NP868R-G4-PAPOLB, pK1Ep-
9 808 575
624 484


Luciferase-4xλBoxBl




(9) Baseline
  22 217
 14 176









In the absence of mRNA capping provided by pNλ-NP868R, non-statistically significant increase of Firefly Luciferase mRNA expression of ˜1.5-fold and 2.5-fold was observed when untethered (row 2 vs. 1) or tethered poly(A) polymerase plasmids (row 3 vs. 1) were transfected, respectively (p=NS, two-way Student t-test). When the Firefly Luciferase mRNA was capped by co-transfection of pNλ-NP868R, a statistically significant increase of expression of ˜1.5-fold (row 5 vs. 4) and ˜2-fold (row 6 vs. 4) was observed when the untethered or tethered poly(A) polymerases plasmids were cotransfected, respectively (p<0.05 for all untethered poly(A) polymerases vs. no poly(A) polymerases, two-way Student t-test).


Poly(A) polymerases were fused to NP868R African Swine Fever virus capping enzyme, together with the Nλ-protein tethering domain. Two types of fusion were tested with poly(A) polymerases subcloned either downstream to Nλ-NP868R through a G4 flexible linker or between the N protein tethering domain from the lambda bacteriophage and NP868R through a Ga flexible linker. All tethered fusions genes of both types increased the expression of Firefly Luciferase mRNA in comparison to non-linked enzymes (row 7 and 8 vs. 6; p<0.05 for all comparisons, two-way Student t-test). Activity of the fusion proteins ranged as follows: Nλ-NP868R-G4-C475L<Nλ-NP868R-G4-R341<Nλ-MG561-G4-NP868R<Nλ-VP55-G4-NP868R<Nλ-C475L-G4-NP868R<Nλ-R341-G4-NP868R<Nλ-NP868R-G4-MG561<Nλ-PAPOLB-G4-NP868R<Nλ-NP868R-G4-PAPOLB<Nλ-NP868R-G4-VP55<Nλ-PAP1-G4-NP868R<Nλ-NP868R-G4-PAPOLA<Nλ-PAPOLA-G4-NP868R<Nλ-NP868R-G4-PAP1


4. Conclusions


The present experiments show that various poly(A) polymerases including mammalian, yeast, viral and bacterial enzymes fused to the African Swine Fever virus NP868R capping enzyme increase the expression of transcripts produced by phage RNA polymerase, when appropriately tethered with the Nλ-4xBoxBl system. Surprisingly, the fusion between various poly(A) polymerases and NP868R capping enzymes, which are not physically linked in the nature and contain no RNA-binding domain, can act synergistically and this effect is even greater when these fusion proteins are appropriately tethered (rows 7 and 8 in the above Table). These results are really surprising and one skilled in the art could have expected to obtain the same expression rate since the components are the same.


Example 6: Fusion of Poly(A) Polymerases and the D12 Subunit of the Heterodimeric Vaccinia Virus Capping Enzyme can Increase the Expression of Luciferase Reporter mRNA Produced by K1E Phage RNA Polymerase when Appropriately Tethered to the Target Transcript

1. Objectives


The present experiments aim to determine if fusions of poly(A) polymerases with D12 subunit of the heterodimeric vaccinia virus capping enzyme can increase the expression of transcripts produced by phage RNA polymerase, when appropriately tethered with the Nλ-4xBoxBl system.


2. Methods


a. Plasmids


The pK1ERNAP expression plasmid was described in Example 1.


The poly(A) polymerases tested were described in previous example and subcloned in-frame (FIG. 12): i) downstream to the Nλ-D12L protein through a G4 flexible linker (e.g. pNλ-D12L-G4-PAP1), or between the N protein tethering domain from the lambda bacteriophage and D12L through a G4 flexible linker (e.g. pNλ-PAP1-G4-D12L).


The Firefly Luciferase reporter plasmids in their untethered (pK1Ep-Luciferase) or tethered version (pK1Ep-Luciferase-4xλBoxBl) were the same as described above.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


The design of the experiment was similar to previous example, except that the capping enzyme consisted of the vaccinia virus heterodimer D1R/D12 (FIG. 13).


Results of these experiments are shown in the table below:














Plasmids
mean
SEM















C475L poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  145 279
116 201


(2) pK1ERNAP, C475L, pK1Ep-Luciferase-4xλBoxBl
  201 661
325 912


(3) pK1ERNAP, pNλ-C475L, pK1Ep-Luciferase-4xλBoxBl
  253 700
466 225


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
3 301 279
429 341


(5) pK1ERNAP, pC475L, pNλ-D12L, pD1R, pK1Ep-Luciferase-
4 196 498
353 954


4xλBoxBl




(6) pK1ERNAP, pNλ-C475L, pNλ-D12L, pD1R, pK1Ep-
5 272 046
95 796


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-C475L-G4-D12L, pD1R, pK1Ep-Luciferase-
7 201 395
453 620


4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-C475L, pK1Ep-Luciferase-
7 173 174
267 557


4xλBoxBl




(9) Baseline
  23 887
 9 510







MG561 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  121 133
125 248


(2) pK1ERNAP, MG561, pK1Ep-Luciferase-4xλBoxBl
  285 286
334 854


(3) pK1ERNAP, pNλ-MG561, pK1Ep-Luciferase-4xλBoxBl
  391 138
866 711


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
2 240 943
218 840


(5) pK1ERNAP, pMG561, pNλ-D12L, pD1R, pK1Ep-Luciferase-
3 682 240
466 962


4xλBoxBl




(6) pK1ERNAP, pNλ-MG561, pNλ-D12L, pD1R, pK1Ep-
5 346 676
374 202


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-MG561-G4-D12L, pD1R, pK1Ep-
8 858 505
603 377


Luciferase-4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-MG561, pK1Ep-
7 449 488
527 949


Luciferase-4xλBoxBl




(9) Baseline
  23 887
 9 510







PAP1 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  120 765
132 669


(2) pK1ERNAP, PAP1, pK1Ep-Luciferase-4xλBoxBl
  215 849
176 186


(3) pK1ERNAP, pNλ-PAP1, pK1Ep-Luciferase-4xλBoxBl
  229 321
646 674


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
3 296 890
441 173


(5) pK1ERNAP, pPAP1, pNλ-D12L, pD1R, pK1Ep-Luciferase-
4 630 608
427 360


4xλBoxBl




(6) pK1ERNAP, pNλ-PAP1, pNλ-D12L, pD1R, pK1Ep-Luciferase-
6 239 087
194 450


4xλBoxBl




(7) pK1ERNAP, pNλ-PAP1-G4-D12L, pD1R, pK1Ep-Luciferase-
10 668 967 
620 415


4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-PAP1, pK1Ep-Luciferase-
9 534 665
223 391


4xλBoxBl




(9) Baseline
  23 887
 9 510







R341 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  92 989
144 738


(2) pK1ERNAP, R341, pK1Ep-Luciferase-4xλBoxBl
  282 582
190 160


(3) pK1ERNAP, pNλ-R341, pK1Ep-Luciferase-4xλBoxBl
  257 822
449 340


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
2 684 956
273 600


(5) pK1ERNAP, pR341, pNλ-D12L, pD1R, pK1Ep-Luciferase-
3 694 895
372 489


4xλBoxBl




(6) pK1ERNAP, pNλ-R341, pNλ-D12L, pD1R, pK1Ep-Luciferase-
5 449 865
413 128


4xλBoxBl




(7) pK1ERNAP, pNλ-R341-G4-D12L, pD1R, pK1Ep-Luciferase-
10 180 543 
694 655


4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-R341, pK1Ep-Luciferase-
8 057 982
766 195


4xλBoxBl




(9) Baseline
  23 887
 9 510







VP55 poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  111 045
 96 804


(2) pK1ERNAP, VP55, pK1Ep-Luciferase-4xλBoxBl
  188 997
328 927


(3) pK1ERNAP, pNλ-VP55, pK1Ep-Luciferase-4xλBoxBl
  280 320
285 568


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
3 466 398
436 289


(5) pK1ERNAP, pVP55, pNλ-D12L, pD1R, pK1Ep-Luciferase-
4 728 261
461 933


4xλBoxBl




(6) pK1ERNAP, pNλ-VP55, pNλ-D12L, pD1R, pK1Ep-Luciferase-
5 758 344
375 637


4xλBoxBl




(7) pK1ERNAP, pNλ-VP55-G4-D12L, pD1R, pK1Ep-Luciferase-
7 863 085
 29 635


4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-VP55, pK1Ep-Luciferase-
8 601 926
252 202


4xλBoxBl




(9) Baseline
  23 887
 9 510







PAPOLA poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  174 916
 96 128


(2) pK1ERNAP, PAPOLA, pK1Ep-Luciferase-4xλBoxBl
  185 461
347 658


(3) pK1ERNAP, pNλ-PAPOLA, pK1Ep-Luciferase-4xλBoxBl
  245 121
282 829


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
3 433 158
309 045


(5) pK1ERNAP, pPAPOLA, pNλ-D12L, pD1R, pK1Ep-Luciferase-
4 454 749
385 659


4xλBoxBl




(6) pK1ERNAP, pNλ-PAPOLA, pNλ-D12L, pD1R, pK1Ep-
6 114 076
325 113


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-PAPOLA-G4-D12L, pD1R, pK1Ep-
9 230 982
731 066


Luciferase-4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-PAPOLA, pK1Ep-
10 284 960 
221 627


Luciferase-4xλBoxBl




(9) Baseline
  23 887
 9 510







PAPOLB poly(A) polymerase series









(1) pK1ERNAP, pK1Ep-Luciferase-4xλBoxBl
  134 279
130 482


(2) pK1ERNAP, PAPOLB, pK1Ep-Luciferase-4xλBoxBl
  236 351
321 719


(3) pK1ERNAP, pNλ-PAPOLB, pK1Ep-Luciferase-4xλBoxBl
  300 637
861 464


(4) pK1ERNAP, pNλ-D12L, pD1R, pK1Ep-Luciferase-4xλBoxBl
3 659 946
363 888


(5) pK1ERNAP, pPAPOLB, pNλ-D12L, pD1R, pK1Ep-Luciferase-
4 154 652
410 206


4xλBoxBl




(6) pK1ERNAP, pNλ-PAPOLB, pNλ-D12L, pD1R, pK1Ep-
5 972 923
350 054


Luciferase-4xλBoxBl




(7) pK1ERNAP, pNλ-PAPOLB-G4-D12L, pD1R, pK1Ep-
7 677 493
552 983


Luciferase-4xλBoxBl




(8) pK1ERNAP, pNλ-D1R, pD12L-G4-PAPOLB, pK1Ep-
8 958 357
480 636


Luciferase-4xλBoxBl




(9) Baseline
  23 887
 9 510









In the absence of mRNA capping provided by pNλ-D12L/D1R, non-statistically significant change of Firefly Luciferase mRNA expression of ˜1.5-fold and 2-fold was observed when untethered (row 2 vs. 1) or tethered poly(A) polymerase plasmids (row 3 vs. 1) were transfected, respectively (p=NS, two-way Student t-test). Similarly to previous findings, when the Firefly Luciferase mRNA was capped by co-transfection of pNλ-D12L/D1R, a statistically significant increase of expression of ˜1.5-fold (row 5 vs. 4) and ˜2-fold (row 6 vs. 4) was observed when the untethered or tethered poly(A) polymerases plasmids were cotransfected, respectively (p<0.05 for all untethered poly(A) polymerases vs. no poly(A) polymerases, two-way Student t-test).


Poly(A) polymerases were fused to the D12 subunit of the heterodimeric vaccinia virus capping enzyme, together with the Nλ-protein domain as described above. All tethered fusions genes of both types increased the expression of Firefly Luciferase mRNA in comparison to non-linked enzymes (row 7 and 8 vs. 6; p<0.05 for all comparisons, two-way Student t-test). Activity of the fusion complexes ranged as follows: Nλ-D12L/D1R-G4-C475L<Nλ-C475L-G4-D12L/D1R<Nλ-D12L/D1R-G4-MG561<Nλ-PAPOLB-G4-D12L/D1R<Nλ-VP55-G4-D12L/D1R<Nλ-D12L/D1R-G4-R341<Nλ-D12L/D1R-G4-VP55<Nλ-MG561-G4-D12L/D1R<Nλ-D12L/D1R-G4-PAPOLB<Nλ-PAPOLA-G4-D12L/D1R<Nλ-D12L/D1R-G4-PAP1<Nλ-R341-G4-D12L/D1R<Nλ-D12L/D1R-G4-PAPOLA<Nλ-PAP1-G4-D12L/D1R.


4. Conclusions


The present experiments that various poly(A) polymerases fused to the D12 subunit of the heterodimeric vaccinia virus capping enzyme together with D1R subunit, which are not physically linked in the nature and contain no RNA-binding domain, can act synergistically and this effect is even greater when these fusion proteins appropriately tethered.


Example 7: Non-Covalent Tethered Coupling Between Acanthamoeba polyphaga Mimivirus R341 Poly(A) Polymerase and African Swine Fever Virus NP868R Capping Enzyme, can Increase the Expression of Luciferase Reporter mRNA Produced by Phage RNA Polymerases

1. Objectives


The objectives of this set of experiments were to determine if non-covalent coupling between the R341 poly(A) polymerase and NP868R capping enzyme also results in an active expression system able to enhance the expression of uncapped Firefly Luciferase reporter mRNA when appropriately tethered. In the present experiments, the non-covalent coupling was generated using complementary leucine zippers that form heterodimers.


2. Methods


a. Plasmids


The pK1ERNAP expression and Firefly Luciferase reporter plasmids in their untethered (pK1Ep-Luciferase) or tethered version (pK1Ep-Luciferase-4xλBoxBr) were the same as described above.


Non-covalent coupling between the Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase and the African swine fever virus NP868R capping enzyme was induced by the EE1234L and RR1234L complementary leucine-zippers (SEQ ID No 28 and SEQ ID No 29 corresponding to the nucleotide and amino-acid sequences of G4-EE1234L leucine-zipper, respectively; SEQ ID No 30 and SEQ ID No 31 corresponding to the nucleotide and amino-acid sequences of RR1234L-G4 leucine-zipper, respectively). These amphipathic α-helices form an antiparallel heterodimer with dissociation affinity of ˜10−15M (Moll, Ruvinov et al. 2001). Two non-covalent heterodimeric complexes were generated between NP868R capping enzyme and R341 RNA polymerase (FIG. 14). Firstly, the EE1234L leucine zipper was fused in-frame to the carboxyl-terminal end of Nλ-R41, while RR1234L was fused in-frame to the amino-terminal end of NP868R. Co-expression of these two plasmids, respectively named pNλ-R341-EE1234L and RR1234L-NP868R, therefore produces a heterodimer with an Nλ-tethering domain carried by R341. Secondly, RR1234L leucine zipper was fused in-frame to the carboxyl-terminal end of Nλ-NP868R, while the EE1234L was fused in-frame to the amino-terminal end of R341. Co-expression of these two plasmids, respectively named pNλ-NP868R-RR1234L and pEE1234L-R341, therefore generates a heterodimer with an Nλ-tethering domain carried by NP868R. These plasmids were synthesized by subcloning the corresponding ORFs in the pCMVScript plasmid backbone at endonuclease restriction enzyme sites.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


Uncapped Firefly Luciferase reporter mRNA was generated by the K1E bacteriophage RNA polymerase, using the pK1Ep-Luciferase and pK1Ep-Luciferase-4xλBoxBr plasmids as previously described (FIG. 15).


Results of these experiments are shown in the table below:














Plasmids
mean
SEM







(1) pK1ERNAP, pNλ-R341
  497 650
182 520


(2) pK1ERNAP, pEE1234L-R341
  257 650
109 512


(3) pK1ERNAP, pNλ-R341-EE1234L
  522 533
146 016


(4) pK1ERNAP, pNλ-NP868R
2 377 957
237 063


(5) pK1ERNAP, pRR1234L-NP868R
1 049 099
104 587


(6) pK1ERNAP, pNλ-NP868R-RR1234L
2 331 330
232 415


(7) pK1ERNAP, pEE1234L-R341, pRR1234L-
4 539 654
325 828


NP868R




(8) pK1ERNAP, pNλ-R341-EE1234L,
12 924 395 
325 828


pRR1234L-NP868R




(9) pK1ERNAP, pNλ-NP868R-RR1234L,
10 191 523 
270 437


pEE1234L-R341




(10) Baseline
   671
   257









The Nλ-tethering of the Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase with or without leucine zipper increased modestly the expression of uncapped 4xλBoxBr-Firefly Luciferase mRNA generated by the phage K1E RNA polymerase in comparison to untethered Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase (row 1 or 3 vs. 2). The Nλ-tethering of the African Swine Fever Virus NP868R capping enzyme with or without leucine zipper increased frankly the expression of uncapped Firefly Luciferase 4xλBoxBr-mRNA generated by the phage K1E RNA polymerase in comparison to untethered NP868R capping enzyme or no capping enzyme (row 4 or 6 vs. 5).


The non-covalent coupling between the Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase and the African swine fever virus NP868R capping enzyme was generated using the EE1234L and RR1234L complementary high-affinity leucine-zippers. This heterodimeric complex without the Nλ-tethering domain resulted in active complex that significantly increased the expression of uncapped Firefly Luciferase mRNA generated by the phage K1E RNA polymerase in comparison to conditions to either untethered R341 or NP868R with leucine zippers alone (row 7 vs. 2 or 5). Noticeably, the addition of Nλ-tethering at amino-terminal end of the Acanthamoeba polyphaga mimivirus R341 poly(A) polymerase or the African swine fever virus NP868R capping enzyme of this heterodimeric complex increased by 2.80- and 2.24-fold the expression of uncapped Firefly Luciferase mRNA in comparison to the untethered complex (row 8 or 9 vs. 7; p<0.05 for both comparisons, two-way Student t-test).


4. Conclusions


The present experiments show that the artificial coupling between the R341 poly(A) polymerase and the African swine fever virus NP868R capping enzyme, i.e. non-covalent through leucine zippers, also results in synergistically active heterodimers and this effect is even greater when these fusion proteins appropriately tethered.


Example 8: Assemblies Between the Acanthamoeba polyphaga mimivirus R341 Poly(A) Polymerase, African Swine Fever Virus NP868R Capping Enzyme and Phage K1E RNA Polymerase Results in Active Expression Complexes

1. Objectives


The objective of the following experiment was to determine if active complexes could be generated by assembling the Acanthamoeba polyphaga mimivirus poly(A) polymerase R341, the African swine fever virus NP868R capping enzyme and the phage K1E RNA polymerase when appropriately Nλ-tethered.


The assemblies tested hereinafter are designed according to the common Nλ-R341-[X1]-NP868R-[X2]-K1ERNAP protein scaffold, where [X1] and [X2] are variable.


The following open-reading-frames were generated to test this hypothesis (FIG. 16):

    • [X1]=G4, [X2]=G4; i.e. Nλ-R341-G4-NP868R-G4-K1ERNAP construction (NCBI accession number J02459-SEQ ID No 40 and SEQ ID No 41 corresponding to the nucleotide and amino-acid sequences of Nλ-R341-G4-NP868R-G4-K1ERNAP, respectively) featured by the fusion of the three enzymatic subunit through flexible linkers with the Nλ-tethering peptide at its N-terminus,
    • [X1]=G4, [X2]=F2A; i.e. Nλ-R341-G4-NP868R-F2A-K1ERNAP construction (SEQ ID No 42 and SEQ ID No 43 corresponding to the nucleotide and amino-acid sequences of Nλ-R341-G4-NP868R-F2A-K1ERNAP, respectively) featured by the substitution of the G4 flexible linker located between the African swine fever virus NP868R capping enzyme and the phage K1E RNA polymerase by the F2A ribosomal skipping sequence from the picornavirus aphtovirus (Foot-and-mouth disease aphovirus type 0 polyprotein, UniProtKB/Swiss-Prot accession number AAT01756, residues 934-955). Ribosomal skipping results in apparent co-translational cleavage of the protein (Donnelly, Luke et al. 2001),
    • [X1]=F2A, [X2]=G4; i.e. Nλ-R341-F2A-NP868R-G4-K1ERNAP construction (SEQ ID No 44 and SEQ ID No 45 corresponding to the nucleotide and amino-acid sequences of Nλ-R341-F2A-NP868R-G4-K1ERNAP, respectively) featured by the substitution of the G4 flexible linker located between the R341 poly(A) polymerase and the African swine fever virus NP868R capping enzyme by the F2A ribosomal skipping sequence.
    • [X1]=T2A, [X2]=G4; i.e. Nλ-R341-T2A-NP868R-G4-K1ERNAP construction (SEQ ID No 46 and SEQ ID No 47 corresponding to the nucleotide and amino-acid sequences of Nλ-R341-T2A-NP868R-G4-K1ERNAP, respectively) featured by the substitution of the G4 flexible linker located between the R341 poly(A) polymerase and the African swine fever virus NP868R capping enzyme by the T2A ribosomal skipping sequence from the porcine teschovirus-1 (UniProtKB/Swiss-Prot accession number Q9WJ28, residues 979-997).


2. Methods


a. Plasmids


The Firefly Luciferase reporter plasmids in their untethered (pK1Ep-Luciferase) or tethered version (pK1Ep-Luciferase-4xλBoxBl) were the same as described above.


The ORFs previously described were subcloned in the pCMVScript backbone, therefore resulting in the pNλ-R341-G4-NP868R-G4-K1ERNAP, pNλ-R341-G4-NP868R-F2A-K1ERNAP, pNλ-R341-F2A-NP868R-G4-K1ERNAP, and pNλ-R341-T2A-NP868R-G4-K1ERNAP plasmids.


b. Cell Culture and Transfection


Same as described in Example 1.


c. Firefly Luciferase Luminescence and SEAP Colorimetric Assays


Same as described in Example 1.


d. Statistical Analysis


Same as described in Example 1.


3. Results


A depiction of the assay is shown FIG. 17.


Results of these experiments are shown in the table below:














Plasmids
mean
SEM







(1) pNλ-R341-G4-NP868R, pK1ERNAP,
2 989 500
321 841


pK1Ep-Luciferase-4xλBoxBl




(2) pNλ-R341-G4-NP868R-G4-K1ERNAP,
3 882 614
649 466


pK1Ep-Luciferase-4xλBoxBl




(3) pNλ-R341-G4-NP868R-F2A-K1ERNAP,
4 393 507
928 702


pK1Ep-Luciferase-4xλBoxBl




(4) pNλ-R341-F2A-NP868R-G4-K1ERNAP,
7 778 512
219 138


pK1Ep-Luciferase-4xλBoxBl




(5) pNλ-R341-T2A-NP868R-G4-K1ERNAP,
7 861 869
1 120 056  


pK1Ep-Luciferase-4xλBoxBl




(6) Baseline
  162 088
 56 504









The various fusion of K1ERNAP coding sequence with Nλ-R341-F2A-NP868R were compared with non-linked Nλ-R341-F2A-NP868R and K1ERNAP. All fusions constructions gave significantly greater expression levels than non-linked Nλ-R341-F2A-NP868R and K1ERNAP (row 2-to-5 vs. 1; p<0.05, two-way Student t-test for all comparisons). Best results were obtained with Nλ-R341-T2A-NP868R-G4-K1ERNAP and Nλ-R341-F2A-NP868R-G4-K1ERNAP fusions, with other conditions ranging in the following order: Nλ-R341-T2A-NP868R-G4-K1ERNAP Nλ-R341-F2A-NP868R-G4-K1ERNAP>>Nλ-R341-G4-NP868R-F2A-K1ERNAP>Nλ-R341-G4-NP868R-G4-K1ERNAP>pNλ-R341-G4-NP868R, pK1ERNAP.


4. Conclusions


The present experiments show that active tethered expression systems can be generated by assembling the poly(A) polymerase R341, African swine fever virus NP868R capping enzyme and phage K1E RNA polymerase under a Nλ-R341-[X1]-NP868R-[X2]-K1ERNAP scaffold, preferably where [X1]=T2A or F2A, and [X2]=G4. Unexpectedly, the construction Nλ-R341-F2A-NP868R-G4-K1ERNAP and Nλ-R341-T2A-NP868R-G4-K1ERNAP allow higher expression rate than the association of the constructions Nλ-R341 with NP868R-G4-K1ERNAP (row 4 and 5 vs. 1). These results are really surprising and one skilled in the art could have expected to obtain the same expression rate since the components are the same and are not physically linked in the nature and nor contain any RNA-binding domain.


BIBLIOGRAPHY



  • Annamalai, P., S. Apte, S. Wilkens and A. L. Rao (2005). “Deletion of highly conserved arginine-rich RNA binding motif in cowpea chlorotic mottle virus capsid protein results in virion structural alterations and RNA packaging constraints.” J Virol 79(6): 3277-3288.

  • Ballaun, C., G. K. Farrington, M. Dobrovnik, J. Rusche, J. Hauber and E. Bohnlein (1991). “Functional analysis of human T-cell leukemia virus type I rex-response element: direct RNA binding of Rex protein correlates with in vivo activity.” J Virol 65(8): 4408-4413.

  • Banerjee, H., A. Rahn, B. Gawande, S. Guth, J. Valcarcel and R. Singh (2004). “The conserved RNA recognition motif 3 of U2 snRNA auxiliary factor (U2AF 65) is essential in vivo but dispensable for activity in vitro.” RNA 10(2): 240-253.

  • Battiste, J. L., H. Mao, N. S. Rao, R. Tan, D. R. Muhandiram, L. E. Kay, A. D. Frankel and J. R. Williamson (1996). “Alpha helix-RNA major groove recognition in an HIV-1 rev peptide-RRE RNA complex.” Science 273(5281): 1547-1551.

  • Bedzyk, W. D., K. M. Weidner, L. K. Denzin, L. S. Johnson, K. D. Hardman, M. W. Pantoliano, E. D. Asel and E. W. Voss, Jr. (1990). “Immunological and structural characterization of a high affinity anti-fluorescein single-chain antibody.” J Biol Chem 265(30): 18615-18620.

  • Belanger, F., J. Stepinski, E. Darzynkiewicz and J. Pelletier (2010). “Characterization of hMTr1, a human Cap1 2′-O-ribose methyltransferase.” J Biol Chem 285(43): 33037-33044.

  • Benarroch, D., M. Jankowska-Anyszka, J. Stepinski, E. Darzynkiewicz and S. Shuman (2010). “Cap analog substrates reveal three clades of cap guanine-N2 methyltransferases with distinct methyl acceptor specificities.” RNA 16(1): 211-220.

  • Benarroch, D., Z. R. Qiu, B. Schwer and S. Shuman (2009). “Characterization of a mimivirus RNA cap guanine-N2 methyltransferase.” RNA 15(4): 666-674.

  • Benarroch, D., P. Smith and S. Shuman (2008). “Characterization of a trifunctional mimivirus mRNA capping enzyme and crystal structure of the RNA triphosphatase domain.” Structure 16(4): 501-512.

  • Bird, R. E., K. D. Hardman, J. W. Jacobson, S. Johnson, B. M. Kaufman, S. M. Lee, T. Lee, S. H. Pope, G. S. Riordan and M. Whitlow (1988). “Single-chain antigen-binding proteins.” Science 242(4877): 423-426.

  • Brisson, M., Y. He, S. Li, J. P. Yang and L. Huang (1999). “A novel T7 RNA polymerase autogene for efficient cytoplasmic expression of target genes.” Gene Ther 6(2): 263-270.

  • Bujnicki, J. M. and L. Rychlewski (2001). “Reassignment of specificities of two cap methyltransferase domains in the reovirus lambda 2 protein.” Genome Biol 2(9): RESEARCH0038.

  • Busch, R., A. Pashine, K. C. Garcia and E. D. Mellins (2002). “Stabilization of soluble, low-affinity HLA-DM/HLA-DR1 complexes by leucine zippers.” J Immunol Methods 263(1-2): 111-121.

  • Busch, R., Z. Reich, D. M. Zaller, V. Sloan and E. D. Mellins (1998). “Secondary structure composition and pH-dependent conformational changes of soluble recombinant HLA-DM.” J Biol Chem 273(42): 27557-27564.

  • Cai, A., M. Jankowska-Anyszka, A. Centers, L. Chlebicka, J. Stepinski, R. Stolarski, E. Darzynkiewicz and R. E. Rhoads (1999). “Quantitative assessment of mRNA cap analogues as inhibitors of in vitro translation.” Biochemistry 38(26): 8538-8547.

  • Carey, J. and O. C. Uhlenbeck (1983). “Kinetic and thermodynamic characterization of the R17 coat protein-ribonucleic acid interaction.” Biochemistry 22(11): 2610-2615.

  • Chang, H. C., Z. Bao, Y. Yao, A. G. Tse, E. C. Goyarts, M. Madsen, E. Kawasaki, P. P. Brauer, J. C. Sacchettini, S. G. Nathenson and et al. (1994). “A general method for facilitating heterodimeric pairing between two proteins: application to expression of alpha and beta T-cell receptor extracellular segments.” Proc Natl Acad Sci USA 91(24): 11408-11412.

  • Chen, Y., H. Cai, J. Pan, N. Xiang, P. Tien, T. Ahola and D. Guo (2009). “Functional screen reveals SARS coronavirus nonstructural protein nsp14 as a novel cap N7 methyltransferase.” Proc Natl Acad Sci USA 106(9): 3484-3489.

  • Chen, Z. and T. D. Schneider (2005). “Information theory based T7-like promoter models: classification of bacteriophages and differential evolution of promoters and their polymerases.” Nucleic Acids Res 33(19): 6172-6187.

  • Cho, E. J., T. Takagi, C. R. Moore and S. Buratowski (1997). “mRNA capping enzyme is recruited to the transcription complex by phosphorylation of the RNA polymerase II carboxy-terminal domain.” Genes Dev 11(24): 3319-3326.

  • Choi, Y. G. and A. L. Rao (2003). “Packaging of brome mosaic virus RNA3 is mediated through a bipartite signal.” J Virol 77(18): 9750-9757.

  • Cilley, C. D. and J. R. Williamson (1997). “Analysis of bacteriophage N protein and peptide binding to boxB RNA using polyacrylamide gel coelectrophoresis (PACE).” RNA 3(1): 57-67.

  • Cong, P. and S. Shuman (1993). “Covalent catalysis in nucleotidyl transfer. A KTDG motif essential for enzyme-GMP complex formation by mRNA capping enzyme is conserved at the active sites of RNA and DNA ligases.” J Biol Chem 268(10): 7256-7260.

  • Cong, P. and S. Shuman (1995). “Mutational analysis of mRNA capping enzyme identifies amino acids involved in GTP binding, enzyme-guanylate formation, and GMP transfer to RNA.” Mol Cell Biol 15(11): 6222-6231.

  • Cronan, J. E., Jr. (1990). “Biotination of proteins in vivo. A post-translational modification to label, purify, and study proteins.” J Biol Chem 265(18): 10327-10333.

  • Daffis, S., K. J. Szretter, J. Schriewer, J. Li, S. Youn, J. Errett, T. Y. Lin, S. Schneller, R. Zust, H. Dong, V. Thiel, G. C. Sen, V. Fensterl, W. B. Klimstra, T. C. Pierson, R. M. Buller, M. Gale, Jr., P. Y. Shi and M. S. Diamond (2010). “2′-O methylation of the viral mRNA cap evades host restriction by IFIT family members.” Nature 468(7322): 452-456.

  • Darzynkiewicz, E., J. Stepinski, I. Ekiel, Y. Jin, D. Haber, T. Sijuwade and S. M. Tahara (1988). “Beta-globin mRNAs capped with m7G, m2.7(2)G or m2.2.7(3)G differ in intrinsic translation efficiency.” Nucleic Acids Res 16(18): 8953-8962.

  • Das, A. (1993). “Control of transcription termination by RNA-binding proteins.” Annu Rev Biochem 62: 893-930.

  • De la Pena, M., 0. J. Kyrieleis and S. Cusack (2007). “Structural insights into the mechanism and evolution of the vaccinia virus mRNA cap N7 methyl-transferase.” EMBO J 26(23): 4913-4925.

  • Decroly, E., F. Ferron, J. Lescar and B. Canard (2011). “Conventional and unconventional mechanisms for capping viral mRNA.” Nat Rev Microbiol 10(1): 51-65.

  • Decroly, E., I. Imbert, B. Coutard, M. Bouvet, B. Selisko, K. Alvarez, A. E. Gorbalenya, E. J. Snijder and B. Canard (2008). “Coronavirus nonstructural protein 16 is a cap-0 binding enzyme possessing (nucleoside-2′O)-methyltransferase activity.” J Virol 82(16): 8071-8084.

  • Dias, N. and C. A. Stein (2002). “Antisense oligonucleotides: basic concepts and mechanisms.” Mol Cancer Ther 1(5): 347-355.

  • Dickson, K. S., S. R. Thompson, N. K. Gray and M. Wickens (2001). “Poly(A) polymerase and the regulation of cytoplasmic polyadenylation.” J Biol Chem 276(45): 41810-41816.

  • Dingwall, C., I. Ernberg, M. J. Gait, S. M. Green, S. Heaphy, J. Karn, A. D. Lowe, M. Singh and M. A. Skinner (1990). “HIV-1 tat protein stimulates transcription by binding to a U-rich bulge in the stem of the TAR RNA structure.” EMBO J 9(12): 4145-4153.

  • Dixon, L. K., D. A. Chapman, C. L. Netherton and C. Upton (2013). “African swine fever virus replication and genomics.” Virus Res 173(1): 3-14.

  • Dong, H., D. C. Chang, M. H. Hua, S. P. Lim, Y. H. Chionh, F. Hia, Y. H. Lee, P. Kukkaro, S. M. Lok, P. C. Dedon and P. Y. Shi (2012). “2′-O methylation of internal adenosine by flavivirus NS5 methyltransferase.” PLoS Pathog 8(4): e1002642.

  • Donnelly, M. L., G. Luke, A. Mehrotra, X. Li, L. E. Hughes, D. Gani and M. D. Ryan (2001). “Analysis of the aphthovirus 2A/2B polyprotein ‘cleavage’ mechanism indicates not a proteolytic reaction, but a novel translational effect: a putative ribosomal ‘skip’.” J Gen Virol 82(Pt 5): 1013-1025.

  • Duconge, F. and J. J. Toulme (1999). “In vitro selection identifies key determinants for loop-loop interactions: RNA aptamers selective for the TAR RNA element of HIV-1.” RNA 5(12): 1605-1614.

  • Elroy-Stein, O. and B. Moss (1990). “Cytoplasmic expression system based on constitutive synthesis of bacteriophage T7 RNA polymerase in mammalian cells.” Proc Natl Acad Sci USA 87(17): 6743-6747.

  • Finn, J., I. MacLachlan and P. Cullis (2005). “Factors limiting autogene-based cytoplasmic expression systems.” FASEB J 19(6): 608-610.

  • Fortes, P., Y. Cuevas, F. Guan, P. Liu, S. Pentlicky, S. P. Jung, M. L. Martinez-Chantar, J. Prieto, D. Rowe and S. I. Gunderson (2003). “Inhibiting expression of specific genes in mammalian cells with 5′ end-mutated U1 small nuclear RNAs targeted to terminal exons of pre-mRNA.” Proc Natl Acad Sci USA 100(14): 8264-8269.

  • Frankel, A. D. and J. A. Young (1998). “HIV-1: fifteen proteins and an RNA.” Annu Rev Biochem 67:1-25.

  • Franklin, N. C. (1985). ““N” transcription antitermination proteins of bacteriophages lambda, phi 21 and P22.” J Mol Biol 181(1): 85-91.

  • Friedman, D. I. and D. L. Court (1995). “Transcription antitermination: the lambda paradigm updated.” Mol Microbiol 18(2): 191-200.

  • Furuichi, Y., A. LaFiandra and A. J. Shatkin (1977). “5′-Terminal structure and mRNA stability.” Nature 266(5599): 235-239.

  • Furuichi, Y. and A. J. Shatkin (2000). “Viral and cellular mRNA capping: past and prospects.” Adv Virus Res 55: 135-184.

  • Gallie, D. R. (1991). “The cap and poly(A) tail function synergistically to regulate mRNA translational efficiency.” Genes Dev 5(11): 2108-2116.

  • Gao, X. and L. Huang (1993). “Cytoplasmic expression of a reporter gene by co-delivery of T7 RNA polymerase and T7 promoter sequence with cationic liposomes.” Nucleic Acids Res 21(12): 2867-2872.

  • Gershon, P. D., B. Y. Ahn, M. Garfield and B. Moss (1991). “Poly(A) polymerase and a dissociable polyadenylation stimulatory factor encoded by vaccinia virus.” Cell 66(6): 1269-1278.

  • Ghosh, I., A. D. Hamilton and L. Regan (2000). “Antiparallel Leucine Zipper-Directed Protein Reassembly: Application to the Green Fluorescent Protein.” Journal of the American Chemical Society 122(23): 5658-5659.

  • Gingras, A. C., B. Raught and N. Sonenberg (1999). “eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation.” Annu Rev Biochem 68: 913-963.

  • Golomb, M. and M. Chamberlin (1974). “Characterization of T7-specific ribonucleic acid polymerase. IV. Resolution of the major in vitro transcripts by gel electrophoresis.” J Biol Chem 249(9): 2858-2863.

  • Gong, C. and S. Shuman (2003). “Mapping the active site of vaccinia virus RNA triphosphatase.” Virology 309(1): 125-134.

  • Grdzelishvili, V. Z., S. Smallwood, D. Tower, R. L. Hall, D. M. Hunt and S. A. Moyer (2005). “A single amino acid change in the L-polymerase protein of vesicular stomatitis virus completely abolishes viral mRNA cap methylation.” J Virol 79(12): 7327-7337.

  • Grdzelishvili, V. Z., S. Smallwood, D. Tower, R. L. Hall, D. M. Hunt and S. A. Moyer (2006). “Identification of a new region in the vesicular stomatitis virus L polymerase protein which is essential for mRNA cap methylation.” Virology 350(2): 394-405.

  • Greenblatt, J., J. R. Nodwell and S. W. Mason (1993). “Transcriptional antitermination.” Nature 364(6436): 401-406.

  • Gregoire, C., S. Y. Lin, G. Mazza, N. Rebai, I. F. Luescher and B. Malissen (1996). “Covalent assembly of a soluble T cell receptor-peptide-major histocompatibility class I complex.” Proc Natl Acad Sci USA 93(14): 7184-7189.

  • Gu, M., K. R. Rajashankar and C. D. Lima (2010). “Structure of the Saccharomyces cerevisiae Cet1-Ceg1 mRNA capping apparatus.” Structure 18(2): 216-227.

  • Gustaysson, M., J. Lehtio, S. Denman, T. T. Teeri, K. Hult and M. Martinelle (2001). “Stable linker peptides for a cellulose-binding domain-lipase fusion protein expressed in Pichia pastoris.” Protein Eng 14(9): 711-715.

  • Haline-Vaz, T., T. C. Silva and N. I. Zanchin (2008). “The human interferon-regulated ISG95 protein interacts with RNA polymerase II and shows methyltransferase activity.” Biochem Biophys Res Commun 372(4): 719-724.

  • Han, Y. T., C. S. Tsai, Y. C. Chen, M. K. Lin, Y. H. Hsu and M. Meng (2007). “Mutational analysis of a helicase motif-based RNA 5′-triphosphatase/NTPase from bamboo mosaic virus.” Virology 367(1): 41-50.

  • Haracska, L., R. E. Johnson, L. Prakash and S. Prakash (2005). “Trf4 and Trf5 proteins of Saccharomyces cerevisiae exhibit poly(A) RNA polymerase activity but no DNA polymerase activity.” Mol Cell Biol 25(22): 10183-10189.

  • Hausmann, S. and S. Shuman (2005). “Giardia lamblia RNA cap guanine-N2 methyltransferase (Tgs2).” J Biol Chem 280(37): 32101-32106.

  • Hausmann, S. and S. Shuman (2005). “Specificity and mechanism of RNA cap guanine-N2 methyltransferase (Tgs1).” J Biol Chem 280(6): 4021-4024.

  • Hausmann, S., S. Zheng, M. Costanzo, R. L. Brost, D. Garcin, C. Boone, S. Shuman and B. Schwer (2008). “Genetic and biochemical analysis of yeast and human cap trimethylguanosine synthase: functional overlap of 2,2,7-trimethylguanosine caps, small nuclear ribonucleoprotein components, pre-mRNA splicing factors, and RNA decay pathways.” J Biol Chem 283(46): 31706-31718.

  • Hemmerich, P., S. Bosbach, A. von Mikecz and U. Krawinkel (1997). “Human ribosomal protein L7 binds RNA with an alpha-helical arginine-rich and lysine-rich domain.” Eur J Biochem 245(3): 549-556.

  • Hennecke, F., C. Krebber and A. Pluckthun (1998). “Non-repetitive single-chain Fv linkers selected by selectively infective phage (SIP) technology.” Protein Eng 11(5): 405-410.

  • Higman, M. A., N. Bourgeois and E. G. Niles (1992). “The vaccinia virus mRNA (guanine-N7-)-methyltransferase requires both subunits of the mRNA capping enzyme for activity.” J Biol Chem 267(23): 16430-16437.

  • Higman, M. A., L. A. Christen and E. G. Niles (1994). “The mRNA (guanine-7-)methyltransferase domain of the vaccinia virus mRNA capping enzyme. Expression in Escherichia coli and structural and kinetic comparison to the intact capping enzyme.” J Biol Chem 269(21): 14974-14981.

  • Higman, M. A. and E. G. Niles (1994). “Location of the S-adenosyl-L-methionine binding region of the vaccinia virus mRNA (guanine-7-)methyltransferase.” J Biol Chem 269(21): 14982-14987.

  • Hornung, V., J. Ellegast, S. Kim, K. Brzozka, A. Jung, H. Kato, H. Poeck, S. Akira, K. K. Conzelmann, M. Schlee, S. Endres and G. Hartmann (2006). “5′-Triphosphate RNA is the ligand for RIG-I.” Science 314(5801): 994-997.

  • Hu, G., P. D. Gershon, A. E. Hodel and F. A. Quiocho (1999). “mRNA cap recognition: dominant role of enhanced stacking interactions between methylated bases and protein aromatic side chains.” Proc Natl Acad Sci USA 96(13): 7149-7154.

  • Hu, W., F. Li, X. Yang, Z. Li, H. Xia, G. Li, Y. Wang and Z. Zhang (2004). “A flexible peptide linker enhances the immunoreactivity of two copies HBsAg preS1 (21-47) fusion protein.” J Biotechnol 107(1): 83-90.

  • Huang, Y. and J. A. Steitz (2005). “SRprises along a messenger's journey.” Mol Cell 17(5): 613-615.

  • Huang, Y. L., Y. T. Han, Y. T. Chang, Y. H. Hsu and M. Meng (2004). “Critical residues for GTP methylation and formation of the covalent m7GMP-enzyme intermediate in the capping enzyme domain of bamboo mosaic virus.” J Virol 78(3): 1271-1280.

  • Huang, Y. L., Y. H. Hsu, Y. T. Han and M. Meng (2005). “mRNA guanylation catalyzed by the S-adenosylmethionine-dependent guanylyltransferase of bamboo mosaic virus.” J Biol Chem 280(13): 13153-13162.

  • Huston, J. S., D. Levinson, M. Mudgett-Hunter, M. S. Tai, J. Novotny, M. N. Margolies, R. J. Ridge, R. E. Bruccoleri, E. Haber, R. Crea and et al. (1988). “Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli.” Proc Natl Acad Sci USA 85(16): 5879-5883.

  • Jais, P. (2011). Capping-prone RNA polymerase enzymes and their applications. Eukarÿs. France.

  • Jais, P. H., E. Decroly, E. Jacquet, M. Le Boulch, A. Jais, H. Eaton, P. Ponien, F. Verdier, B. Canard, S. Gonvalves, S. Chiron, M. Le Gall, P. Mayeux and M. Shmulevitz (2018). “C3P3-G1: first generation of an artificial cytoplasmic expression system that recapitulates mRNA capping and polyadenylation.” Under review.

  • Jiang, F., A. Gorin, W. Hu, A. Majumdar, S. Baskerville, W. Xu, A. Ellington and D. J. Patel (1999). “Anchoring an extended HTLV-1 Rex peptide within an RNA major groove containing junctional base triples.” Structure 7(12): 1461-1472.

  • Kaneko, S., C. Chu, A. J. Shatkin and J. L. Manley (2007). “Human capping enzyme promotes formation of transcriptional R loops in vitro.” Proc Natl Acad Sci USA 104(45): 17620-17625.

  • Karn, J., C. Dingwall, J. T. Finch, S. Heaphy and M. J. Gait (1991). “RNA binding by the tat and rev proteins of HIV-1.” Biochimie 73(1): 9-16.

  • Kashiwabara, S., T. Zhuang, K. Yamagata, J. Noguchi, A. Fukamizu and T. Baba (2000). “Identification of a novel isoform of poly(A) polymerase, TPAP, specifically present in the cytoplasm of spermatogenic cells.” Dev Biol 228(1): 106-115.

  • Kashiwabara, S. I., S. Tsuruta, K. Okada, Y. Yamaoka and T. Baba (2016). “Adenylation by testis-specific cytoplasmic poly(A) polymerase, PAPOLB/TPAP, is essential for spermatogenesis.” J Reprod Dev 62(6): 607-614.

  • Keene, J. D., C. C. Query and R. O. Bentley (1999). Ribonucleoproteins and RNA-binding proteins useful for the specific recognition and binding to RNA, and for control of cellular genetic processing and expression. U.S. Pat. No. 5,866,680A.

  • Keith, J. M., M. J. Ensinger and B. Mose (1978). “HeLa cell RNA (2′-O-methyladenosine-N6-)-methyltransferase specific for the capped 5′-end of messenger RNA.” J Biol Chem 253(14): 5033-5039.

  • Kohler, A. and E. Hurt (2007). “Exporting RNA from the nucleus to the cytoplasm.” Nat Rev Mol Cell Biol 8(10): 761-773.

  • Komarnitsky, P., E. J. Cho and S. Buratowski (2000). “Different phosphorylated forms of RNA polymerase II and associated mRNA processing factors during transcription.” Genes Dev 14(19): 2452-2460.

  • Kozak, M. (2005). “Regulation of translation via mRNA structure in prokaryotes and eukaryotes.” Gene 361: 13-37.

  • Kyriakopoulou, C. B., H. Nordvarg and A. Virtanen (2001). “A novel nuclear human poly(A) polymerase (PAP), PAP gamma.” J Biol Chem 276(36): 33504-33511.

  • Lamla, T. and V. A. Erdmann (2004). “The Nano-tag, a streptavidin-binding peptide for the purification and detection of recombinant proteins.” Protein Expr Purif 33(1): 39-47.

  • Langberg, S. R. and B. Moss (1981). “Post-transcriptional modifications of mRNA. Purification and characterization of cap I and cap II RNA (nucleoside-2′-)-methyltransferases from HeLa cells.” J Biol Chem 256(19): 10054-10060.

  • LeGuyer, K. A., L. S. Behlen and O. C. Uhlenbeck (1996). “Mutagenesis of a stacking contact in the MS2 coat protein-RNA complex.” EMBO J 15(24): 6847-6853.

  • Lee, Y. J., Y. Lee and J. H. Chung (2000). “An intronless gene encoding a poly(A) polymerase is specifically expressed in testis.” FEBS Lett 487(2): 287-292.

  • Leppek, K. and G. Stoecklin (2014). “An optimized streptavidin-binding RNA aptamer for purification of ribonucleoprotein complexes identifies novel ARE-binding proteins.” Nucleic Acids Res 42(2): e13.

  • Li, J., E. C. Fontaine-Rodriguez and S. P. Whelan (2005). “Amino acid residues within conserved domain VI of the vesicular stomatitis virus large polymerase protein essential for mRNA cap methyltransferase activity.” J Virol 79(21): 13373-13384.

  • Li, J., A. Rahmeh, M. Morelli and S. P. Whelan (2008). “A conserved motif in region v of the large polymerase proteins of nonsegmented negative-sense RNA viruses that is essential for mRNA capping.” J Virol 82(2): 775-784.

  • Li, Y. I., Y. J. Chen, Y. H. Hsu and M. Meng (2001). “Characterization of the AdoMet-dependent guanylyltransferase activity that is associated with the N terminus of bamboo mosaic virus replicase.” J Virol 75(2): 782-788.

  • Li, Y. I., T. W. Shih, Y. H. Hsu, Y. T. Han, Y. L. Huang and M. Meng (2001). “The helicase-like domain of plant potexvirus replicase participates in formation of RNA 5′ cap structure by exhibiting RNA 5′-triphosphatase activity.” J Virol 75(24): 12114-12120.

  • Lian, Y., M. B. De Young, A. Siwkowski, A. Hampel and J. Rappaport (1999). “The sCYMV1 hairpin ribozyme: targeting rules and cleavage of heterologous RNA.” Gene Ther 6(6): 1114-1119.

  • Lieschke, G. J., P. K. Rao, M. K. Gately and R. C. Mulligan (1997). “Bioactive murine and human interleukin-12 fusion proteins which retain antitumor activity in vivo.” Nat Biotechnol 15(1): 35-40.

  • Lingner, J., J. Kellermann and W. Keller (1991). “Cloning and expression of the essential gene for poly(A) polymerase from S. cerevisiae.” Nature 354(6353): 496-498.

  • Liu, Z. and G. G. Carmichael (1994). “Nuclear antisense RNA. An efficient new method to inhibit gene expression.” Mol Biotechnol 2(2): 107-118.

  • Lo, H. J., H. K. Huang and T. F. Donahue (1998). “RNA polymerase I-promoted HIS4 expression yields uncapped, polyadenylated mRNA that is unstable and inefficiently translated in Saccharomyces cerevisiae.” Mol Cell Biol 18(2): 665-675.

  • Lugari, A., S. Betzi, E. Decroly, E. Bonnaud, A. Hermant, J. C. Guillemot, C. Debarnot, J. P. Borg, M. Bouvet, B. Canard, X. Morelli and P. Lecine (2010). “Molecular mapping of the RNA Cap 2′-O-methyltransferase activation interface between severe acute respiratory syndrome coronavirus nsp10 and nsp16.” J Biol Chem 285(43): 33230-33241.

  • Lumb, K. J. and P. S. Kim (1995). “A buried polar interaction imparts structural uniqueness in a designed heterodimeric coiled coil.” Biochemistry 34(27): 8642-8648.

  • Lyakhov, D. L., B. He, X. Zhang, F. W. Studier, J. J. Dunn and W. T. McAllister (1997). “Mutant bacteriophage T7 RNA polymerases with altered termination properties.” J Mol Biol 269(1): 28-40.

  • Makarova, 0. V., E. M. Makarov, R. Sousa and M. Dreyfus (1995). “Transcribing of Escherichia coli genes with mutant T7 RNA polymerases: stability of lacZ mRNA inversely correlates with polymerase speed.” Proc Natl Acad Sci USA 92(26): 12250-12254.

  • Malone, R. W., P. L. Feigner and I. M. Verma (1989). “Cationic liposome-mediated RNA transfection.” Proc Natl Acad Sci USA 86(16): 6077-6081.

  • Mao, X., B. Schwer and S. Shuman (1995). “Yeast mRNA cap methyltransferase is a 50-kilodalton protein encoded by an essential gene.” Mol Cell Biol 15(8): 4167-4174.

  • Mao, X. and S. Shuman (1994). “Intrinsic RNA (guanine-7) methyltransferase activity of the vaccinia virus capping enzyme D1 subunit is stimulated by the D12 subunit. Identification of amino acid residues in the D1 protein required for subunit association and methyl group transfer.” J Biol Chem 269(39): 24472-24479.

  • Mao, X. and S. Shuman (1996). “Vaccinia virus mRNA (guanine-7-)methyltransferase: mutational effects on cap methylation and AdoHcy-dependent photo-cross-linking of the cap to the methyl acceptor site.” Biochemistry 35(21): 6900-6910.



Martinez-Costas, J., G. Sutton, N. Ramadevi and P. Roy (1998). “Guanylyltransferase and RNA 5′-triphosphatase activities of the purified expressed VP4 protein of bluetongue virus.” J Mol Biol 280(5): 859-866.


Marzluff, W. F., E. J. Wagner and R. J. Duronio (2008). “Metabolism and regulation of canonical histone mRNAs: life without a poly(A) tail.” Nat Rev Genet 9(11): 843-854.

  • Mauer, J., X. Luo, A. Blanjoie, X. Jiao, A. V. Grozhik, D. P. Patil, B. Linder, B. F. Pickering, J. J. Vasseur, Q. Chen, S. S. Gross, O. Elemento, F. Debart, M. Kiledjian and S. R. Jaffrey (2017). “Reversible methylation of m(6)Am in the 5′ cap controls mRNA stability.” Nature 541(7637): 371-375.
  • McClain, D. L., H. L. Woods and M. G. Oakley (2001). “Design and characterization of a heterodimeric coiled coil that forms exclusively with an antiparallel relative helix orientation.” J Am Chem Soc 123(13): 3151-3152.
  • McCracken, S., N. Fong, E. Rosonina, K. Yankulov, G. Brothers, D. Siderovski, A. Hessel, S. Foster, S. Shuman and D. L. Bentley (1997). “5′-Capping enzymes are targeted to pre-mRNA by binding to the phosphorylated carboxy-terminal domain of RNA polymerase II.” Genes Dev 11(24): 3306-3318.
  • Mix, H., A. V. Lobanov and V. N. Gladyshev (2007). “SECIS elements in the coding regions of selenoprotein transcripts are functional in higher eukaryotes.” Nucleic Acids Res 35(2): 414-423.
  • Moll, J. R., S. B. Ruvinov, I. Pastan and C. Vinson (2001). “Designed heterodimerizing leucine zippers with a ranger of pls and stabilities up to 10(-15) M.” Protein Sci 10(3): 649-655.
  • Myette, J. R. and E. G. Niles (1996). “Domain structure of the vaccinia virus mRNA capping enzyme. Expression in Escherichia coli of a subdomain possessing the RNA 5′-triphosphatase and guanylyltransferase activities and a kinetic comparison to the full-size enzyme.” J Biol Chem 271(20): 11936-11944.
  • Natalizio, B. J., N. D. Robson-Dixon and M. A. Garcia-Blanco (2009). “The Carboxyl-terminal Domain of RNA Polymerase II Is Not Sufficient to Enhance the Efficiency of Pre-mRNA Capping or Splicing in the Context of a Different Polymerase.” J Biol Chem 284(13): 8692-8702.
  • Newton, D. L., Y. Xue, K. A. Olson, J. W. Fett and S. M. Rybak (1996). “Angiogenin single-chain immunofusions: influence of peptide linkers and spacers between fusion protein domains.” Biochemistry 35(2): 545-553.
  • Niles, E. G. and L. Christen (1993). “Identification of the vaccinia virus mRNA guanyltransferase active site lysine.” J Biol Chem 268(33): 24986-24989.
  • O'Shea, E. K., J. D. Klemm, P. S. Kim and T. Alber (1991). “X-ray structure of the GCN4 leucine zipper, a two-stranded, parallel coiled coil.” Science 254(5031): 539-544.
  • O'Shea, E. K., K. J. Lumb and P. S. Kim (1993). “Peptide ‘Velcro’: design of a heterodimeric coiled coil.” Curr Biol 3(10): 658-667.
  • Oakley, M. G. and P. S. Kim (1998). “A buried polar interaction can direct the relative orientation of helices in a coiled coil.” Biochemistry 37(36): 12603-12610.
  • Ogino, T. and A. K. Banerjee (2007). “Unconventional mechanism of mRNA capping by the RNA-dependent RNA polymerase of vesicular stomatitis virus.” Mol Cell 25(1): 85-97.
  • Ogino, T. and A. K. Banerjee (2008). “Formation of guanosine(5′)tetraphospho(5′)adenosine cap structure by an unconventional mRNA capping enzyme of vesicular stomatitis virus.” J Virol 82(15): 7729-7734.
  • Ohlmann, T., M. Rau, S. J. Morley and V. M. Pain (1995). “Proteolytic cleavage of initiation factor elF-4 gamma in the reticulocyte lysate inhibits translation of capped mRNAs but enhances that of uncapped mRNAs.” Nucleic Acids Res 23(3): 334-340.
  • Osumi-Davis, P. A., M. C. de Aguilera, R. W. Woody and A. Y. Woody (1992). “Asp537, Asp812 are essential and Lys631, His811 are catalytically significant in bacteriophage T7 RNA polymerase activity.” J Mol Biol 226(1): 37-45.
  • Osumi-Davis, P. A., N. Sreerama, D. B. Volkin, C. R. Middaugh, R. W. Woody and A. Y. Woody (1994). “Bacteriophage T7 RNA polymerase and its active-site mutants. Kinetic, spectroscopic and calorimetric characterization.” J Mol Biol 237(1): 5-19.
  • Palancade, B. and O. Bensaude (2003). “Investigating RNA polymerase II carboxyl-terminal domain (CTD) phosphorylation.” Eur J Biochem 270(19): 3859-3870.
  • Pantoliano, M. W., R. E. Bird, S. Johnson, E. D. Asel, S. W. Dodd, J. F. Wood and K. D. Hardman (1991). “Conformational stability, folding, and ligand-binding affinity of single-chain Fv immunoglobulin fragments expressed in Escherichia coli.” Biochemistry 30(42): 10117-10125.
  • Pashine, A., R. Busch, M. P. Belmares, J. N. Munning, R. C. Doebele, M. Buckingham, G. P. Nolan and E. D. Mellins (2003). “Interaction of HLA-DR with an acidic face of HLA-DM disrupts sequence-dependent interactions with peptides.” Immunity 19(2): 183-192.
  • Pavlinkova, G., G. W. Beresford, B. J. Booth, S. K. Batra and D. Colcher (1999). “Pharmacokinetics and biodistribution of engineered single-chain antibody constructs of MAb CC49 in colon carcinoma xenografts.” J Nucl Med 40(9): 1536-1546.
  • Peabody, D. S. (1993). “The RNA binding site of bacteriophage MS2 coat protein.” EMBO J 12(2): 595-600.
  • Pena, L., R. J. Yanez, Y. Revilla, E. Vinuela and M. L. Salas (1993). “African swine fever virus guanylyltransferase.” Virology 193(1): 319-328.
  • Pillutla, R. C., Z. Yue, E. Maldonado and A. J. Shatkin (1998). “Recombinant human mRNA cap methyltransferase binds capping enzyme/RNA polymerase IIo complexes.” J Biol Chem 273(34): 21443-21446.
  • Puglisi, J. D., L. Chen, S. Blanchard and A. D. Frankel (1995). “Solution structure of a bovine immunodeficiency virus Tat-TAR peptide-RNA complex.” Science 270(5239): 1200-1203.
  • Puglisi, J. D., R. Tan, B. J. Calnan, A. D. Frankel and J. R. Williamson (1992). “Conformation of the TAR RNA-arginine complex by NMR spectroscopy.” Science 257(5066): 76-80.
  • Raab, D., M. Graf, F. Notka, T. Schodl and R. Wagner (2010). “The GeneOptimizer Algorithm: using a sliding window approach to cope with the vast sequence space in multiparameter DNA sequence optimization.” Syst Synth Biol 4(3): 215-225.
  • Raabe, T., K. G. Murthy and J. L. Manley (1994). “Poly(A) polymerase contains multiple functional domains.” Mol Cell Biol 14(5): 2946-2957.
  • Rahmeh, A. A., J. Li, P. J. Kranzusch and S. P. Whelan (2009). “Ribose 2′-O methylation of the vesicular stomatitis virus mRNA cap precedes and facilitates subsequent guanine-N-7 methylation by the large polymerase protein.” J Virol 83(21): 11043-11050.
  • Ramadevi, N., N. J. Burroughs, P. P. Mertens, I. M. Jones and P. Roy (1998). “Capping and methylation of mRNA by purified recombinant VP4 protein of bluetongue virus.” Proc Natl Acad Sci USA 95(23): 13537-13542.
  • Ramadevi, N., J. Rodriguez and P. Roy (1998). “A leucine zipper-like domain is essential for dimerization and encapsidation of bluetongue virus nucleocapsid protein VP4.” J Virol 72(4): 2983-2990.
  • Raoult, D., S. Audic, C. Robert, C. Abergel, P. Renesto, H. Ogata, B. La Scola, M. Suzan and J. M. Claverie (2004). “The 1.2-megabase genome sequence of Mimivirus.” Science 306(5700): 1344-1350.
  • Reinisch, K. M., M. L. Nibert and S. C. Harrison (2000). “Structure of the reovirus core at 3.6 A resolution.” Nature 404(6781): 960-967.
  • Rhoads, R. E. (1999). “Signal transduction pathways that regulate eukaryotic protein synthesis.” J Biol Chem 274(43): 30337-30340.
  • Robert, F., M. Gagnon, D. Sans, S. Michnick and L. Brakier-Gingras (2000). “Mapping of the RNA recognition site of Escherichia coli ribosomal protein S7.” RNA 6(11): 1649-1659.
  • Robinson, C. R. and R. T. Sauer (1998). “Optimizing the stability of single-chain proteins by linker length and composition mutagenesis.” Proc Natl Acad Sci USA 95(11): 5929-5934.
  • Romac, J. M., D. H. Graff and J. D. Keene (1994). “The U1 small nuclear ribonucleoprotein (snRNP) 70K protein is transported independently of U1 snRNP particles via a nuclear localization signal in the RNA-binding domain.” Molecular and Cellular Biology 14(7): 4662-4670.
  • Rouault, T. A. (2006). “The role of iron regulatory proteins in mammalian iron homeostasis and disease.” Nat Chem Biol 2(8): 406-414.
  • Sacher, R. and P. Ahlquist (1989). “Effects of deletions in the N-terminal basic arm of brome mosaic virus coat protein on RNA packaging and systemic infection.” J Virol 63(11): 4545-4552.
  • Salehi-Ashtiani, K. and J. W. Szostak (2001). “In vitro evolution suggests multiple origins for the hammerhead ribozyme.” Nature 414(6859): 82-84.
  • Schmid, M., B. Kuchler and C. R. Eckmann (2009). “Two conserved regulatory cytoplasmic poly(A) polymerases, GLD-4 and GLD-2, regulate meiotic progression in C. elegans.” Genes Dev 23(7): 824-836.
  • Schmidt, T. G. and A. Skerra (1993). “The random peptide library-assisted engineering of a C-terminal affinity peptide, useful for the detection and purification of a functional Ig Fv fragment.” Protein Eng 6(1): 109-122.
  • Schnierle, B. S., P. D. Gershon and B. Moss (1994). “Mutational analysis of a multifunctional protein, with mRNA 5′ cap-specific (nucleoside-2′-O-)-methyltransferase and 3′-adenylyltransferase stimulatory activities, encoded by vaccinia virus.” The Journal of biological chemistry 269(32): 20700-20706.
  • Schroeder, S. C., B. Schwer, S. Shuman and D. Bentley (2000). “Dynamic association of capping enzymes with transcribing RNA polymerase II.” Genes Dev 14(19): 2435-2440.
  • Schwer, B., S. Hausmann, S. Schneider and S. Shuman (2006). “Poxvirus mRNA cap methyltransferase. Bypass of the requirement for the stimulatory subunit by mutations in the catalytic subunit and evidence for intersubunit allostery.” J Biol Chem 281(28): 18953-18960.
  • Schwer, B., N. Saha, X. Mao, H. W. Chen and S. Shuman (2000). “Structure-function analysis of yeast mRNA cap methyltransferase and high-copy suppression of conditional mutants by AdoMet synthase and the ubiquitin conjugating enzyme Cdc34p.” Genetics 155(4): 1561-1576.
  • Shao, W. H., X. E. Zhang, H. Liu, Z. P. Zhang and A. E. Cass (2000). “Anchor-chain molecular system for orientation control in enzyme immobilization.” Bioconjug Chem 11(6): 822-826.
  • Shi, X., P. Yao, T. Jose and P. Gershon (1996). “Methyltransferase-specific domains within VP-39, a bifunctional protein that participates in the modification of both mRNA ends.” RNA 2(1): 88-101.
  • Shibagaki, Y., N. Itoh, H. Yamada, S. Nagata and K. Mizumoto (1992). “mRNA capping enzyme. Isolation and characterization of the gene encoding mRNA guanylytransferase subunit from Saccharomyces cerevisiae.” J Biol Chem 267(14): 9521-9528.
  • Smith, C. A., V. Calabro and A. D. Frankel (2000). “An RNA-binding chameleon.” Mol Cell 6(5): 1067-1076.
  • Tan, R. and A. D. Frankel (1995). “Structural variety of arginine-rich RNA-binding peptides.” Proc Natl Acad Sci USA 92(12): 5282-5286.
  • Tang, Y., N. Jiang, C. Parakh and D. Hilvert (1996). “Selection of linkers for a catalytic single-chain antibody using phage display technology.” J Biol Chem 271(26): 15682-15686.
  • Theil, E. C. (1994). “Iron regulatory elements (IREs): a family of mRNA non-coding sequences.” Biochem J 304 (Pt 1): 1-11.
  • Tiggemann, M., S. Jeske, M. Larsen and F. Meinhardt (2001). “Kluyveromyces lactis cytoplasmic plasmid pGKL2: heterologous expression of Orf3p and proof of guanylyltransferase and mRNA-triphosphatase activities.” Yeast 18(9): 815-825.
  • Ting, A. Y., K. H. Kain, R. L. Klemke and R. Y. Tsien (2001). “Genetically encoded fluorescent reporters of protein tyrosine kinase activities in living cells.” Proc Natl Acad Sci USA 98(26): 15003-15008.
  • Tommasino, M., S. Ricci and C. L. Galeotti (1988). “Genome organization of the killer plasmid pGK12 from Kluyveromyces lactis.” Nucleic Acids Res 16(13): 5863-5878.
  • Trippe, R., B. Sandrock and B. J. Benecke (1998). “A highly specific terminal uridylyl transferase modifies the 3′-end of U6 small nuclear RNA.” Nucleic Acids Res 26(13): 3119-3126.
  • Tsukamoto, T., Y. Shibagaki, S. Imajoh-Ohmi, T. Murakoshi, M. Suzuki, A. Nakamura, H. Gotoh and K. Mizumoto (1997). “Isolation and characterization of the yeast mRNA capping enzyme beta subunit gene encoding RNA 5′-triphosphatase, which is essential for cell viability.” Biochem Biophys Res Commun 239(1): 116-122.
  • Tsukamoto, T., Y. Shibagaki, Y. Niikura and K. Mizumoto (1998). “Cloning and characterization of three human cDNAs encoding mRNA (guanine-7-)-methyltransferase, an mRNA cap methylase.” Biochem Biophys Res Commun 251(1): 27-34.
  • Turner, B., S. E. Melcher, T. J. Wilson, D. G. Norman and D. M. Lilley (2005). “Induced fit of RNA on binding the L7Ae protein to the kink-turn motif.” RNA 11(8): 1192-1200.
  • Turner, D. J., M. A. Ritter and A. J. George (1997). “Importance of the linker in expression of single-chain Fv antibody fragments: optimisation of peptide sequence using phage display technology.” J Immunol Methods 205(1): 43-54.
  • Valegard, K., J. B. Murray, P. G. Stockley, N. J. Stonehouse and L. Liljas (1994). “Crystal structure of an RNA bacteriophage coat protein-operator complex.” Nature 371(6498): 623-626.
  • Valegard, K., J. B. Murray, N. J. Stonehouse, S. van den Worm, P. G. Stockley and L. Liljas (1997). “The three-dimensional structures of two complexes between recombinant MS2 capsids and RNA operator fragments reveal sequence-specific protein-RNA interactions.” J Mol Biol 270(5): 724-738.
  • Vethantham, V., N. Rao and J. L. Manley (2008). “Sumoylation regulates multiple aspects of mammalian poly(A) polymerase function.” Genes Dev 22(4): 499-511.
  • Wang, L., C. R. Eckmann, L. C. Kadyk, M. Wickens and J. Kimble (2002). “A regulatory cytoplasmic poly(A) polymerase in Caenorhabditis elegans.” Nature 419(6904): 312-316.
  • Weeks, K. M., C. Ampe, S. C. Schultz, T. A. Steitz and D. M. Crothers (1990). “Fragments of the HIV-1 Tat protein specifically bind TAR RNA.” Science 249(4974): 1281-1285.
  • Wells, J. A. and D. B. Powers (1986). “In vivo formation and stability of engineered disulfide bonds in subtilisin.” J Biol Chem 261(14): 6564-6570.
  • Whitlow, M., B. A. Bell, S. L. Feng, D. Filpula, K. D. Hardman, S. L. Hubert, M. L. Rollence, J. F. Wood, M. E. Schott, D. E. Milenic and et al. (1993). “An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability.” Protein Eng 6(8): 989-995.
  • Wickham, T. J., M. E. Carrion and I. Kovesdi (1995). “Targeting of adenovirus penton base to new receptors through replacement of its RGD motif with other receptor-specific peptide motifs.” Gene Ther 2(10): 750-756.
  • Wu, S. C., J. C. Yeung, P. M. Hwang and S. L. Wong (2002). “Design, production, and characterization of an engineered biotin ligase (BirA) and its application for affinity purification of staphylokinase produced from Bacillus subtilis via secretion.” Protein Expr Purif 24(3): 357-365.
  • Wu, X. and L. A. Guarino (2003). “Autographa californica nucleopolyhedrovirus orf69 encodes an RNA cap (nucleoside-2′-O)-methyltransferase.” J Virol 77(6): 3430-3440.
  • Xiao, F., W. D. Moll, S. Guo and P. Guo (2005). “Binding of pRNA to the N-terminal 14 amino acids of connector protein of bacteriophage phi29.” Nucleic Acids Res 33(8): 2640-2649.
  • Yamada-Okabe, T., R. Doi, O. Shimmi, M. Arisawa and H. Yamada-Okabe (1998). “Isolation and characterization of a human cDNA for mRNA 5′-capping enzyme.” Nucleic Acids Res 26(7): 1700-1706.
  • Ye, W., J. Yang, Q. Yu, W. Wang, J. Nancy, R. Luo and H. F. Chen (2013). “Kink turn sRNA folding upon L7Ae binding using molecular dynamics simulations.” Phys Chem Chem Phys 15(42): 18510-18522.
  • Yi, G., R. C. Vaughan, I. Yarbrough, S. Dharmaiah and C. C. Kao (2009). “RNA binding by the brome mosaic virus capsid protein and the regulation of viral RNA accumulation.” J Mol Biol 391(2): 314-326.
  • Yip, M. T., W. S. Dynan, P. L. Green, A. C. Black, S. J. Arrigo, A. Torbati, S. Heaphy, C. Ruland, J. D. Rosenblatt and I. S. Chen (1991). “Human T-cell leukemia virus (HTLV) type II Rex protein binds specifically to RNA sequences of the HTLV long terminal repeat but poorly to the human immunodeficiency virus type 1 Rev-responsive element.” J Virol 65(5): 2261-2272.
  • Yu, L., A. Martins, L. Deng and S. Shuman (1997). “Structure-function analysis of the triphosphatase component of vaccinia virus mRNA capping enzyme.” J Virol 71(12): 9837-9843.
  • Yu, L. and S. Shuman (1996). “Mutational analysis of the RNA triphosphatase component of vaccinia virus mRNA capping enzyme.” J Virol 70(9): 6162-6168.
  • Yue, Z., E. Maldonado, R. Pillutla, H. Cho, D. Reinberg and A. J. Shatkin (1997). “Mammalian capping enzyme complements mutant Saccharomyces cerevisiae lacking mRNA guanylyltransferase and selectively binds the elongating form of RNA polymerase II.” Proc Natl Acad Sci USA 94(24): 12898-12903.
  • Zamore, P. D., J. G. Patton and M. R. Green (1992). “Cloning and domain structure of the mammalian splicing factor U2AF.” Nature 355(6361): 609-614.
  • Zhang, X. and F. W. Studier (1997). “Mechanism of inhibition of bacteriophage T7 RNA polymerase by T7 lysozyme.” J Mol Biol 269(1): 10-27.
  • Zhelkovsky, A., S. Helmling and C. Moore (1998). “Processivity of the Saccharomyces cerevisiae poly(A) polymerase requires interactions at the carboxyl-terminal RNA binding domain.” Mol Cell Biol 18(10): 5942-5951.
  • Zhu, Y., C. Qi, W. Q. Cao, A. V. Yeldandi, M. S. Rao and J. K. Reddy (2001). “Cloning and characterization of PIMT, a protein with a methyltransferase domain, which interacts with and enhances nuclear receptor coactivator PRIP function.” Proc Natl Acad Sci USA 98(18): 10380-10385.

Claims
  • 1. A non-natural hetero-oligomeric enzyme comprising components i) through iv) linked together covalently or non-covalently: i) at least one catalytic domain of a RNA triphosphatase;ii) at least one catalytic domain of a guanylyltransferase;iii) at least one catalytic domain of a N7-guanine methyltransferase; andiv) at least one RNA-binding domain of a protein-RNA tethering system; and wherein said RNA-binding domain is a bacteriophage RNA-binding domain of a bacteriophage protein-RNA tethering system, and wherein said non-natural hetero-oligomeric enzyme adds a cap to RNA produced by a RNA polymerase.
  • 2. The non-natural hetero-oligomeric enzyme according to claim 1, wherein the RNA-binding domain is a bacteriophage RNA-binding domain of a bacteriophage protein selected from the group consisting of the MS2 coat protein, the R17 coat protein and lambdoid N antitermination proteins.
  • 3. A method for the in vitro or ex vivo production of an RNA molecule with a 5′-terminal cap, and optionally said method comprising at least one chemical modification, wherein said method comprises in vitro or ex vivo use of: A) expressing, in a host cell: an isolated nucleic acid molecule encoding a non-natural hetero-oligomeric enzyme according to claim 1; and/orB) expressing in one or more host cells a group of isolated nucleic acid molecules encoding a non-natural hetero-oligomeric enzyme according to claim 1.
  • 4. A kit for the production of an RNA molecule with a 5′-terminal cap, comprising: i) at least one non-natural hetero-oligomeric enzyme according to claim 1; and/orii) an isolated nucleic acid molecule encoding a non-natural hetero-oligomeric enzyme according to claim 1;and/oriii) a group of isolated nucleic acid molecules encoding a non-natural hetero-oligomeric enzyme according to claim 1.
  • 5. A pharmaceutical composition comprising: at least one non-natural hetero-oligomeric enzyme according to claim 1.
  • 6. A pharmaceutical composition comprising an isolated nucleic acid molecule encoding a non-natural hetero-oligomeric enzyme according to claim 1.
Priority Claims (1)
Number Date Country Kind
17306006 Jul 2017 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2018/070479 7/27/2018 WO
Publishing Document Publishing Date Country Kind
WO2019/020811 1/31/2019 WO A
US Referenced Citations (1)
Number Name Date Kind
5866680 Keene et al. Feb 1999 A
Foreign Referenced Citations (1)
Number Date Country
2011128444 Oct 2011 WO
Non-Patent Literature Citations (4)
Entry
Keryer-Bobens, Biol. Cell, 2008, 100, 125-138.
Bharati, A.P., et al., “The mRNA capping enzyme of Saccharomyces cerevisiae has dual specificity to interact with CTD of RNA Polymerase II.” Scientific Reports, 2016, 6: 31294, pp. 1-12.
Ho, C.K., et al., “A Yeast-Based Genetic System for Functional Analysis of Viral mRNA Capping Enzymes.” Journal of Virology, Jun. 2000, 74(12): 5486-5494.
Wen, Y., et al., “Mammalian capping enzyme binds RNA and uses protein tyrosine phosphatase mechanism.” Proc. Natl. Acad. Sci. USA, Oct. 1998, 95: 12226-12231.
Related Publications (1)
Number Date Country
20200165585 A1 May 2020 US