WO2025249887A1 - Composition for increasing stability or expression of exogenous rna in cells, comprising inhibitor of trim25 expression or activity - Google Patents
Composition for increasing stability or expression of exogenous rna in cells, comprising inhibitor of trim25 expression or activityInfo
- Publication number
- WO2025249887A1 WO2025249887A1 PCT/KR2025/007217 KR2025007217W WO2025249887A1 WO 2025249887 A1 WO2025249887 A1 WO 2025249887A1 KR 2025007217 W KR2025007217 W KR 2025007217W WO 2025249887 A1 WO2025249887 A1 WO 2025249887A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- trim25
- rna
- expression
- mrna
- cells
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
Definitions
- the present invention relates to the use of increasing the stability or expression of exogenous RNA in a cell by inhibiting TRIM25 expression or activity.
- RNA Exogenous RNA, such as viral RNA and therapeutic mRNA, must interact with cellular factors to enter cells while simultaneously evading cellular defense mechanisms. Therefore, developing effective mRNA therapeutics requires exploiting and overcoming these cellular barriers. Recent advances in mRNA technology have significantly improved gene delivery efficiency, establishing mRNA as a crucial therapeutic tool and reaching billions of people during the COVID-19 pandemic.
- Therapeutic mRNA is synthesized via in vitro transcription (IVT) and encapsulated in lipid nanoparticles (LNPs) composed of ionic lipids, phospholipids, cholesterol, and polyethylene glycol (PEG)-linked lipids. Within acidified endosomes, the ionic lipids protonate, disrupting the endosomal membrane and releasing the mRNA into the cytoplasm.
- IVTT in vitro transcription
- LNPs lipid nanoparticles
- PEG polyethylene glycol
- mRNA vaccines have a linear structure, including a 5′ cap, a poly(A) tail, and an N1-methylpseudouridine (m1 ⁇ ) modification, which enhances protein expression.
- Alternative platforms such as circular RNA (circRNA) and self-amplifying RNA (saRNA), are also being explored for sustained mRNA vaccine expression. While the m1 ⁇ modification is advantageous in linear mRNA, it interferes with cis-elements such as the internal ribosome entry site (IRES), making it incompatible with alternative platforms and requiring further research.
- IRS internal ribosome entry site
- One aspect is to provide a composition for increasing the stability or expression of intracellular RNA comprising an inhibitor of TRIM25 expression or activity.
- Another aspect is to provide a method for increasing the expression or activity of a target RNA within a cell in vitro.
- Another aspect is to provide a method for screening substances for increasing the stability or expression of RNA within a cell.
- Another aspect is to provide a method for screening chemical modifications of nucleic acids that enhance expression or activity of RNA.
- One aspect provides a composition for increasing the stability or expression of intracellular RNA comprising an inhibitor of TRIM25 expression or activity.
- TAM25 Tripartite motif containing 25
- TRIM25 Tripartite motif containing 25
- TRIM25 functions to conjugate ubiquitin to specific target proteins and plays a major role mainly in the innate immune response and antiviral response.
- the composition may further comprise an inhibitor of the expression or activity of a TRIM25 cofactor.
- the composition may further comprise an inhibitor of the expression or activity of an endoribonuclease capable of acting as a TRIM25 cofactor.
- composition may be administered in combination with an inhibitor of TRIM25 cofactor expression or activity.
- composition may further comprise one or more expression or activity inhibitors selected from the group consisting of N4BP1, KHNYN, and ZAP.
- composition may be treated in combination with one or more expression or activity inhibitors selected from the group consisting of N4BP1, KHNYN, and ZAP.
- N4BP1 NEDD4 Binding Protein 1
- NEDD4 Binding Protein 1 refers to a protein that binds to NEDD4, an E3 ubiquitin ligase, and is a multifunctional regulatory protein that mainly functions in protein degradation regulation, immune signaling, RNA metabolism, etc.
- KHNYN KH And NYN Domain Containing
- ZAP Zinc-finger Antiviral Protein
- the above combination treatment may mean administering the TRIM25 expression or activity inhibitor and the N4BP1, KHNYN and/or ZAP expression or activity inhibitor sequentially, separately, or in any order.
- the RNA may be exogenous RNA.
- Exogenous RNA refers to RNA derived from outside the cell and introduced into the cell through artificial or natural means, and is generally distinguished from RNA synthesized endogenously within the body.
- the exogenous RNA may have various biological origins, such as eukaryotic cells, prokaryotic cells, or viruses, and may also include artificially synthesized RNA.
- the composition may increase the stability or expression of exogenous RNA introduced into a cell by inhibiting the expression or activity of TRIM25 within the cell.
- TRIM25 can induce a cellular immune response to exogenous RNA or induce RNA degradation, and thus the composition may prevent degradation of exogenous RNA and increase the biological efficacy of the RNA by inhibiting the function of TRIM25.
- the exogenous RNA may be an mRNA vaccine.
- An mRNA vaccine comprises messenger RNA encoded to express a specific antigen, and refers to a vaccine platform that induces an immune response by expressing the antigen protein within a host cell.
- the composition can increase the stability and protein expression efficiency of the mRNA vaccine.
- the exogenous RNA may be encapsulated in a lipid nanoparticle (LNP) and delivered into the cell.
- LNP lipid nanoparticle
- the lipid nanoparticle refers to a nanometer-sized particle composed of lipid molecules used as a carrier for nucleic acid or drug delivery.
- the lipid nanoparticle may include one or more lipids selected from the group consisting of ionic lipids, phospholipids, cholesterol, and polyethylene glycol (PEG)-linked lipids.
- the lipid nanoparticles may be composed of a single or multiple types of lipid components, and the components may affect the size, charge, stability, and cell permeability of the LNPs depending on their composition ratio and structure.
- the lipid nanoparticles include cationic ionic lipids to electrostatically bind to RNA, thereby enabling efficient capture and delivery of RNA.
- the RNA may be at least one selected from the group consisting of mRNA, circular RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, ribozyme, and ncRNA.
- the above TRIM25 expression inhibitor may include, but is not limited to, one or more selected from the group consisting of siRNA, shRNA, oligonucleotide, antisense nucleotide, and sgRNA that specifically bind to the TRIM25 gene.
- the above TRIM25 activity inhibitor may include, but is not limited to, one or more selected from the group consisting of antibodies or antigen-binding fragments thereof that specifically bind to TRIM25 protein, interacting proteins, PROTACs, oligopeptides, ligands, nanoparticles, aptamers, avidity multimers, and peptidomimetics.
- the above N4BP1, KHNYN and/or ZAP expression inhibitor may include, but is not limited to, one or more selected from the group consisting of siRNA, shRNA, oligonucleotide, antisense nucleotide and sgRNA that specifically bind to the N4BP1, KHNYN and/or ZAP genes, respectively.
- N4BP1, KHNYN and/or ZAP activity inhibitor may include, but is not limited to, one or more selected from the group consisting of antibodies or antigen-binding fragments thereof, interacting proteins, oligopeptides, ligands, nanoparticles, aptamers, PROTACs, avidity multimers and peptidomimetics that specifically bind to N4BP1, KHNYN and/or ZAP proteins, respectively.
- composition may be administered orally or parenterally during clinical administration.
- it may be administered by one or more of various routes, including oral, intravenous, intramuscular, intraarterial, intramedullary, intramedullary, subcutaneous, intraventricular, transdermal, intradermal, rectally, intravaginally, intraperitoneally, intraocularly, subretinal, intravitreal, topical, mucosal, transnasal, buccal, enteral, intravitreal, intratumoral, sublingual (under the tongue), and intranasal.
- routes including oral, intravenous, intramuscular, intraarterial, intramedullary, intramedullary, subcutaneous, intraventricular, transdermal, intradermal, rectally, intravaginally, intraperitoneally, intraocularly, subretinal, intravitreal, topical, mucosal, transnasal, buccal, enteral, intravitreal, intratumoral, sublingual (under the tongue), and intranasal
- the above composition is not particularly limited in its formulation, and may be formulated and used in the form of oral formulations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., as well as external preparations, suppositories, and sterile injectable solutions, according to conventional methods.
- oral formulations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc.
- external preparations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc.
- external preparations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc.
- diluents or excipients such as fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants commonly used in the art.
- TRIM25 can act as a negative regulator of exogenous RNA within cells.
- the composition of the present invention can prevent cells from recognizing exogenous RNA as foreign RNA and degrading it or inducing an immune response by inhibiting the expression or activity of TRIM25.
- the composition can induce increased stability of exogenous RNA within cells, increased expression of exogenous RNA within cells, or both. Therefore, the composition of the present invention can be useful for utilizing exogenous RNA as a therapeutic agent.
- the composition can increase the stability and/or expression of a target intracellular RNA in vitro, in vivo, or ex vivo.
- Another aspect provides a method for increasing the stability or expression of RNA within a cell, comprising treating the subject with an inhibitor of TRIM25 expression or activity.
- Another aspect provides the use of TRIM25 expression or activity inhibitors to increase the stability or expression of intracellular RNA.
- Another aspect provides a method for increasing the expression or activity of a target RNA in a cell in vitro. Specifically, the method comprises:
- It comprises a step of introducing target RNA into a cell.
- the method may further comprise a step of inhibiting the expression or activity of one or more selected from the group consisting of N4BP1, KHNYN, and ZAP.
- the step of inhibiting the expression or activity of N4BP1, KHNYN and/or ZAP may be performed sequentially, individually, or in any order with the step of inhibiting the expression or activity of TRIM25.
- increasing the expression or activity of the target RNA may increase the production of the target RNA or a polypeptide encoded by the target RNA.
- the target RNA may be an exogenous RNA.
- the target RNA may be encapsulated in a lipid nanoparticle (LNP) and delivered into the cell.
- LNP lipid nanoparticle
- the RNA may be at least one selected from the group consisting of mRNA, circular RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, ribozyme, and ncRNA.
- Another aspect provides a method for screening a substance for increasing the stability or expression of RNA within a cell. Specifically, the method
- It includes a step of selecting a candidate substance that changes the intracellular expression or activity of TRIM25 compared to an untreated control group.
- the selection step may further include a step of determining the candidate substance as a substance for increasing the stability or expression of intracellular RNA when the intracellular expression or activity of TRIM25 is reduced compared to an untreated control group.
- the measurement of the intracellular expression of TRIM25 may be performed by labeling with a label that generates a detectable signal (the label is, for example, chemically (e.g., covalently or non-covalently), recombinantly, or physically bound) or by labeling in a form in which a tag to which the label can be bound is attached, and then measuring a signal generated from the label through a conventional enzymatic reaction, fluorescence, luminescence, and/or radiological detection.
- the measurement of the signal may be measured by any signal detection means conventionally used to detect or measure it (e.g., a conventional fluorescence microscope, a fluorescence camera, a fluorescence intensity measurement (quantitation) device, etc.).
- Another aspect is,
- RNA having a candidate chemical modification to experimental cells and normal cells in which TRIM25 expression or activity is suppressed
- a method for screening a chemical modification of a nucleic acid that enhances RNA expression or activity comprising a step of determining that the chemical modification enhances RNA expression or activity when the expression level of RNA of an experimental cell or the activity level of an expressed protein is equivalent to the expression level of RNA of a normal cell or the activity level of an expressed protein.
- the above RNA may be an exogenous RNA encapsulated in a lipid nanoparticle (LNP) and delivered into the cell.
- LNP lipid nanoparticle
- nucleic acid in this specification means that the chemical structure of a nucleic acid (DNA or RNA) such as a base, sugar, or phosphate has been modified, and includes both natural chemical modification and artificial chemical modification.
- the exogenous RNA exhibits substantially equivalent expression or activity levels in experimental cells in which TRIM25 expression or activity is suppressed and in normal cells, the exogenous RNA can be considered to contain a chemical modification capable of evading TRIM25. Accordingly, the exogenous RNA can be considered to contain a chemical modification that enhances RNA expression or activity.
- Another aspect is,
- a method for screening a chemical modification of a nucleic acid that enhances expression or activity of RNA comprising a step of determining that the candidate chemical modification is a chemical modification that enhances RNA expression or activity if TRIM25 does not bind or interact with the RNA having the candidate chemical modification.
- the RNA may be an exogenous RNA encapsulated in a lipid nanoparticle (LNP) and delivered into cells.
- LNP lipid nanoparticle
- TRIM25 can act as a negative regulator of exogenous RNA within cells. If RNA with a candidate chemical modification does not bind or interact with TRIM25, the exogenous RNA can be considered to contain a chemical modification that evades TRIM25. If the RNA containing the candidate chemical modification evades TRIM25, the RNA can be prevented from being degraded by the cell or from inducing an immune response. Therefore, in such cases, the exogenous RNA can be considered to contain a chemical modification that enhances RNA expression or activity.
- the measurement of the binding or interaction may be performed at a pH of 7.0 or lower.
- TRIM25 acts as a key negative regulator in LNP RNA delivery. Inhibiting TRIM25 expression or activity can significantly increase gene delivery efficiency and is useful for designing and developing therapeutic RNA vaccines.
- Figure 1 is a schematic diagram of whole-genome CRISPR-Cas9 knockout screening.
- Figure 2 is a schematic diagram of mRNA formulated in LNP (LNP-mRNAs).
- Figure 3 shows the results of evaluating GFP expression in EGFP LNP-mRNA treated cells with or without m1 ⁇ using flow cytometry and signal quantification analysis.
- Figure 4 shows the results of a dot blot analysis showing the IVT mRNA gene expression level after PKR knockdown and the dsRNA byproduct according to its quantification.
- Figure 5 is a graph showing the results of Western blot analysis confirming phosphorylation of PKR and eIF2 ⁇ , polysome profiling results, IFNB1 mRNA levels, and representative ISG expression levels.
- Figure 6 is a diagram analyzing the distribution of sgRNAs in the GFPLow and GFPHigh groups.
- A is the read count distribution of individual sgRNAs
- B is a scatter plot showing the log2 enrichment of all sgRNAs within the CRISPR library
- C is a scatter plot based on FDR rank distinguishing the GFPLow and GFPHigh groups
- D is a heatmap showing the Pearson correlation coefficient for the gene-level log2 enrichment of significant genes (FDR ⁇ 0.05) across different conditions.
- Figure 7 shows the results of the CRISPR-KO screen and the major GO term enrichment results of significant genes derived from GFPHigh and GFPLow conditions.
- Figure 8 shows the results of STRING analysis showing the protein-protein interaction network of positive regulators identified through the screen and the GO term enrichment results of the top 10 candidates.
- Figure 9 is a heatmap showing the log2 enrichment of genes involved in HSPG biosynthesis and V-ATPase.
- Figure 10 shows (top) an observation of a decrease in protein production after knocking down a HSPG pathway-related gene or a V-ATPase subunit gene, and (bottom) the results of measuring the knockdown efficiency using RT-qPCR.
- Figure 11 is a graph showing luciferase expression in HCT116 cells deficient in HSPG biosynthesis factors or V-ATPase subunits.
- Figure 12 shows the results of observing a decrease in protein production using HEK293T and HeLa cells (top) and measuring the knockdown efficiency using RT-qPCR (bottom).
- Figure 13 is a diagram showing the amount of protein produced from EGFP mRNA transfected into HCT116 cells after heparin treatment.
- Figure 14 shows the amount of protein produced from EGFP mRNA transfected into HCT116 cells after treatment with BafA1, Pitstop-2, or Dynole.
- Figure 15 is a graph showing the results of RT-qPCR analysis of intracellular EGFP IVT mRNA levels after heparin or BafA1 treatment.
- Figure 16 is a graph showing the poly(A) length distribution measured by Hire-PAT analysis after heparin or BafA1 treatment.
- Figure 17 is a diagram confirming the luciferase expression level of Fluc mRNA delivered to HCT116 cells via LNP 2.
- Figure 18 shows the FDR ranking-based gene scatter plot (left) and the fold enrichment of sgRNAs targeting TRIM25 (right) under GFPHigh conditions.
- Figure 19 is a graph confirming protein production from unmodified EGFP LNP-mRNA when TRIM25 or WDR77 was knocked down in HCT116 cells.
- Figure 20 shows the results of Western blot analysis of TRIM25 in HCT116 WT or TRIM25 KO-1, KO-2 cells.
- Figure 21 is a graph confirming luciferase expression and GFP expression from Fluc LNP-mRNA in HCT116 WT or TRIM25 KO-1, KO-2 cells.
- Figure 22 is a graph showing relative luciferase expression for m1 ⁇ and U Fluc LNP-mRNA in HCT116 cell lines.
- Figure 23 is a graph showing the measurement of luciferase expression by Fluc LNP-mRNA in primary mouse BMDM cells and various human cell lines (HeLa, THP-1, Jurkat, HEK293T) (top) and representative ISG expression levels (bottom).
- Figure 24 is a graph measuring luciferase expression from LNP-mRNA (top) and Fluc mRNA-2 or Rluc mRNA (bottom) with the 5′ end in an uncapped, cap0, or cap1 state.
- Figure 25 is a graph showing the distribution of Fluc LNP-mRNA extracted by collecting the polysome profiling and ribosome fractions after transfecting HCT116 WT or TRIM25 KO cells with Fluc LNP-mRNA, measured by RT-qPCR.
- Figure 26 is a graph measuring the intracellular abundance of Fluc LNP-mRNA by RT-qPCR.
- Figure 27 is a graph outlining the pulse-chase experiment after Fluc LNP-mRNA transfection and measuring protein expression levels by luciferase analysis, mRNA abundance by RT-qPCR, and poly(A) length distribution by Hire-PAT analysis.
- Figure 28 is a graph measuring the abundance of LNP-mRNA by RT-qPCR.
- EGFP mRNA in TRIM25-deficient HCT116 GFP-stable cells or parental cell line Fluc mRNA in HCT116 TRIM25 KO cells, and Fluc mRNA in GFP-stable cells.
- Figure 29 is a schematic diagram of Fluc circular RNA and the automated electrophoresis results showing its integrity at each experimental step.
- Figure 30 shows the results of measuring the mRNA level of ISG, luciferase expression, and its intracellular abundance by qRT-PCR after transfecting HCT116 WT or TRIM25 KO cells with Fluc circular RNA using LNP.
- Figure 31 shows the results of RT-qPCR measurement of LNP-mRNA-derived luciferase expression, mRNA abundance, and knockdown efficiency of each gene under conditions where exonuclease (XRN1) and exosome components (DIS3, RRP41) were knocked down in HCT116 WT cells.
- exonuclease XRN1
- exosome components DIS3, RRP41
- Figure 32 shows the results of measuring the expression of LNP-mRNA-derived luciferase, mRNA abundance, and knockdown efficiency of each gene by RT-qPCR under conditions in which endoribonuclease (RNASEL) involved in cytoplasmic innate immune response or RNASET2/RNASE4 and PLD3 involved in RNA degradation within endosomes were knocked down in HCT116 WT cells.
- RNASEL endoribonuclease
- Figure 33 shows gene-level enrichment (left) and domain structures (right) of human TRIM25, N4BP1, KHNYN, and ZAP proteins in a CRISPR KO screen using unmodified IVT mRNA.
- Figure 34 is a graph measuring luciferase expression from linear Fluc LNP-mRNA along with KHNYN/ZAP knockdown after N4BP1 siRNA treatment in HCT116 WT or TRIM25 KO cells (top), the results of measuring the efficiency of N4BP1, KHNYN, and ZAP knockdown by RT-qPCR (middle), and the results of measuring the abundance of LNP-mRNA (bottom).
- Figure 35 is a graph showing the results of western blot confirming the knockout of N4BP1 in HCT116 WT or TRIM25 KO cells and measuring luciferase expression from linear Fluc LNP-mRNA along with lentiviral KHNYN/ZAP knockdown.
- Figure 36 is a graph measuring luciferase expression from circular RNA along with KHNYN/ZAP knockdown after N4BP1 siRNA treatment or gene deletion by lentiviral sgRNA in HCT116 WT or TRIM25 KO cells.
- Figure 37 is a graph showing luciferase expression after transfection of Fluc LNP-mRNA with various base modifications under TRIM25 knockdown conditions.
- Figure 38 is a schematic diagram of a TRIM25 RNA immunoprecipitation (RNA-IP) experiment and a graph showing the relative enrichment compared to input as measured by RT-qPCR.
- RNA-IP RNA immunoprecipitation
- Figure 39 shows an overview of RNA pulldown experiments performed by immobilizing poly(A)+ UTR-2 RNA bait containing m1 ⁇ or U onto oligo-dT beads and the results of experiments performed on endogenous or exogenous TRIM25 protein.
- Figure 40 shows the results of in vitro RNA binding experiments, SDS-PAGE analysis, and size exclusion chromatography using purified recombinant human TRIM25 protein and poly(A)+ UTR-2 RNA bait.
- Figure 41 shows the results of RNA pulldown and western blot experiments using UTR-1 RNA as bait.
- Figure 42 is a graph showing the results of a TRIM25 restoration experiment and a western blot analysis of an exogenously expressed TRIM25 variant.
- Figure 43 is a schematic diagram of an in vitro ubiquitination experiment using immunoprecipitated TRIM25 as an E3 ligase and the results of an in vitro ubiquitination experiment comparing TRIM25 WT, 7KA RNA binding mutant, and R54P E3 mutant using Fluc IVT mRNA.
- Figure 44 is a schematic diagram of Fluc LNP-mRNA transfection and TRIM25 knockdown in GFP stable expressing cells, and a graph showing GFP (intracellular mRNA) and luciferase (LNP-mRNA) expression measured by flow cytometry and luciferase analysis, respectively.
- GFP intracellular mRNA
- LNP-mRNA luciferase
- Figures 45, 46, and 47 show the results of confirming luciferase expression, GFP expression, or intracellular abundance after transfection of Fluc mRNA into HCT116 TRIM25 WT or KO-1 cells.
- Figure 45 shows Fluc mRNA transfection via Lipofectamine, Figure 46 via electroporation, and Figure 47 via LNP-2.
- Figure 48 is a graph showing RNA quality and binding of 30-nt poly(A)+ IVT RNA bait immobilized on oligo-dT beads at a specified pH.
- Figure 49 shows the RNA pulldown experiment performed at the indicated pH using HCT116 WT cell extracts with UTR-1 or UTR-2 as bait and the corresponding western blot results.
- Figure 50 shows the results of an RNA pulldown experiment performed using CALM1 UTR as bait and an in vitro RNA binding experiment using purified TRIM25 protein.
- Figure 51 shows the results of an in vitro RNA binding experiment performed in a potassium phosphate buffer (pH-wise) adjusted at pH intervals of 0.2 using purified TRIM25 protein.
- Figure 52 is a proposed model of the intracellular regulatory mechanism for LNP-mRNA.
- Example 1 CRISPR-Cas9 screening to identify regulatory factors of LNP-mRNA.
- sgRNA libraries were introduced into HCT116, a human epithelial cell line characterized by a stable diploid karyotype, a rapid and organized endocytosis pathway capable of efficient LNP delivery, and a well-characterized interferon (IFN) response to exogenous nucleic acids.
- IFN interferon
- the introduced mRNA contained a 5′ cap1 structure, a 120-nt poly(A) tail, a synthetic 5′ UTR, and a human alpha-globin-derived 3′ UTR, which are similar to the UTR used in the mRNA-1273 COVID-19 vaccine (Fig. 2). These mRNAs were produced in two forms, one with and one without the m1 ⁇ modification, to enable analysis of the regulatory mechanism dependent on base modification.
- mRNA containing m1 ⁇ produced more protein than unmodified mRNA (Fig. 3).
- the IVT mRNA was encapsulated in LNPs composed of the same four lipid components (ALC-0315, DSPC, cholesterol, and ALC-0159) used in the BNT162b2 COVID-19 vaccine.
- LNP formulation and transfection conditions were optimized to minimize cell-to-cell variation in transfection efficiency, enabling GFP expression-based screening. Since we aimed to identify single-stranded mRNA-specific regulatory factors, we induced dsRNA responses through PKR, OAS, TLR3, RIG-I, and MDA5 to reduce dsRNA byproducts derived from the IVT reaction that could nonspecifically interfere with gene regulation.
- the IVT DNA template was constructed by PCR using a reverse primer with a 2′-O-methyl group introduced at the terminal and terminal positions to ensure accurate transcription termination.
- PKR knockdown affected gene expression from IVT mRNA only when large amounts of dsRNA were co-transfected.
- Fluc IVT mRNA was encapsulated in LNPs and transfected in the same manner as in the previous experiments.
- Fluc mRNA was transcribed from template DNA (PCR product) lacking a terminal 2'-O-methyl group, resulting in increased production of dsRNA byproducts (Fig. 4).
- the generated RNA did not induce IFN responses, PKR phosphorylation, or overall translational inhibition (Fig. 5).
- EGFP mRNA produced in this cell line is identical to IVT EGFP mRNA, except for additional sequences derived from the polyadenylation signal.
- Example 1 Screening using IVT mRNA in Example 1 identified several positive regulatory factor candidates, including genes involved in heparan sulfate proteoglycan (HSPG) biosynthesis, vesicle transport, and V-ATPase components (Fig. 8). These genes were commonly enriched in both unmodified and m1 ⁇ -modified mRNA screens, suggesting that their roles are independent of RNA modification.
- HSPG heparan sulfate proteoglycan
- V-ATPase an ATP-driven proton pump that acidifies the lumen of the endosomes.
- V-ATPase subunits decreased protein production from IVT mRNA (EGFP and Fluc) in HCT116, HEK293T, and HeLa cells (Figs. 10 and 11). Furthermore, treatment with Bafilomycin A1 (BafA1), which inhibits V-ATPase, also reduced GFP production from IVT mRNA. Furthermore, when GFP-stable cells were transfected with Fluc mRNA and treated with BafA1, BafA1 selectively inhibited exogenous mRNA, suppressing only luciferase expression without affecting GFP. Furthermore, Pitstop-2 and Dynole, which are endocytic inhibitors targeting clathrin and dynamin I/II, respectively, also suppressed LNP-mRNA expression (Fig. 13).
- RNA analysis revealed that heparin reduced the intracellular abundance of IVT mRNA, whereas BafA1 increased mRNA levels (Fig. 15). This suggests that heparin inhibits initial uptake by interfering with LNP attachment to the cell surface, whereas BafA1 inhibits endosomal escape, leading to mRNA accumulation within endosomes.
- Hire-PAT high-resolution poly(A) tail analysis
- LNP-2 another type of LNP consisting of the ionic lipid SM-102 used in the mRNA-1273 COVID-19 vaccine and the PEGylated lipid DMG-PEG 2000.
- TRIM25 was the most strongly enriched with the lowest FDR value in the unmodified mRNA screening, but was not enriched in the GFP-stable cells or m1 ⁇ -modified mRNA screening (Fig. 18). This suggests that TRIM25 selectively represses unmodified IVT mRNA.
- TRIM25 is an RNA-binding protein and E3 ligase known to have antiviral activity against RNA viruses.
- TRIM25 is also known to interact with immune sensors such as RIG-I and ZAP, as well as the G3BP protein.
- TRIM25 and WDR77 which are candidates existing in the cytoplasm, were knocked down.
- TRIM25 knockout (KO) cell lines (TRIM25 KO-1 and KO-2) were generated using different sgRNAs (Fig. 20).
- unmodified mRNA produced more protein than the parental cell line (Fig. 21).
- unmodified mRNA produced luciferase at levels comparable to those of m1 ⁇ -modified mRNA. This demonstrates that TRIM25 is a key repressor of unmodified mRNA (Fig. 22).
- TRIM25 human embryonic kidney cells
- HeLa human epithelial cells
- THP-1 human monocytes
- Jurkat human T lymphocytes
- HEK293T human embryonic kidney cells
- mBMDM bone marrow-derived macrophages
- TRIM25 depletion significantly increased the expression of intact mRNA in HeLa, THP-1, Jurkat, and mBMDM cells, but not in HEK293T cells.
- the surveillance mechanism mediated by TRIM25 shows some differences depending on the cell type, it was confirmed that this mechanism is universal and evolutionarily conserved (Fig. 23).
- mRNAs with different cap structures were compared.
- mRNAs with cap0 (N7-methylguanosine, m7G) and cap1 (m7G plus 2′-O-methylation at the +1 position) exhibited different capping efficiencies, allowing us to analyze the impact of the 5′-terminal structure.
- TRIM25 depletion significantly increased luciferase expression regardless of the cap structure, indicating that TRIM25's action is independent of the 5′-terminal structure.
- Polysome profiling is a method for separating mRNAs undergoing translation based on the number of ribosomes bound on a sugar gradient.
- TRIM25 deletion or nucleotide modification did not significantly affect overall polysome distribution, indicating that TRIM25 does not affect global translation.
- TRIM25 deletion or nucleotide modification did not significantly affect overall polysome distribution, indicating that TRIM25 does not affect global translation.
- Fig. 25 when the amount of IVT mRNA in each gradient fraction was quantified using RT-qPCR, no significant changes were observed due to TRIM25 deletion (Fig. 25). Thus, while the possibility that TRIM25 regulates translation cannot be completely ruled out, this is unlikely to be the primary mechanism of TRIM25 action.
- RNA amount was analyzed using RT-qPCR.
- TRIM25-deficient cells showed a dramatic increase in the level of unmodified IVT mRNA.
- the level of modified mRNA increased only slightly.
- no significant increase was observed when treated with BafA1, which inhibits endosomal escape. This indicates that TRIM25 primarily acts by reducing mRNA levels after mRNA enters the cytoplasm (Fig. 26).
- TRIM25 activates a very rapid repression mechanism (Fig. 27). Increases in both EGFP and Fluc mRNA levels were also observed in TRIM25 knockdown or deficient cells. These results suggest that TRIM25 promotes mRNA degradation (Fig. 28).
- deadenylation is generally considered a rate-determining step in mRNA degradation
- TRIM25 deletion did not significantly affect the deadenylation rate, indicating that TRIM25 does not act to accelerate deadenylation (Fig. 27).
- TRIM25 induces endonucleolytic cleavage
- circRNA circular RNA lacking a 5′ cap and poly(A) tail
- Fig. 29 we constructed a circRNA without the m1 ⁇ modification.
- circRNAs did not significantly induce the expression of IFN ⁇ and ISGs.
- circRNA-derived luciferase expression was significantly increased. RNA levels were also higher in TRIM25-deficient cells than in the control.
- circRNAs did not accumulate in cells treated with BafA1 (Fig. 30).
- N4BP1 NEDD4-binding protein 1
- N4BP1 contains a NYN domain (endoribonuclease domain), a KH-like domain (putative RNA-binding domain), and ubiquitin-binding domains, UBA and CoCUN.
- N4BP1 has a potential paralog, KHNYN, which ranked 4027th in the screening of unmodified mRNA. KHNYN interacts with TRIM25 and ZAP and is known to inhibit HIV-1 replication.
- N4BP1 knockout cells were generated. Single deletion of N4BP1 resulted in a modest but significant increase in luciferase expression, and the inhibitory effect was further alleviated when KHNYN and ZAP were additionally knocked down with siRNA. In contrast, no effect was observed in TRIM25-deficient cells (Fig. 35). This regulatory effect was much stronger with unmodified mRNA than with modified mRNA.
- RNAs were also derepressed in a TRIM25-dependent manner in N4BP1/KHNYN/ZAP-depleted cells (Fig. 36).
- Fig. 36 circular RNAs
- ⁇ variant mRNA showed only a minimal increase in expression upon TRIM25 knockdown. This suggests that both m1 ⁇ and ⁇ variant mRNAs evade TRIM25 to some extent, but not completely. In contrast, mo5U and m5C variant mRNAs showed a moderate increase in expression. This suggests that TRIM25 can still target these mRNAs, and that mo5U and m5C variants provide only partial protection from TRIM25.
- TRIM25 is known to interact directly with RNA, and TRIM25 mutants defective in RNA binding lack antiviral activity. Therefore, we hypothesized that TRIM25 might differentially recognize unmodified mRNA and regulate it through its interaction with RNA. To verify this, we transfected HCT116 cells with LNP329 mRNA, followed by RNA immunoprecipitation and RT-qPCR (RIP-qPCR) (Figure 38).
- TRIM25 appears to preferentially interact with exogenous RNA without base modifications.
- RNA-binding specificity of TRIM25 was further demonstrated through pull-down experiments centered on RNA336 (Fig. 39).
- Single-stranded RNA was constructed by adding a 30-nt poly(A) fragment to the 100-nt sequence at the 3′ UTR of UTR-1 or UTR-2 shown in Fig. 24. This was immobilized on oligo-dT beads and reacted with HCT116 cell lysate.
- RNA To determine whether TRIM25 directly interacts with RNA, we purified recombinant TRIM25 protein. As a result, unmodified RNA effectively pulled down TRIM25 protein ( Figure 40), indicating a direct interaction between TRIM25 and RNA. In contrast, m1 ⁇ -modified RNA did not significantly precipitate TRIM25, indicating that the m1 ⁇ modification inhibits TRIM25's interaction with RNA.
- TRIM25 is composed of a RING domain, a B-box, a coiled-coil domain, a positively charged linker, and a PRY/SPRY domain.
- a seven-lysine sequence (381KKVSKEEKKSKK392, the "7K motif" within the linker is essential for RNA binding.
- a mutant in which the lysine residues in this 7K motif are substituted with alanines (“7KA”) is known to reduce the affinity of TRIM25 for RNA.
- the 7KA mutant showed little pull-down with RNA bait, implying the importance of the 7K motif in the RNA-TRIM25 interaction ( Figure 41).
- TRIM25 functions as an E3 ubiquitin ligase.
- a mutation R54P was introduced at an arginine residue within the RING domain. This mutation is known to impair interaction with the E2 ubiquitin ligase, resulting in loss of E3 ligase activity.
- the R54P mutant failed to repress the expression of unmodified IVT mRNA ( Figure 42), indicating that E3 ligase activity contributes to TRIM25-mediated repression.
- RNA binding enhances the E3 ligase activity of TRIM25 we performed an in vitro ubiquitination experiment. To this end, FLAG-tagged TRIM25, which is expected to co-precipitate with substrate proteins, was immunoprecipitated. E1, E2, and ubiquitin were added to the immunoprecipitate and reacted in the presence or absence of IVT mRNA. The addition of IVT mRNA increased the level of ubiquitinated proteins, indicating that RNA binding induces the ubiquitination ligase activity of TRIM25 ( Figure 43). Notably, unmodified mRNA exhibited a stronger effect than m1 ⁇ -modified mRNA.
- RNA-binding mutant (7KA) showed reduced ubiquitination levels, and a mutant lacking E3 ligase activity (R54P) served as a negative control.
- R54P E3 ligase activity
- TRIM25 was not detected in a control screening using GFP-stabilized cells. Knockdown experiments performed in GFP392 stably expressed cells confirmed that TRIM25 can distinguish between exogenous mRNA and endogenous mRNA exported from the nucleus (Fig. 44).
- RNA entry pathway into the cytoplasm we investigated whether TRIM25's RNA targeting is influenced by the RNA entry pathway into the cytoplasm.
- Lipofectamine a cationic lipid-based transfection reagent. Lipofectamine, which maintains a persistent positive charge at physiological pH, is known to transport transfection agents across the plasma membrane and early endosomes.
- RNA was introduced into the cytoplasm using electroporation. Electroporation is a method that creates temporary pores in the plasma membrane to allow mRNA to enter the cell. When electroporation was used, deletion or knockdown of TRIM25 did not significantly affect reporter mRNA, indicating that TRIM25 does not inhibit mRNA delivered directly through the plasma membrane ( Figure 46). In contrast, when another type of LNP consisting of SM-102 (ionized lipid), DMG-PEG 2000, DSPC, and cholesterol was used, as in mRNA-1273, TRIM25 deficiency strongly affected Fluc expression (Figure 47). These results indicate that TRIM25 can selectively target exogenous mRNA delivered via LNPs.
- RNA-binding activity of TRIM25 is affected by pH changes. Specifically, IVT RNA was immobilized on oligo-dT beads, and RNA-binding experiments were performed using cell lysates at pH 6.5-8.0.
- TRIM25 strongly bound to UTR-1 RNA under slightly acidic conditions (pH 6.5), and the binding strength showed a sharp change between pH 6.5 and 7.0. Similar pH sensitivity was also observed in similar RNA pull-down experiments using another RNA (UTR-2) (Figs. 48 and 49).
- RNA-binding properties of TRIM25 were further confirmed by performing RNA-binding experiments in two different buffer conditions after immobilizing biochemically synthesized RNA via biotin-avidin conjugation. Unlike TRIM25, other RNA-binding proteins, such as hnRNPA1 and G3BP1, were hardly affected by pH changes. RNA-binding experiments using purified recombinant TRIM25 protein showed that TRIM25's direct interaction with RNA increased under acidic conditions (Figure 50).
- RNA-binding affinity of TRIM25 significantly increased between pH 7.2 and pH 7.0, which are physiological cytoplasmic pHs, and further increased at lower pHs. This suggests that even subtle pH changes within cells can induce TRIM25-RNA interactions (Fig. 51).
- RNAs with m1 ⁇ introduced showed lower binding affinity to TRIM25 than unmodified RNA under all experimental conditions. This indicates that the m1 ⁇ modification inhibits TRIM25 binding even under acidic conditions.
- a sequence containing a single SNP (dbSNP rs205498, a G/A mutation corresponding to the P358L point mutation, found in more than 77% of purified alleles) in the reference coding sequence of TRIM25 (RefSeq NM_005082) was PCR amplified and inserted into the pCK vector with a FLAG tag at the N terminus to construct a plasmid for exogenous expression.
- the same sequence was subcloned into the pX vector with a His10-eYFP-SUMOstar-Strep tag at the N terminus.
- mutants of TRIM25 such as the 7KA RNA-binding defective mutant and the R54P E3 ligase defective mutant, were constructed by site-directed mutagenesis.
- the CMV promoter was replaced with the PGK promoter, and the FLAG tag was removed to ensure stable expression.
- the list of plasmids used in this study is summarized in Table 1.
- HCT116 wild-type parental cell line and cell lines derived from it were cultured in McCoy's 5A medium (Welgene, LM 005-01) supplemented with 9.1% fetal bovine serum (FBS) (Welgene, S001-01).
- FBS fetal bovine serum
- HEK293T, lenti-X HEK293T, HeLa, and MEF cells were cultured in DMEM (Welgene, LM 001-05) containing 9.1% FBS, and suspended HEK293E cells were cultured in DMEM (Welgene, LM001-170) containing 5% FBS and 50 ⁇ g/mL G418 sulfate (Gibco, 10131-027).
- THP-1 cells were cultured in RPMI 1640 (Welgene, LM 011-03) supplemented with 9.1% FBS and 0.05 mM ⁇ -mercaptoethanol (Thermo Scientific, 35602BID), and Jurkat cells were cultured in the same RPMI 1640 medium supplemented with 9.1% FBS. All cell lines were confirmed to be mycoplasma-negative.
- mBMDMs Mouse-derived bone marrow macrophages
- mBMDMs mouse-derived bone marrow macrophages
- mBMDMs mouse-derived bone marrow macrophages
- L929 conditioned medium was prepared using L929 cells (ATCC, CCL-1) according to the Bowdish Lab (McMaster University) protocol and cultured in IMDM medium (Thermo Scientific, 12440053) supplemented with 10% FBS and 1% penicillin/streptomycin. On days 3 and 5, 10 mL of fresh medium containing M-CSF was added, and on day 6, cells were gently scraped and harvested. In all subsequent experiments, antibiotic-free medium was used.
- a plasmid containing the EGFP coding sequence including the minimal CMV promoter, the 5′ and 3′ untranslated regions (UTRs) of the in vitro-transcribed RNA used for CRISPR screening, and the downstream SV40 poly(A) signal was constructed using a lentiviral backbone vector (Addgene #60954).
- Lentivirus was produced by cotransfecting lenti-X HEK293T cells with the plasmids containing the lentiviral packaging plasmid, and the produced virus was filtered through a 0.45 ⁇ m filter (Millipore, SLHVR33RS).
- the harvested lentivirus was then used to infect the HCT116 parental cell line together with 8 ⁇ g/mL polybrene (Millipore, TR-1003-G). GFP-positive cells were selected using flow cytometry (FACS) and isolated as single cells. After expansion of the clones, the single clone with the most uniform GFP fluorescence was used in subsequent experiments.
- the HCT116 TRIM25 knockout cell line was constructed by subcloning sgRNAs targeting the canonical TRIM25 mRNA isoform (RefSeq NM_005082.4) into a Cas9 expression backbone plasmid (Addgene #48148).
- sgRNA targeting exon 3 sgTRIM25-1, used in the TRIM25 KO-1 clone in this study; GCAGCTACAACAAGAATACA, SEQ ID NO: 165
- sgRNA targeting exon 7 sgTRIM25-2, used in the TRIM25 KO-2 clone; TGTTCCGGGGCTCCAAACGT, SEQ ID NO: 166
- HCT116 wild-type parental cells were cotransfected with plasmids expressing Cas9 and sgRNA, and single cells were isolated by limiting dilution. TRIM25 deletion was confirmed by Sanger sequencing and Western blotting.
- the HCT116 pooled N4BP1 knockout cell line was generated via lentiviral infection.
- Lentiviral vectors expressing Cas9 and a non-targeting control sgRNA or an N4BP1-targeting sgRNA were cloned (Addgene #52961).
- sgRNA sequences used were as follows: sgCtrl-1 (CTATATTGTCGCGCAGTGGA, SEQ ID NO: 1), sgCtrl-2 (CTCCCTGCCGGCCGGGTTAG, SEQ ID NO: 2), sgN4BP1-1 (AGAAAGAGAATGTTACCCCA, SEQ ID NO: 3), and sgN4BP1-2 (TTACACAGAATGCTGCCACA, SEQ ID NO: 4). These sgRNAs were selected from the Brunello human CRISPR knockout pooled library (Addgene #73179).
- CRISPR lentiviruses were generated and harvested by transfecting lentiviral packaging plasmids (Addgene #12259, pMD2.G and #12260, psPAX2) and each CRISPR lentiviral backbone plasmid into lenti-X HEK293T cells.
- lentiviral packaging plasmids Additional plasmids
- Each generated lentivirus was used to infect HCT116 wild-type or TRIM25-deficient cells for 1 day under conditions containing 8 ⁇ g/mL polybrene, and the medium was replaced with fresh medium the following day.
- Infected cells were selected for 5 days under 1 ⁇ g/mL puromycin, and N4BP1 deletion was verified by Western blotting.
- CDSs coding sequences of the reporter genes used in this study (Enhanced green fluorescent protein, EGFP; Firefly luciferase, Fluc; Renilla luciferase, Rluc) were subcloned into mRNA production plasmids flanked by 5′ and 3′ untranslated regions (UTRs).
- the standard EGFP sequence was used as the open reading frame including the initiation codon, and for Fluc/Rluc, the sequence derived from the pmirGLO-3XmiR-1 dual luciferase plasmid (backbone plasmid from Promega) used in a previous study was used.
- the UTR composition followed the combination reported in a previous study by Moderna [Cell 168, 1114-1125.e10] in CRISPR screening and validation experiments.
- the 5′ UTR is the original 41-nucleotide synthetic sequence of Moderna COVID-19 mRNA vaccine (mRNA-1273) (GenBank: OR134578.1), which includes the Kozak sequence, and the 3′ UTR uses a sequence derived from human alpha-globin mRNA.
- mRNA-1273 GenBank: OR134578.1
- the 3′ UTR uses a sequence derived from human alpha-globin mRNA.
- Fluc mRNA-2 and Rluc mRNA used in Figure 24 the UTR configuration was changed based on the sequence predicted to be expressed in the pmirGLO-3XmiR-1 dual luciferase vector.
- the IVT template was generated by PCR amplification using a forward primer containing the T7 promoter sequence and a reverse primer containing a poly(T) sequence. These primers were designed to target the 5′ and 3′ UTRs of interest, respectively, and the 5′-terminal two nucleotides of the reverse primer were 2′-O-methylated to increase the accuracy of transcription termination and prevent byproduct formation.
- a reverse primer without 2′-O-methylation was used.
- the PCR-amplified IVT DNA template was subjected to agarose gel electrophoresis, gel-extracted (Qiagen, 28704), and further oligo-purified (Zymo Research, D4061) to synthesize IVT mRNA using the mMESSAGE mMACHINE T7 Transcription Kit (Invitrogen, AM1344).
- mRNA capping and polyadenylation were performed concurrently with transcription.
- Representative cap1 analogs (CleanCap AG (3′-OMe), TriLink, N-7413) were used for capping.
- Experiments comparing differences in cap structure (Fig. 24) used first-generation cap0 analogs (mCAP, TriLink, N-7001) and later-developed cap0 analogs (ARCA, TriLink, N-7003).
- mCAP first-generation cap0 analogs
- ARCA TriLink, N-7003
- a modified design was applied using a forward primer starting with A instead of G at the +1 position in the PCR for IVT templates, and the conditions were applied to the mRNA synthesis used in the following figures: Fig. 4, Fig. 17, Fig. 23, Fig. 24, Fig. 26, Fig. 27, Fig.
- the poly(A) tail of the mRNA was generated by transcribing the poly(T) sequence inserted into the template, and a fixed length of 120 nt was used for most IVT mRNAs, while a length of 60 nt was used for Fluc mRNA in Fig. 4 and Rluc mRNA in Fig. 24.
- each NTP in the IVT reaction was completely replaced with the desired modified NTP.
- the modified NTPs used were as follows:
- TriLink N1-methylpseudoUTP (TriLink, N-1081), pseudoUTP (TriLink, N-1019), 5-methoxyUTP (TriLink, N-1093), 5-methylCTP (TriLink, N-1014).
- RNA cleanup After transcription, the DNA template was removed with RNase-free DNase (Takara, 2270A), and the generated mRNA was purified according to the RNA purification protocol using Qiagen's RNeasy Mini Kit (74106), with additional on-column DNase treatment (Qiagen, 79254). To minimize NTP residues and maximize RNA purity, the purified RNA was further purified using RNA cleanup.
- IVT templates were generated by PCR from a plasmid containing the Fluc sequence. This template was derived from circRNA-synIRES-R25-NanoLuc (Addgene #188116), and in this study, the NanoLuc CDS was replaced with the Fluc CDS. IVT template generation and circRNA synthesis were performed according to previously reported methods [Nat. Biotechnol. 41, 262-272 and Nat. Commun. 9, 2629] with slight modifications. Briefly, circRNAs were transcribed in vitro using the HiScribeTM T7 High Yield RNA Synthesis Kit (NEB, E2040S), and the DNA template was removed by DNase treatment after transcription.
- HiScribeTM T7 High Yield RNA Synthesis Kit NEB, E2040S
- RNA circularization was promoted by adding 2 mM GTP and incubating at 55°C for 15 min.
- Circular RNA was purified by column chromatography, then treated with RNase R (Abm, E049) at 37°C for 15 min to remove linear RNA, followed by another column chromatography purification. Finally, circRNA was further purified by agarose gel electrophoresis using the Zymoclean Gel RNA Recovery Kit (Zymo Research, R1011).
- N6-methylATP TriLink, N-1013
- ATP TriLink, N-1013
- the integrity of the final IVT RNA was assessed using an automated electrophoresis device (Agilent TapeStation, RNA ScreenTape analysis), and RNA concentration was quantified spectrophotometrically prior to LNP formulation.
- the sequences of the plasmids and PCR primers used for constructing the IVT templates used in this study, as well as the CDS and UTR sequences of the IVT mRNA, are listed in Tables 1 and 2.
- the LNP formulation was based on a previous report [Sci. Adv. 7, eabf4398] with some modifications to suit the mRNA formulation.
- LNPs were dissolved in the organic phase, and the target IVT mRNA was dissolved in the aqueous phase, and mixed in a volume ratio of 1:3.
- the LNP used in this study mimicked the composition of the BNT162b2 COVID-19 mRNA vaccine developed by Pfizer-BioNTech [Nature 595, 572-577 and Int. J. Pharm.
- lipid components 601, 120586] and contained the following four lipid components: ionizable lipid (ALC-0315; Echelon Biosciences, N-1020), phospholipid (DSPC; Avanti, 850365), cholesterol (Sigma, C8667), and PEG-linked lipid (ALC-0159; Echelon Biosciences, N-2010).
- lipids were dissolved in ethanol (Sigma, E7023) at a molar ratio of 46.3:9.4:42.7:1.6 to prepare a lipid master mix.
- LNP-2 LNP-1273 COVID-19 vaccine
- SM-102 MedChemExpress, HY-134541
- DMG-PEG 2000 Advanti, 880151P
- the formulated LNP-mRNA mixture was diluted with PBS, and the encapsulation efficiency was measured using the Quant-iT RiboGreen RNA Assay (Invitrogen, R11491) prior to transfection.
- CRISPR-Cas9 knockout screening For genome-wide CRISPR-Cas9 knockout screening, the Brunello human CRISPR knockout pooled library (Addgene #73179) was used in this study.
- the library contains 77,441 sgRNAs (approximately four independent sgRNAs per gene) targeting 19,114 protein-coding genes, plus 1,000 non-targeting sgRNAs for normalization. Screening was performed in two biological replicates. Plasmids containing Cas9 and the sgRNA library were amplified according to a previously reported method [Cell 186, 3291-3306/e21], and the integrity of the sgRNAs was verified by next-generation sequencing (NGS) before screening.
- CRISPR lentiviruses were produced and harvested from lenti-X HEK293T cells, and the multiplicity of infection (MOI) was calculated by measuring cell viability under various concentrations of puromycin.
- MOI multiplicity of infection
- HCT116 cells were seeded in fresh medium containing 8 ⁇ g/mL polybrene and pooled CRISPR lentivirus at a limited MOI (approximately 0.3) to generate single-particle infected cells with only one gene silenced per cell. After 1 day of infection, the medium was replaced with fresh medium, and on day 2, cells were passaged and selected in medium containing 1 ⁇ g/mL puromycin for 7 days. To ensure at least 300-fold coverage of the CRISPR library, cells were maintained at a density of at least 3 ⁇ 107 cells during the puromycin treatment period.
- GFP-stable cell line screening only CRISPR infection was performed on HCT116 GFP-stable cells, and LNP delivery of EGFP mRNA was omitted.
- genomic DNA from sorted and unsorted control cells was extracted using the MasterPure Complete DNA and RNA Purification Kit (Lucigen, MC85200) according to the manufacturer's protocol. Subsequently, a two-step PCR reaction was performed to prepare a sequencing library using Herculase II Fusion DNA polymerase (Agilent, 600677) in the same manner as previously reported [Nature 542, 197-202 and Science. 370(6523):eabc9546]. For the unsorted control, 6.6 ⁇ g of gDNA was used per 100 ⁇ L reaction, and a total of 24 PCR reactions were performed to ensure 300-fold coverage.
- PCR1_F CRISPR screen 1st PCR 25 PCR1_R CRISPR screen 1st PCR 26 P7-A01 CRISPR screen 2nd PCR barcoded reverse primer 27 P7-A02 CRISPR screen 2nd PCR barcoded reverse primer 28 P7-A03 CRISPR screen 2nd PCR barcoded reverse primer 29 P7-A04 CRISPR screen 2nd PCR barcoded reverse primer 30 P7-A05 CRISPR screen 2nd PCR barcoded reverse primer 31 P7-A06 CRISPR screen 2nd PCR barcoded reverse primer 32 P7-A07 CRISPR screen 2nd PCR barcoded reverse primer 33 P7-A08 CRISPR screen 2nd PCR barcoded reverse primer 34 P7-A09 CRISPR screen 2nd PCR barcoded reverse primer 35 P7-A10 CRISPR screen 2nd PCR barcoded reverse primer 36 P7-A11 CRISPR screen 2nd PCR barcoded reverse primer 37 P7-A12 CRISPR screen 2nd PCR barcode
- sgRNA sequences per read were extracted from demultiplexed FASTQ files and aligned to the sgRNA reference sequence constructed by Addgene based on CRISPR library information using Bowtie 2 under unique alignment and no mismatch tolerance conditions. A total of more than 3 ⁇ 107 sequencing reads were secured by counting the uniquely aligned reads, achieving at least 300x sgRNA coverage.
- RNA transfection using Lipofectamine MessengerMAX was performed by treating cells with IVT mRNA at various concentrations for 1 day using Lipofectamine MessengerMAX (Invitrogen, LMRNA015) according to the manufacturer's instructions.
- RNA pulldown, and in vitro ubiquitination experiments requiring exogenous expression of TRIM25 the TRIM25 expression plasmid was delivered to cells using FuGENE HD transfection reagent (Promega, E2312) according to the manufacturer's instructions.
- siRNA was introduced via electroporation, as described in the Electroporation section below.
- the siRNAs used in this study were either ON-TARGETplus SMARTpool siRNA (Dharmacon) or individually designed siRNAs using Invitrogen Block-iT RNAi Designer (Invitrogen).
- ON-TARGETplus Non-targeting Control Dharmacon
- AccuTarget Negative Control siRNA Bioneer served as negative controls.
- the sequences of the siRNAs used are presented in Table 4.
- siRNA L-003527-00 sihTLR3 Gene specific siRNA L-007745-00 sihTLR7
- siRNA L-004714-00 sihTLR8 Gene specific siRNA L-004715-00 sihDDX58 (RIG-I) Gene specific siRNA L-012511-00 sihIFIH1 (MDA5) Gene specific siRNA L-013041-00 sihOAS1 Gene specific siRNA L-011344-00 sihOASL Gene specific siRNA L-012617-00 sihEXT1-1 Gene specific siRNA 52 sihEXT1-4 Gene specific siRNA 53 sihEXT1-8 Gene specific siRNA 54 sihEXT2-1 Gene specific siRNA 55 sihEXT2-6 Gene specific siRNA 56 sihNDST1-3 Gene specific siRNA 57 sihNDST1-6 Gene specific siRNA 58 sihNDST1-10 Gene specific siRNA 59 sihATP6AP1-2 Gene specific siRNA 60 sihATP6AP1-7
- HCT116 wild-type or GFP-stable cell lines were transfected with LNP-formulated mRNA, followed by treatment with inhibitors of intracellular positive regulator candidates for 1 day.
- HCT116 wild-type cells were treated with six concentrations of heparin (0, 0.1, 0.2, 0.5, 2, and 10 ⁇ g/mL) or Bafilomycin A1 (0, 0.1, 0.2, 0.3, 0.5, and 1 nM).
- Electroporation was performed using the Neon NxT System (Invitrogen, MPK5000) according to the manufacturer's instructions.
- HCT116 cells were trypsinized and washed with DPBS. Cells were then resuspended in 50 ⁇ L of R buffer (Invitrogen, MPK1096) at a concentration of 5 ⁇ 10 6 cells/mL, and 100 ng of Fluc IVT mRNA was added and mixed well.
- Ten ⁇ L of the cell-RNA mixture was used per pulse, and a total of 40 ⁇ L was processed per sample over four pulses.
- Electroporation was performed under the following conditions: 1530 V, 20 ms, 1 pulse/10 ⁇ L. Immediately after electroporation, cells were immediately transferred to 1 mL of pre-warmed McCoy's medium in a 12-well plate.
- TRIM25 knockdown experiment in Jurkat or mBMDM in Figure 23, cells were washed with DPBS and resuspended in T buffer (Invitrogen, MPK10096) at a concentration of 2 ⁇ 10 7 cells/mL (Jurkat) and 1.2 ⁇ 10 7 cells/mL (mBMDM), respectively. mBMDM were collected by gentle scraping before resuspension. siRNA was then added to a final concentration of 1 ⁇ M and mixed well. 100 ⁇ L of the cell-siRNA mixture was used for electroporation under the following conditions: Jurkat: 1350 V, 10 ms, 3 pulses; mBMDM: 1500 V, 20 ms, 1 pulse.
- Jurkat cells were seeded in 1.9 mL of RPMI 1640 medium in 6-well plates, and mBMDM cells were seeded in 2 ⁇ 105 cells in 12-well plates, transferred to 1 mL of L929-conditioned DMEM, and 1 mL of DMEM was added 2 h later. The following day, mRNA was transfected into the siRNA-transduced cells via LNP.
- GFP fluorescence in live cells including GFP-stable cells and wild-type cells transfected with EGFP mRNA, was measured using a BD Accuri C6 Plus Flow Cytometer (BD Biosciences). Similar to FACS separation, cells were trypsinized, resuspended, and filtered through a tube containing a 35 ⁇ m cell strainer cap. The filtered cells were injected into the analyzer, and GFP fluorescence in live cells was measured in the appropriate fluorescence channel (FITC-A).
- FITC-A fluorescence channel
- Luciferase assays were performed by lysing cells with 1X passive lysis buffer and measuring luminescence using the Dual-Luciferase Reporter 1000 Assay System (Promega, E1910) according to the manufacturer's instructions. In LNP-mRNA experiments, mRNA expressing Fluc or Rluc was transfected. Luciferase Assay Reagent II (LAR II) was used for measuring firefly luciferase activity, and LAR II and Stop & Glo Reagent were used together for measuring renilla luciferase activity.
- LAR II Luciferase Assay Reagent II
- PCR amplification was performed using a 6-FAM-labeled universal reverse primer and a gene-specific forward primer.
- the primer sequences used are presented in Table 6.
- NP-40 phosphatase inhibitor cocktail
- AG Scientific P-1518
- 1X passive lysis buffer Promega, E1910
- Protein samples were loaded onto 8–16% Novex Tris-Glycine protein gels (Invitrogen, XP08162BOX) along with Thermo Scientific protein marker (26616), and then transferred to PVDF membranes (Millipore) activated with methanol.
- the transferred membranes were blocked with 5% skim milk or BSA-supplemented PBS-T solution, and immunoblotted with primary antibodies and HRP-conjugated secondary antibodies.
- Chemiluminescence was performed using SuperSignal West Pico or Femto reagents (Thermo Scientific, 34580 or 34095), and signals were detected with a ChemiDoc XRS+ System (Bio-Rad).
- ⁇ -TRIM25 Abcam, ab167154, 1:1000
- ⁇ -Fluc Invitrogen, PA5-32209, 1:1000
- ⁇ -RIG-I Cell Signaling, 3743S, 1:1000
- ⁇ -eIF2 ⁇ Cell Signaling, 5324S, 1:1000
- ⁇ -phospho-eIF2 ⁇ Cell Signaling, 3398S, 1:1000
- ⁇ -PKR Cell Signaling, 12297S, 1:1000
- ⁇ -phospho-PKR Thermo Scientific, MA5-38282, 1:500
- ⁇ -Ubiquitin Abcam, ab7254, 1:2000
- ⁇ -hnRNPA1 Santa Cruz, sc-32301, 1:1000
- ⁇ - ⁇ -Tubulin Abcam, ab52866, 1:1000
- ⁇ -GAPDH Santa Cruz, sc-32233, 1:1000
- Polysome profiling was performed as follows. A 10–50% sucrose gradient solution (Acros Organics, AC419760050) containing 100 ⁇ g/mL cycloheximide (CHX) (Sigma, C4859) was prepared using Gradient Master (Biocomp, B108-2). Prior to profiling, LNP-formulated mRNA was treated for 1 day in TRIM25 knockdown or knockout HCT116 cells.
- sucrose gradient solution A 10–50% sucrose gradient solution (Acros Organics, AC419760050) containing 100 ⁇ g/mL cycloheximide (CHX) (Sigma, C4859) was prepared using Gradient Master (Biocomp, B108-2). Prior to profiling, LNP-formulated mRNA was treated for 1 day in TRIM25 knockdown or knockout HCT116 cells.
- CHX cycloheximide
- PBS polysome-extraction buffer
- PBS polysome-extraction buffer
- PBS 50 mM Tris-HCl pH 7.5, 100 mM KCl, 5 mM MgCl2, 0.5 mM TCEP, 0.5% NP-40
- CHX polysome-extraction buffer
- protease inhibitor phosphatase inhibitor
- RNase inhibitor RNase inhibitor
- the obtained lysate was carefully layered onto a sucrose gradient and centrifuged at 36,000 rpm for 2 h using an ultracentrifuge (Beckman Coulter Ultracentrifuge Optima XE, SW41Ti rotor). The fractionated samples were then analyzed using a Bio-Rad EM-1 Econo UV detector, and 1 mL of each sample was collected using Bio-Rad 7318303 and subjected to RT-qPCR analysis.
- RNA immunoprecipitation was performed as follows. Normal rabbit IgG (Cell Signaling, 2792S) or a monoclonal rabbit antibody specific for TRIM25 (Abcam, ab167154) was conjugated to protein A sepharose beads (GE Healthcare, 17-5138-01) pretreated with salmon sperm DNA (Invitrogen, 15632011).
- HCT116 cells were transfected with LNP formulations containing Fluc mRNA containing m1 ⁇ or U and cultured for 1 day. Cells were then lysed in 0.2% NP-40 RIP buffer containing protease inhibitors, phosphatase inhibitors, and RNase inhibitors.
- RNA samples were incubated with antibody-conjugated beads at 4°C for 2 h and then washed five times. In the final washing step, some beads were reserved for Western blotting to confirm TRIM25 immunoprecipitation, while the coprecipitated RNA and input samples from the remaining beads were extracted using TRIzol. For quantification, 1 ng of spike-in RNA was added. The extracted RNA was reverse-transcribed and quantified by RT-qPCR, and the relative enrichment compared to the input was calculated based on the spike-in-normalized RNA level.
- Recombinant TRIM25 protein with a His10-eYFP-SUMOstar-Strep tag at its N-terminus was heterologously expressed in suspension-cultured HEK293E cells.
- Cells were transiently transfected at a density of approximately 7 ⁇ 10 5 cells/mL, and 0.15 mg of plasmid DNA and 1.5 mg of linear polyethylenimine (PEI) were mixed in the presence of 1% DMSO per 0.5 L culture. After transfection, cells were cultured at 33 °C for 72 h.
- PEI linear polyethylenimine
- the supernatant was loaded onto Ni-NTA Superflow resin (Qiagen, 1018142), washed with buffer A containing 20 mM imidazole (Sigma-Aldrich, I202), and eluted with buffer A containing 200 mM imidazole.
- the mixture was then incubated overnight with SUMOstar protease (LifeSensors, 4110) and Benzonase nuclease (Sigma-Aldrich, E1014).
- the sample was then loaded onto Strep-Tactin Superflow resin (IBA Lifesciences, 4-4030-025), washed with buffer A, and eluted with buffer A containing 50 mM biotin (IBA Lifesciences, 2-1016-005).
- the eluted protein was filtered through a 0.22 ⁇ m membrane filter to remove aggregates, and then buffer exchange was performed using a HiPrep 26/10 Desalting column (GE Healthcare, 17-5087-01).
- the final purified TRIM25 protein was equilibrated in a buffer containing 5 mM Tris-HCl pH 7.0, 150 mM NaCl, and 0.5 mM TCEP, rapidly frozen in liquid nitrogen, and stored at -80 °C.
- RNA pulldown experiments were performed as follows. Briefly, 94-nt long UTR1 or UTR2 RNAs with a 30-nt poly(A) at the 3′ end were synthesized through T7 polymerase-based in vitro transcription, resulting in a total RNA length of 130 nt, consisting of a 6-nt T7 promoter sequence at the 5′ end, the UTR body (94 nt), and a 30-nt poly(A) at the 3′ end.
- Poly(A)+ IVT RNA bait was bound to Oligo d(T)25 magnetic beads (NEB, S1419S) by incubation at 25 °C and 1,400 rpm in a thermomixer for 1 h, followed by overnight rotation at 4 °C.
- RNA baits 20-nt CALM1 UTR RNA (Dharmacon) with biotin attached to the terminal was synthesized and bound to Streptavidin magnetic beads (Thermo Scientific, 88816) by rotation overnight at 4 °C.
- Tris-HCl-based RNA pulldown buffer 50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 5% glycerol, 0.5 mM TCEP, 1% Triton X-100
- potassium phosphate buffer was used instead of Tris-HCl for pH adjustment.
- Protease inhibitors, phosphatase inhibitors, and RNase inhibitors were added to all buffers according to the designated pH conditions before the experiment. Information on the sequences of the RNA baits used is shown in Table 7.
- RNA-bound beads were reacted with RNA-bound beads at 4 °C for 2 hours.
- the beads were then washed five times and transferred to new protein low-binding tubes (Eppendorf, Z666505) to minimize contamination in the final wash step.
- the precipitated protein samples were eluted with sample buffer (Bio-Rad, 161-0747) containing 50 mM TCEP (Thermo Scientific, 77720) and analyzed by western blotting.
- TRIM25 knockout cells For endogenous TRIM25 analysis, HCT116 wild-type or TRIM25 knockout cells were used, and for exogenous TRIM25 analysis, TRIM25 knockout cells were transfected with GFP, TRIM25 wild-type, or TRIM25 7KA RNA-binding mutant expression plasmids and then lysed.
- FLAG-tagged TRIM25 wild-type, 7KA, or R54P mutant proteins were exogenously expressed in HCT116 TRIM25 knockout cells. Two days after plasmid transfection, cells were lysed and FLAG-tagged TRIM25 proteins were pulled down by reaction with ⁇ -FLAG M2 magnetic beads (Millipore, M8823).
- the amount of protein was measured by BSA-based quantification on SDS-PAGE gels, and beads corresponding to 4.5 pmol per protein were used for the in vitro ubiquitination reaction.
- the reaction was performed in a final volume of 50 ⁇ L of in vitro ubiquitination buffer (50 mM Tris pH 7.5, 150 mM NaCl, 5 mM MgCl2, 2 mM ATP, 0.5 mM TCEP) containing the following components: E1 enzyme: UBE1 125 ng (UBPBio, B1101), E2 enzyme: 6XHis-UbE2D3 250 ng (UBPBio, C1601), ubiquitin: 500 ng (UBPBio, E1100), m1 ⁇ or U IVT RNA: 5 pmol
- the mixture was shaken at 1,300 rpm in a thermomixer and incubated at 37 °C for 30 min.
- the reaction was terminated by adding sample buffer containing 50 mM TCEP and heating, and then analyzed by Western blotting using ⁇ -TRIM25 and ⁇ -Ubiquitin antibodies.
- HCT116 wild-type cells were seeded on 18-mm diameter microscope coverslips (Marienfeld, HSU-0111580) one day before LNP transfection.
- Fluc mRNA containing m1 ⁇ or U was formulated into LNPs and transfected for 6 h at two concentrations.
- the low concentration was 10 ng/mL, which is the standard condition for this study, and the high concentration was 500 ng/mL, which is 50 times the standard condition.
- cells were treated with PBS or LNP alone in the same volume as that used for LNP-mRNA transfection.
- As a stress condition control that induces G3BP focus formation cells were treated with 0.5 mM sodium arsenite (NaAsO2) (Sigma, S7400) for 45 min or transfected with 2 ⁇ g/mL of poly(I:C) for 12 h using Lipofectamine RNAiMAX.
- the coverslips were washed six times with PBS-T and stained with Alexa Fluor fluorescent secondary antibodies corresponding to the corresponding antibodies: Alexa Fluor 488 ( ⁇ -Mouse, Invitrogen, A-21202, 1:400) for ⁇ -G3BP1 and Alexa Fluor 594 ( ⁇ -Rabbit, Invitrogen, A-21207, 1:400) for ⁇ -TRIM25.
- Alexa Fluor 488 ⁇ -Mouse, Invitrogen, A-21202, 1:400
- Alexa Fluor 594 ⁇ -Rabbit, Invitrogen, A-21207, 1:400
- Protein-protein interaction network analysis of candidate genes for key positive regulators of LNP-mRNA was performed using STRING (v12.0).
- the connection lines between nodes in the network represent the strength of the interaction evidence calculated by the STRING software and were visualized with default settings.
- Reliable candidate genes under the GFPLow condition were defined as those that met both of the following two criteria: (1) FDR ⁇ 0.05 under the most stringent condition and (2) genes in the top 1% of pairwise binning conditions (e.g., genes with FDR ⁇ 0.05 at 0-2.5% GFPLow and in the top 1% at 2.5-5% GFPLow).
- High-confidence candidate genes selected under four GFPLow conditions [m1 ⁇ 0-2.5% and 2.5-5%], [U 0-2.5% and 2.5-5%]) were used as inputs for STRING analysis to construct protein interaction networks.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- General Health & Medical Sciences (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
본 발명은 TRIM25 발현 또는 활성 억제를 통한 세포 내 외인성 RNA의 안정성 또는 발현 증가용도에 관한 것이다.The present invention relates to the use of increasing the stability or expression of exogenous RNA in a cell by inhibiting TRIM25 expression or activity.
바이러스 RNA 및 치료용 mRNA와 같은 외인성 RNA는 세포 내로 진입하기 위해 세포 인자들과 상호작용해야 하며, 동시에 세포의 방어 기전을 회피해야 한다. 즉, 효과적인 mRNA 치료제를 개발하려면 이러한 세포 장벽을 활용하고 극복해야 한다. 최근 mRNA 기술의 발전은 유전자 전달 효율을 크게 향상시켰으며, mRNA를 중요한 치료 수단으로 자리잡게 하였고, COVID-19 팬데믹 동안 수십억 명에게 사용되었다. 치료용 mRNA는 in vitro transcription(IVT)을 통해 합성되며, 이온성 지질, 인지질, 콜레스테롤, 폴리에틸렌글라이콜(PEG) 결합 지질로 구성된 지질나노입자(LNP)에 캡슐화된다. 이후, 산성화된 엔도솜 내에서 이온성 지질이 양성화되어 엔도솜 막을 파괴하고 mRNA가 세포질로 방출된다.Exogenous RNA, such as viral RNA and therapeutic mRNA, must interact with cellular factors to enter cells while simultaneously evading cellular defense mechanisms. Therefore, developing effective mRNA therapeutics requires exploiting and overcoming these cellular barriers. Recent advances in mRNA technology have significantly improved gene delivery efficiency, establishing mRNA as a crucial therapeutic tool and reaching billions of people during the COVID-19 pandemic. Therapeutic mRNA is synthesized via in vitro transcription (IVT) and encapsulated in lipid nanoparticles (LNPs) composed of ionic lipids, phospholipids, cholesterol, and polyethylene glycol (PEG)-linked lipids. Within acidified endosomes, the ionic lipids protonate, disrupting the endosomal membrane and releasing the mRNA into the cytoplasm.
현재 임상에서 사용되는 mRNA 백신은 5′ 캡, poly(A) 꼬리, N1-메틸슈도유리딘(m1Ψ) 변형을 포함한 선형 구조를 가지며, 이는 단백질 발현을 향상시킨다. mRNA 백신의 지속적인 발현을 위해 환형 RNA(circRNA) 및 자가증폭 RNA(saRNA)와 같은 대체 플랫폼도 연구되고 있다. m1Ψ 변형은 선형 mRNA에서는 유리하지만, 내부 리보솜 진입 부위(IRES)와 같은 시스-요소를 방해하여 대체 플랫폼과는 호환되지 않으며, 이에 대한 추가 연구가 필요하다.Current clinically used mRNA vaccines have a linear structure, including a 5′ cap, a poly(A) tail, and an N1-methylpseudouridine (m1Ψ) modification, which enhances protein expression. Alternative platforms, such as circular RNA (circRNA) and self-amplifying RNA (saRNA), are also being explored for sustained mRNA vaccine expression. While the m1Ψ modification is advantageous in linear mRNA, it interferes with cis-elements such as the internal ribosome entry site (IRES), making it incompatible with alternative platforms and requiring further research.
mRNA의 생물학적 중요성과 다양한 응용 가능성에도 불구하고, 외인성 mRNA가 세포 내 기전과 어떻게 상호작용하는지에 대한 이해는 아직 제한적이다. 일 예시로, IVT mRNA를 이용한 어레이 기반 유전체 기능 교란 스크리닝이 수행된 바 있으며, 이 연구를 통해 막 수송과 엔도솜 성숙 과정의 중요성이 밝혀졌다[SLAS Discov 25, 605-617]. 하지만 이러한 어레이 스크린은 5-메톡시유리딘(mo5U)으로 변형된 mRNA만을 사용하였고, 확장성 측면에서도 제한이 있다. LNP-mRNA의 다양한 응용 가능성과는 달리, 그 조절 기전은 아직 충분히 밝혀지지 않은 바, 외인성 RNA의 생애주기를 조절하는 세포 내 과정을 더 깊이 이해하는 것은 mRNA 치료제 개발을 진전시키는 데 중요한 과제라고 볼 수 있다.Despite the biological importance and diverse applications of mRNA, our understanding of how exogenous mRNA interacts with cellular mechanisms remains limited. For example, array-based genome functional disruption screening using IVT mRNA has been performed, revealing the importance of membrane trafficking and endosomal maturation [SLAS Discov 25, 605-617]. However, this array screen only used mRNA modified with 5-methoxyuridine (mo5U), limiting scalability. Despite the diverse applications of LNP-mRNA, its regulatory mechanisms remain largely unknown. Therefore, a deeper understanding of the cellular processes that regulate the life cycle of exogenous RNA is crucial for advancing the development of mRNA therapeutics.
본 연구자들은 외인성 mRNA에 영향을 미치는 세포 내 조절 인자를 체계적으로 규명하기 위해, 전장 유전체 수준의 CRISPR-Cas9 knockout 스크리닝을 수행하였다. 3개의 전체 유전체 녹아웃(KO) 스크리닝을 통합하여 LNP mRNA 전달에 중요한 세포 단백질을 확인하였으며, TRIM25가 핵심 음성 조절 인자임을 확인하여 본 발명을 완성하게 되었다.To systematically identify intracellular regulatory factors affecting exogenous mRNA, we performed a genome-wide CRISPR-Cas9 knockout screen. By integrating three genome-wide knockout (KO) screens, we identified cellular proteins critical for LNP mRNA delivery, confirming TRIM25 as a key negative regulator, thereby completing the present invention.
일 양상은 TRIM25의 발현 또는 활성 억제제를 포함하는 세포 내 RNA의 안정성 또는 발현 증가용 조성물을 제공하는 것이다.One aspect is to provide a composition for increasing the stability or expression of intracellular RNA comprising an inhibitor of TRIM25 expression or activity.
다른 양상은 인비트로에서 세포 내 목적 RNA의 발현 또는 활성을 증가시키는 방법을 제공하는 것이다.Another aspect is to provide a method for increasing the expression or activity of a target RNA within a cell in vitro.
다른 양상은 세포 내 RNA의 안정성 또는 발현 증가용 물질을 스크리닝하는 방법을 제공하는 것이다.Another aspect is to provide a method for screening substances for increasing the stability or expression of RNA within a cell.
다른 양상은 RNA의 발현 또는 활성을 증진시키는 핵산의 화학적 변형을 스크리닝하는 방법을 제공하는 것이다.Another aspect is to provide a method for screening chemical modifications of nucleic acids that enhance expression or activity of RNA.
일 양상은 TRIM25의 발현 또는 활성 억제제를 포함하는 세포 내 RNA의 안정성 또는 발현 증가용 조성물을 제공한다.One aspect provides a composition for increasing the stability or expression of intracellular RNA comprising an inhibitor of TRIM25 expression or activity.
본 명세서에서 용어 "TRIM25(Tripartite motif containing 25)"는 TRIM (Tripartite motif) 단백질 계열에 속하는 E3 유비퀴틴 리가아제로, RING 지니어스 구조(RING finger), B-box 도메인, coiled-coil 도메인으로 구성된 tripartite motif를 가진다. TRIM25는 특정 표적 단백질에 유비퀴틴을 결합시키는 기능을 수행하며 주로 선천면역 반응 및 항바이러스 반응에서 주요한 역할을 한다.The term "TRIM25 (Tripartite motif containing 25)" used herein refers to an E3 ubiquitin ligase belonging to the TRIM (Tripartite motif) protein family, which has a tripartite motif consisting of a RING genius structure (RING finger), a B-box domain, and a coiled-coil domain. TRIM25 functions to conjugate ubiquitin to specific target proteins and plays a major role mainly in the innate immune response and antiviral response.
일 구체예에 있어서, 상기 조성물은 TRIM25 보조인자의 발현 또는 활성 억제제를 추가로 포함할 수 있다. 구체적으로, 상기 조성물은 TRIM25 보조인자로 작용할 수 있는 엔도리보뉴클레이스(endoribonuclease)의 발현 또는 활성 억제제를 추가로 포함할 수 있다.In one specific example, the composition may further comprise an inhibitor of the expression or activity of a TRIM25 cofactor. Specifically, the composition may further comprise an inhibitor of the expression or activity of an endoribonuclease capable of acting as a TRIM25 cofactor.
일 구체예에 있어서, 상기 조성물은 TRIM25 보조인자의 발현 또는 활성 억제제와 병용하여 처리될 수 있다.In one specific example, the composition may be administered in combination with an inhibitor of TRIM25 cofactor expression or activity.
일 구체예에 있어서, 상기 조성물은 N4BP1, KHNYN 및 ZAP으로 이루어지는 군에서 선택된 어느 하나 이상의 발현 또는 활성 억제제를 추가로 포함할 수 있다.In one specific example, the composition may further comprise one or more expression or activity inhibitors selected from the group consisting of N4BP1, KHNYN, and ZAP.
일 구체예에 있어서, 상기 조성물은 N4BP1, KHNYN 및 ZAP으로 이루어지는 군에서 선택된 어느 하나 이상의 발현 또는 활성 억제제와 병용하여 처리될 수 있다.In one specific example, the composition may be treated in combination with one or more expression or activity inhibitors selected from the group consisting of N4BP1, KHNYN, and ZAP.
본 명세서에서 용어 "N4BP1 (NEDD4 Binding Protein 1)"은 E3 유비퀴틴 리가아제인 NEDD4와 결합하는 단백질로, 주로 단백질 분해 조절, 면역 신호전달, RNA 대사 등에서 기능하는 다기능성 조절 단백질이다.The term "N4BP1 (NEDD4 Binding Protein 1)" in this specification refers to a protein that binds to NEDD4, an E3 ubiquitin ligase, and is a multifunctional regulatory protein that mainly functions in protein degradation regulation, immune signaling, RNA metabolism, etc.
본 명세서에서 용어 "KHNYN (KH And NYN Domain Containing)"은 KH(K-homology) 도메인과 NYN 뉴클레이스 도메인을 모두 포함하는 단백질로, 주로 RNA 결합 및 RNA 절단 활성과 관련된 항바이러스성 효소 단백질로 알려져 있다.The term "KHNYN (KH And NYN Domain Containing)" in this specification refers to a protein containing both a KH (K-homology) domain and a NYN nuclease domain, which is known as an antiviral enzyme protein mainly associated with RNA binding and RNA cleavage activities.
본 명세서에서 용어 "ZAP (Zinc-finger Antiviral Protein)"은 아연지손(zinc-finger) 구조를 가진 항바이러스 단백질로, 바이러스 RNA를 인식하여 분해를 유도함으로써 바이러스 복제를 억제하는 선천면역 인자이다.The term "ZAP (Zinc-finger Antiviral Protein)" used herein refers to an antiviral protein with a zinc-finger structure, which is an innate immune factor that inhibits viral replication by recognizing viral RNA and inducing its degradation.
상기 병용 처리는 상기 TRIM25의 발현 또는 활성 억제제 및 상기 N4BP1, KHNYN 및/또는 ZAP의 발현 또는 활성 억제제를 순차적으로, 개별적으로, 또는 임의 순서로 투여하는 것을 의미할 수 있다.The above combination treatment may mean administering the TRIM25 expression or activity inhibitor and the N4BP1, KHNYN and/or ZAP expression or activity inhibitor sequentially, separately, or in any order.
일 구체예에 있어서, 상기 RNA는 외인성 RNA(exogenus RNA)일 수 있다. 상기 외인성 RNA란 세포 외부에서 유래하여 세포 내로 인위적 또는 자연적 방법에 의해 도입된 RNA를 의미하며 일반적으로 생체 내에서 내인적으로 합성되는 RNA와는 구별된다. 상기 외인성 RNA는 진핵세포, 원핵세포 또는 바이러스 등 다양한 생물학적 기원을 가질 수 있으며, 인공적으로 합성된 RNA도 포함될 수 있다. In one specific example, the RNA may be exogenous RNA. Exogenous RNA refers to RNA derived from outside the cell and introduced into the cell through artificial or natural means, and is generally distinguished from RNA synthesized endogenously within the body. The exogenous RNA may have various biological origins, such as eukaryotic cells, prokaryotic cells, or viruses, and may also include artificially synthesized RNA.
상기 조성물은 세포 내 TRIM25의 발현 또는 활성을 억제하여 세포 내로 도입된 외인성 RNA의 안정성 또는 발현을 증가하는 것일 수 있다. TRIM25는 외인성 RNA에 대한 세포의 면역 반응을 유도하거나 RNA의 분해를 유도할 수 있는 바, 상기 조성물은 상기 TRIM25의 기능을 억제함으로써 외인성 RNA의 분해를 방지하고 상기 RNA의 생물학적 효능을 증가시킬 수 있다.The composition may increase the stability or expression of exogenous RNA introduced into a cell by inhibiting the expression or activity of TRIM25 within the cell. TRIM25 can induce a cellular immune response to exogenous RNA or induce RNA degradation, and thus the composition may prevent degradation of exogenous RNA and increase the biological efficacy of the RNA by inhibiting the function of TRIM25.
일 구체예에 있어서, 상기 외인성 RNA는 mRNA 백신일 수 있다. 상기 mRNA 백신은 특정 항원을 발현하도록 암호화된 메신저 RNA를 포함하며, 숙주 세포 내에서 항원 단백질이 발현됨으로써 면역 반응을 유도하는 백신 플랫폼을 의미한다. 상기 조성물은 mRNA 백신의 안정성 및 단백질 발현 효율을 증가시킬 수 있다.In one specific example, the exogenous RNA may be an mRNA vaccine. An mRNA vaccine comprises messenger RNA encoded to express a specific antigen, and refers to a vaccine platform that induces an immune response by expressing the antigen protein within a host cell. The composition can increase the stability and protein expression efficiency of the mRNA vaccine.
일 구체예에 있어서, 상기 외인성 RNA는 지질나노입자(Lipid Nanoparticle, LNP)에 캡슐화되어 세포 내로 전달된 것일 수 있다. 상기 지질나노입자는 핵산 또는 약물 전달을 위한 운반체로 사용되는 지질 분자로 구성된 나노미터 크기의 입자를 의미한다. 상기 지질나노입자는 이온성 지질, 인지질, 콜레스테롤 및 폴리에틸렌글라이콜(PEG) 결합 지질로 이루어진 군에서 선택되는 어느 하나 이상의 지질을 포함하는 것일 수 있다. In one specific example, the exogenous RNA may be encapsulated in a lipid nanoparticle (LNP) and delivered into the cell. The lipid nanoparticle refers to a nanometer-sized particle composed of lipid molecules used as a carrier for nucleic acid or drug delivery. The lipid nanoparticle may include one or more lipids selected from the group consisting of ionic lipids, phospholipids, cholesterol, and polyethylene glycol (PEG)-linked lipids.
상기 지질나노입자는 단일 또는 복수 종류의 지질 성분으로 구성될 수 있으며, 상기 성분들은 상호간의 조성비 및 구조에 따라 LNP의 크기, 전하, 안정성 및 세포 투과성 등에 영향을 미칠 수 있다. 바람직하게는, 상기 지질나노입자는 양이온성 이온성 지질을 포함하여 RNA와 정전기적으로 결합함으로써 RNA의 효율적인 포집 및 전달을 가능하게 한다.The lipid nanoparticles may be composed of a single or multiple types of lipid components, and the components may affect the size, charge, stability, and cell permeability of the LNPs depending on their composition ratio and structure. Preferably, the lipid nanoparticles include cationic ionic lipids to electrostatically bind to RNA, thereby enabling efficient capture and delivery of RNA.
일 구체예에 있어서, 상기 RNA는 mRNA, 원형 RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, 리보자임 및 ncRNA로 이루어지는 군에서 선택되는 어느 하나 이상일 수 있다.In one specific example, the RNA may be at least one selected from the group consisting of mRNA, circular RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, ribozyme, and ncRNA.
상기 TRIM25의 발현 억제제는 TRIM25 유전자에 특이적으로 결합하는 siRNA, shRNA, 올리고뉴클레오타이드, 안티센스 뉴클레오티드(antisense nucleotide) 및 sgRNA로 구성된 군에서 선택되는 어느 하나 이상을 포함할 수 있으나, 이에 제한되지 않는다.The above TRIM25 expression inhibitor may include, but is not limited to, one or more selected from the group consisting of siRNA, shRNA, oligonucleotide, antisense nucleotide, and sgRNA that specifically bind to the TRIM25 gene.
상기 TRIM25의 활성 억제제는 TRIM25 단백질에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편, 상호작용 단백질, PROTAC, 올리고펩타이드, 리간드, 나노입자 (nanoparticles), 압타머 (aptamer), 아비머 (avidity multimer) 및 펩티도모방체 (peptidomimetics)로 구성된 군에서 선택되는 어느 하나 이상을 포함할 수 있으나, 이에 제한되지 않는다.The above TRIM25 activity inhibitor may include, but is not limited to, one or more selected from the group consisting of antibodies or antigen-binding fragments thereof that specifically bind to TRIM25 protein, interacting proteins, PROTACs, oligopeptides, ligands, nanoparticles, aptamers, avidity multimers, and peptidomimetics.
상기 N4BP1, KHNYN 및/또는 ZAP의 발현 억제제는 각각 N4BP1, KHNYN 및/또는 ZAP 유전자에 특이적으로 결합하는 siRNA, shRNA, 올리고뉴클레오타이드, 안티센스 뉴클레오티드(antisense nucleotide) 및 sgRNA로 구성된 군에서 선택되는 어느 하나 이상을 포함할 수 있으나, 이에 제한되지 않는다.The above N4BP1, KHNYN and/or ZAP expression inhibitor may include, but is not limited to, one or more selected from the group consisting of siRNA, shRNA, oligonucleotide, antisense nucleotide and sgRNA that specifically bind to the N4BP1, KHNYN and/or ZAP genes, respectively.
상기 N4BP1, KHNYN 및/또는 ZAP의 활성 억제제는 각각 N4BP1, KHNYN 및/또는 ZAP 단백질에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편, 상호작용 단백질, 올리고펩타이드, 리간드, 나노입자 (nanoparticles), 압타머 (aptamer), PROTAC, 아비머 (avidity multimer) 및 펩티도모방체 (peptidomimetics)로 구성된 군에서 선택되는 어느 하나 이상을 포함할 수 있으나, 이에 제한되지 않는다.The above N4BP1, KHNYN and/or ZAP activity inhibitor may include, but is not limited to, one or more selected from the group consisting of antibodies or antigen-binding fragments thereof, interacting proteins, oligopeptides, ligands, nanoparticles, aptamers, PROTACs, avidity multimers and peptidomimetics that specifically bind to N4BP1, KHNYN and/or ZAP proteins, respectively.
상기 조성물은 임상 투여 시 경구 또는 비경구로 투여가 가능하다. 예를 들어, 경구, 정맥 내, 근육내, 동맥 내, 골수 내, 골수강 내, 피하, 심실 내, 경피, 피 내, 직장, 질 내, 복강내, 안 내, 망막하, 초자체 내, 국소, 점막, 경비, 구강, 경장, 초자체, 종양 내, 설하(혀 밑), 비강 내를 포함한, 여러가지 경로의 하나 이상에 의해 투여될 수 있다.The composition may be administered orally or parenterally during clinical administration. For example, it may be administered by one or more of various routes, including oral, intravenous, intramuscular, intraarterial, intramedullary, intramedullary, subcutaneous, intraventricular, transdermal, intradermal, rectally, intravaginally, intraperitoneally, intraocularly, subretinal, intravitreal, topical, mucosal, transnasal, buccal, enteral, intravitreal, intratumoral, sublingual (under the tongue), and intranasal.
상기 조성물은 제형이 특히 제한되지는 않으며, 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다. 상기 조성물을 제제화 할 경우 당업계에서 일반적으로 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. The above composition is not particularly limited in its formulation, and may be formulated and used in the form of oral formulations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., as well as external preparations, suppositories, and sterile injectable solutions, according to conventional methods. When formulating the above composition, it may be prepared using diluents or excipients such as fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants commonly used in the art.
TRIM25는 세포 내에서 외인성 RNA의 음성 조절 인자로 작용할 수 있다. 본 발명의 상기 조성물은 TRIM25의 발현 또는 활성을 억제함으로써, 세포가 외인성 RNA를 이질적인 RNA로 인식하여 이를 분해하거나 면역반응을 유도하는 것을 방지할 수 있다. 이를 통해, 상기 조성물은 세포 내 외인성 RNA의 안정성 증가, 세포 내 외인성 RNA의 발현 증가 또는 둘 모두를 유도할 수 있다. 따라서, 본 발명의 상기 조성물은 외인성 RNA를 치료제로 활용하는데 유용하게 사용될 수 있다. 상기 조성물은 시험관내(in vitro), 생체내(in vivo), 또는 생체외(ex vivo)에서 목적하는 세포 내 RNA의 안정성 및/또는 발현을 증가시킬 수 있다.TRIM25 can act as a negative regulator of exogenous RNA within cells. The composition of the present invention can prevent cells from recognizing exogenous RNA as foreign RNA and degrading it or inducing an immune response by inhibiting the expression or activity of TRIM25. Thus, the composition can induce increased stability of exogenous RNA within cells, increased expression of exogenous RNA within cells, or both. Therefore, the composition of the present invention can be useful for utilizing exogenous RNA as a therapeutic agent. The composition can increase the stability and/or expression of a target intracellular RNA in vitro, in vivo, or ex vivo.
다른 양상은 개체에 TRIM25의 발현 또는 활성 억제제를 처리하는 단계를 포함하는 세포 내 RNA의 안정성 또는 발현을 증가시키는 방법을 제공한다.Another aspect provides a method for increasing the stability or expression of RNA within a cell, comprising treating the subject with an inhibitor of TRIM25 expression or activity.
다른 양상은 TRIM25의 발현 또는 활성 억제제의 세포 내 RNA의 안정성 또는 발현 증가 용도를 제공한다.Another aspect provides the use of TRIM25 expression or activity inhibitors to increase the stability or expression of intracellular RNA.
다른 양상은 인비트로(in vitro)에서 세포 내 목적 RNA의 발현 또는 활성을 증가시키는 방법을 제공한다. 구체적으로, 상기 방법은 Another aspect provides a method for increasing the expression or activity of a target RNA in a cell in vitro. Specifically, the method comprises:
인비트로에서 세포 내 TRIM25의 발현 또는 활성을 억제시키는 단계; 및A step of inhibiting the expression or activity of TRIM25 in cells in vitro; and
목적 RNA를 세포에 도입시키는 단계를 포함한다.It comprises a step of introducing target RNA into a cell.
상기 TRIM25의 발현 또는 활성의 억제는 TRIM25 유전자 녹아웃, 녹다운, 조건부 유전자 녹아웃, 유전자 변경 및 RNA 간섭으로 이루어진 군에서 선택되는 어느 하나 이상을 포함할 수 있으나, 이에 제한되지 않는다.The inhibition of the expression or activity of TRIM25 may include, but is not limited to, any one or more selected from the group consisting of TRIM25 gene knockout, knockdown, conditional gene knockout, genetic modification, and RNA interference.
일 구체예에 있어서, 상기 방법은 N4BP1, KHNYN 및 ZAP으로 이루어지는 군에서 선택된 어느 하나 이상의 발현 또는 활성을 추가로 억제시키는 단계를 포함할 수 있다.In one embodiment, the method may further comprise a step of inhibiting the expression or activity of one or more selected from the group consisting of N4BP1, KHNYN, and ZAP.
상기 N4BP1, KHNYN 및/또는 ZAP의 발현 또는 활성을 억제시키는 단계는 TRIM25의 발현 또는 활성을 억제시키는 단계와 순차적으로, 개별적으로, 또는 임의 순서로 수행될 수 있다.The step of inhibiting the expression or activity of N4BP1, KHNYN and/or ZAP may be performed sequentially, individually, or in any order with the step of inhibiting the expression or activity of TRIM25.
일 구체예에 있어서, 상기 목적 RNA의 발현 또는 활성 증가는 목적 RNA 또는 상기 목적 RNA가 암호화하는 폴리펩타이드의 생산을 증가시키는 것일 수 있다.In one specific example, increasing the expression or activity of the target RNA may increase the production of the target RNA or a polypeptide encoded by the target RNA.
일 구체예에 있어서, 상기 목적 RNA는 외인성 RNA일 수 있다.In one specific example, the target RNA may be an exogenous RNA.
일 구체예에 있어서, 상기 목적 RNA는 지질나노입자(Lipid Nanoparticle, LNP)에 캡슐화되어 세포 내로 전달된 것일 수 있다.In one specific example, the target RNA may be encapsulated in a lipid nanoparticle (LNP) and delivered into the cell.
일 구체예에 있어서, 상기 RNA는 mRNA, 원형 RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, 리보자임 및 ncRNA로 이루어지는 군에서 선택되는 어느 하나 이상일 수 있다.In one specific example, the RNA may be at least one selected from the group consisting of mRNA, circular RNA, dsRNA, shRNA, miRNA, gRNA, saRNA, lncRNA, taRNA, ribozyme, and ncRNA.
다른 양상은 세포 내 RNA의 안정성 또는 발현 증가용 물질을 스크리닝하는 방법을 제공한다. 구체적으로, 상기 방법은 Another aspect provides a method for screening a substance for increasing the stability or expression of RNA within a cell. Specifically, the method
후보 물질을 세포에 처리하는 단계; A step of treating a candidate substance into cells;
상기 후보 물질이 처리된 세포 내에서 TRIM25의 세포 내 발현 또는 활성을 측정하는 단계; 및 A step of measuring the intracellular expression or activity of TRIM25 in cells treated with the above candidate substance; and
무처리 대조군 대비 TRIM25의 세포 내 발현 또는 활성을 변화시킨 후보 물질을 선별하는 단계를 포함한다.It includes a step of selecting a candidate substance that changes the intracellular expression or activity of TRIM25 compared to an untreated control group.
일 구체예에 있어서, 상기 선별 단계는 무처리 대조군 대비 TRIM25의 세포 내 발현 또는 활성이 감소한 경우, 상기 후보 물질을 세포 내 RNA의 안정성 또는 발현 증가용 물질로 판단하는 단계를 추가로 포함할 수 있다. In one specific example, the selection step may further include a step of determining the candidate substance as a substance for increasing the stability or expression of intracellular RNA when the intracellular expression or activity of TRIM25 is reduced compared to an untreated control group.
일 구체예에 있어서, 상기 TRIM25의 세포 내 발현의 측정은 검출 가능한 신호를 발생시키는 표지 물질로 표지되거나 (표지 물질이, 예컨대, 화학적 (예컨대, 공유적 또는 비공유적), 재조합적, 또는 물리적으로, 결합되거나), 표지물질이 결합될 수 있는 tag이 부착된 형태로 표지한 후, 통상적인 효소 반응, 형광, 발광, 및/또는 방사선 검출을 통하여 표지물질로부터 발생하는 신호를 측정하는 것일 수 있다. 상기 신호의 측정은 이를 검출 또는 측정하는데 통상적으로 사용되는 모든 신호 검출 수단 (예컨대, 통상의 형광 현미경, 형광 카메라, 형광세기 측정 (정량) 장치 등)에 의하여 측정될 수 있다.In one specific example, the measurement of the intracellular expression of TRIM25 may be performed by labeling with a label that generates a detectable signal (the label is, for example, chemically (e.g., covalently or non-covalently), recombinantly, or physically bound) or by labeling in a form in which a tag to which the label can be bound is attached, and then measuring a signal generated from the label through a conventional enzymatic reaction, fluorescence, luminescence, and/or radiological detection. The measurement of the signal may be measured by any signal detection means conventionally used to detect or measure it (e.g., a conventional fluorescence microscope, a fluorescence camera, a fluorescence intensity measurement (quantitation) device, etc.).
다른 양상은, Another aspect is,
세포 내 TRIM25 유전자 또는 상기 유전자가 코딩하는 단백질의 발현 또는 활성을 억제시킨 실험 세포를 얻는 단계; A step of obtaining an experimental cell in which the expression or activity of the TRIM25 gene or the protein encoded by the gene is suppressed within the cell;
TRIM25의 발현 또는 활성이 억제된 실험 세포 및 정상 세포에 후보 화학적 변형을 갖는 RNA를 처리하는 단계; A step of treating RNA having a candidate chemical modification to experimental cells and normal cells in which TRIM25 expression or activity is suppressed;
상기 실험 세포와 정상 세포에서 후보 화학적 변형을 갖는 RNA의 발현 수준 또는 발현된 단백질의 활성 수준을 측정하는 단계; 및 A step of measuring the expression level of RNA having a candidate chemical modification or the activity level of the expressed protein in the above experimental cells and normal cells; and
실험 세포의 RNA의 발현 수준 또는 발현된 단백질의 활성 수준이 정상 세포의 RNA의 발현 수준 또는 발현된 단백질의 활성 수준과 동등한 경우 RNA 발현 또는 활성을 증진시키는 화학적 변형으로 판단하는 단계를 포함하는 RNA 발현 또는 활성을 증진시키는 핵산의 화학적 변형을 스크리닝하는 방법을 제공한다.A method for screening a chemical modification of a nucleic acid that enhances RNA expression or activity is provided, comprising a step of determining that the chemical modification enhances RNA expression or activity when the expression level of RNA of an experimental cell or the activity level of an expressed protein is equivalent to the expression level of RNA of a normal cell or the activity level of an expressed protein.
상기 RNA 및 TRIM25의 발현 또는 활성 억제에 관해서는 상기에서 기술한 바와 같다. 구체적으로, 상기 RNA는 지질나노입자(Lipid Nanoparticle, LNP)에 캡슐화되어 세포 내로 전달된 외인성 RNA일 수 있다.Regarding the inhibition of expression or activity of the above RNA and TRIM25, it is as described above. Specifically, the above RNA may be an exogenous RNA encapsulated in a lipid nanoparticle (LNP) and delivered into the cell.
본 명세서에서 용어 "핵산의 화학적 변형"은 핵산(DNA 또는 RNA)의 염기, 당, 인산 등의 화학구조가 변형된 것을 의미하며, 자연적 화학적 변형 및 인위적 화학적 변형을 모두 포함한다. The term "chemical modification of nucleic acid" in this specification means that the chemical structure of a nucleic acid (DNA or RNA) such as a base, sugar, or phosphate has been modified, and includes both natural chemical modification and artificial chemical modification.
TRIM25의 발현 또는 활성이 억제된 실험 세포 및 정상 세포에서 상기 외인성 RNA가 실질적으로 동등한 발현 또는 활성 수준을 보이는 경우, 상기 외인성 RNA는 TRIM25를 회피할 수 있는 화학적 변형을 포함하는 것으로 판단할 수 있다. 이에 따라, 상기 외인성 RNA는 RNA의 발현 또는 활성을 증진시키는 화학적 변형을 포함하는 것으로 간주될 수 있다.If the exogenous RNA exhibits substantially equivalent expression or activity levels in experimental cells in which TRIM25 expression or activity is suppressed and in normal cells, the exogenous RNA can be considered to contain a chemical modification capable of evading TRIM25. Accordingly, the exogenous RNA can be considered to contain a chemical modification that enhances RNA expression or activity.
또 다른 양상은, Another aspect is,
후보 화학적 변형을 갖는 RNA와 TRIM25의 결합 또는 상호작용을 측정하는 단계; 및 A step of measuring the binding or interaction of RNA with a candidate chemical modification and TRIM25; and
상기 후보 화학적 변형을 갖는 RNA와 TRIM25가 결합 또는 상호작용을 하지 않는 경우 후보 화학적 변형을 RNA 발현 또는 활성을 증진시키는 화학적 변형으로 판단하는 단계를 포함하는, RNA의 발현 또는 활성을 증진시키는 핵산의 화학적 변형을 스크리닝하는 방법을 제공한다.A method for screening a chemical modification of a nucleic acid that enhances expression or activity of RNA is provided, comprising a step of determining that the candidate chemical modification is a chemical modification that enhances RNA expression or activity if TRIM25 does not bind or interact with the RNA having the candidate chemical modification.
상기 RNA, TRIM25의 발현 또는 활성 억제, 핵산의 화학적 변형에 관해서는 상기에서 기술한 바와 같다. 구체적으로, 상기 RNA는 지질나노입자(Lipid Nanoparticle, LNP)에 캡슐화되어 세포 내로 전달된 외인성 RNA일 수 있다.Regarding the above RNA, inhibition of TRIM25 expression or activity, and chemical modification of nucleic acids, the same is as described above. Specifically, the RNA may be an exogenous RNA encapsulated in a lipid nanoparticle (LNP) and delivered into cells.
TRIM25는 세포 내에서 외인성 RNA의 음성 조절 인자로 작용할 수 있다. 후보 화학적 변형을 갖는 RNA 및 TRIM25가 결합 또는 상호작용하지 않는 경우, 상기 외인성 RNA는 TRIM25를 회피할 수 있는 화학적 변형을 포함하는 것으로 판단할 수 있다. 상기 후보 화학적 변형을 포함하는 RNA가 TRIM25를 회피하는 경우, 상기 RNA는 세포에 의해 분해되거나 면역 반응이 유도되는 것을 방지할 수 있다. 따라서, 상기와 같은 경우, 외인성 RNA는 RNA의 발현 또는 활성을 증진시키는 화학적 변형을 포함하는 것으로 간주될 수 있다.TRIM25 can act as a negative regulator of exogenous RNA within cells. If RNA with a candidate chemical modification does not bind or interact with TRIM25, the exogenous RNA can be considered to contain a chemical modification that evades TRIM25. If the RNA containing the candidate chemical modification evades TRIM25, the RNA can be prevented from being degraded by the cell or from inducing an immune response. Therefore, in such cases, the exogenous RNA can be considered to contain a chemical modification that enhances RNA expression or activity.
일 구체예에 있어서, 상기 결합 또는 상호작용의 측정은 pH 7.0 이하에서 수행되는 것일 수 있다.In one specific example, the measurement of the binding or interaction may be performed at a pH of 7.0 or lower.
본 발명은 다양한 변환을 가할 수 있고 여러 가지 실시예를 가질 수 있는 바, 이하 특정 실시예들을 도면에 예시하고 상세한 설명에 상세하게 설명하고자 한다. 그러나, 이는 본 발명을 특정한 실시 형태에 대해 한정하려는 것이 아니며, 본 발명의 사상 및 기술 범위에 포함되는 모든 변환, 균등물 내지 대체물을 포함하는 것으로 이해되어야 한다. 본 발명을 설명함에 있어서 관련된 공지 기술에 대한 구체적인 설명이 본 발명의 요지를 흐릴 수 있다고 판단되는 경우 그 상세한 설명을 생략한다.The present invention is susceptible to various modifications and embodiments. Specific embodiments are illustrated in the drawings and described in detail in the following detailed description. However, this is not intended to limit the present invention to specific embodiments, but rather to encompass all modifications, equivalents, and alternatives falling within the spirit and technical scope of the present invention. In describing the present invention, detailed descriptions of related known technologies will be omitted if they are deemed to obscure the gist of the present invention.
TRIM25는 LNP RNA 전달에 있어 핵심 음성 조절 인자로 작용하는 바, TRIM25의 발현 또는 활성을 억제하면 유전자 전달 효율을 크게 증가시킬 수 있으며 치료용 RNA 백신 설계 및 개발에 유용한 쓰임이 있다.TRIM25 acts as a key negative regulator in LNP RNA delivery. Inhibiting TRIM25 expression or activity can significantly increase gene delivery efficiency and is useful for designing and developing therapeutic RNA vaccines.
도 1은 전장 유전체 CRISPR-Cas9 녹아웃 스크리닝의 개략도이다. Figure 1 is a schematic diagram of whole-genome CRISPR-Cas9 knockout screening.
도 2는 LNP에 포뮬레이션된 mRNA(LNP-mRNAs)의 개략도이다.Figure 2 is a schematic diagram of mRNA formulated in LNP (LNP-mRNAs).
도 3은 m1Ψ 유무에 따른 EGFP LNP-mRNA 처리 세포의 GFP 발현을 유세포 분석 및 신호 정량 분석으로 평가한 결과이다.Figure 3 shows the results of evaluating GFP expression in EGFP LNP-mRNA treated cells with or without m1Ψ using flow cytometry and signal quantification analysis.
도 4는 PKR 녹다운 후 IVT mRNA 유전자 발현량 확인 및 이의 정량에 따른 dsRNA 부산물을 보여주는 dot blot 분석 결과이다.Figure 4 shows the results of a dot blot analysis showing the IVT mRNA gene expression level after PKR knockdown and the dsRNA byproduct according to its quantification.
도 5는 PKR 및 eIF2α의 인산화를 확인한 웨스턴 블롯 분석 결과, 폴리솜 프로파일링 결과, IFNB1 mRNA 수준 및 대표 ISG 발현 수준을 나타낸 그래프이다.Figure 5 is a graph showing the results of Western blot analysis confirming phosphorylation of PKR and eIF2α, polysome profiling results, IFNB1 mRNA levels, and representative ISG expression levels.
도 6은 GFPLow 및 GFPHigh 군의 sgRNA 분포를 분석한 도이다. A는 개별 sgRNA들의 리드 카운트 분포, B는 CRISPR 라이브러리 내 전체 sgRNA의 log2 농축도를 나타낸 산점도, C는 GFPLow 및 GFPHigh 군을 구분하는 FDR 순위 기반 산점도, 및 D는 서로 다른 조건 간 유의한 유전자들(FDR < 0.05)의 유전자 수준 log2 농축도에 대한 피어슨 상관계수를 나타낸 히트맵이다.Figure 6 is a diagram analyzing the distribution of sgRNAs in the GFPLow and GFPHigh groups. A is the read count distribution of individual sgRNAs, B is a scatter plot showing the log2 enrichment of all sgRNAs within the CRISPR library, C is a scatter plot based on FDR rank distinguishing the GFPLow and GFPHigh groups, and D is a heatmap showing the Pearson correlation coefficient for the gene-level log2 enrichment of significant genes (FDR < 0.05) across different conditions.
도 7은 CRISPR-KO 스크린 결과 및 GFPHigh 및 GFPLow 조건으로부터 유도된 유의한 유전자들의 주요 GO term 농축 결과이다.Figure 7 shows the results of the CRISPR-KO screen and the major GO term enrichment results of significant genes derived from GFPHigh and GFPLow conditions.
도 8은 스크린을 통해 확인된 양성 조절인자들의 단백질-단백질 상호작용 네트워크를 보여주는 STRING 분석 결과 및 상위 10개 후보의 GO term 농축 결과이다.Figure 8 shows the results of STRING analysis showing the protein-protein interaction network of positive regulators identified through the screen and the GO term enrichment results of the top 10 candidates.
도 9는 HSPG 생합성 및 V-ATPase에 관여하는 유전자들의 log2 농축도를 보여주는 히트맵이다.Figure 9 is a heatmap showing the log2 enrichment of genes involved in HSPG biosynthesis and V-ATPase.
도 10은 HSPG 경로 관련 유전자 또는 V-ATPase 서브유닛 유전자를 knockdown시킨 후 단백질 생성이 감소함을 관찰한 도(상) 및 녹다운 효율을 RT-qPCR로 측정한 결과(하)이다.Figure 10 shows (top) an observation of a decrease in protein production after knocking down a HSPG pathway-related gene or a V-ATPase subunit gene, and (bottom) the results of measuring the knockdown efficiency using RT-qPCR.
도 11은 HSPG 생합성 인자 또는 V-ATPase 서브유닛이 결핍된 HCT116 세포에서의 루시퍼레이스 발현을 관찰한 그래프이다.Figure 11 is a graph showing luciferase expression in HCT116 cells deficient in HSPG biosynthesis factors or V-ATPase subunits.
도 12는 HEK293T및 HeLa 세포를 사용하여 단백질 생성이 감소함을 관찰한 도(상) 및 녹다운 효율을 RT-qPCR로 측정한 결과(하)이다.Figure 12 shows the results of observing a decrease in protein production using HEK293T and HeLa cells (top) and measuring the knockdown efficiency using RT-qPCR (bottom).
도 13은 헤파린 처리 후 HCT116 세포에 형질감염된 EGFP mRNA로부터의 단백질 생성량을 관찰한 도이다.Figure 13 is a diagram showing the amount of protein produced from EGFP mRNA transfected into HCT116 cells after heparin treatment.
도 14는 BafA1, Pitstop-2 또는 Dynole 처리 후 HCT116 세포에 형질감염된 EGFP mRNA로부터의 단백질 생성량을 관찰한 도이다.Figure 14 shows the amount of protein produced from EGFP mRNA transfected into HCT116 cells after treatment with BafA1, Pitstop-2, or Dynole.
도 15는 헤파린 또는 BafA1 처리 후 세포 내 EGFP IVT mRNA 수준을 RT-qPCR 분석한 결과 그래프이다.Figure 15 is a graph showing the results of RT-qPCR analysis of intracellular EGFP IVT mRNA levels after heparin or BafA1 treatment.
도 16은 헤파린 또는 BafA1 처리 후 poly(A) 길이 분포를 Hire-PAT 분석으로 측정한 그래프이다.Figure 16 is a graph showing the poly(A) length distribution measured by Hire-PAT analysis after heparin or BafA1 treatment.
도 17은 LNP 2를 통해 HCT116 세포에 전달된 Fluc mRNA의 루시퍼레이스 발현량을 확인한 도이다.Figure 17 is a diagram confirming the luciferase expression level of Fluc mRNA delivered to HCT116 cells via LNP 2.
도 18은 GFPHigh 조건에서의 FDR 순위 기반 유전자 산점도(좌) 및 TRIM25를 표적하는 sgRNA들의 fold 농축도(우)이다.Figure 18 shows the FDR ranking-based gene scatter plot (left) and the fold enrichment of sgRNAs targeting TRIM25 (right) under GFPHigh conditions.
도 19는 HCT116 세포에서 TRIM25 또는 WDR77를 knockdown하였을 때, 비변형 EGFP LNP-mRNA로부터의 단백질 생성을 확인한 그래프이다.Figure 19 is a graph confirming protein production from unmodified EGFP LNP-mRNA when TRIM25 or WDR77 was knocked down in HCT116 cells.
도 20은 HCT116 WT 또는 TRIM25 KO-1, KO-2 세포에서 TRIM25의 웨스턴 블롯 분석 결과이다.Figure 20 shows the results of Western blot analysis of TRIM25 in HCT116 WT or TRIM25 KO-1, KO-2 cells.
도 21은 HCT116 WT 또는 TRIM25 KO-1, KO-2 세포에서 Fluc LNP-mRNA로부터의 루시퍼레이스 발현 및 GFP 발현을 확인한 그래프이다.Figure 21 is a graph confirming luciferase expression and GFP expression from Fluc LNP-mRNA in HCT116 WT or TRIM25 KO-1, KO-2 cells.
도 22는 HCT116 세포주에서 m1Ψ 및 U Fluc LNP-mRNA에 대한 상대적 루시퍼레이스 발현을 나타낸 그래프이다.Figure 22 is a graph showing relative luciferase expression for m1Ψ and U Fluc LNP-mRNA in HCT116 cell lines.
도 23은 일차 생쥐 BMDM 세포 및 다양한 인간 세포주(HeLa, THP-1, Jurkat, HEK293T)에서 Fluc LNP-mRNA에 의한 루시퍼레이스 발현을 측정한 그래프(상) 및 대표적 ISG 발현 수준(하)를 나타낸 그래프이다.Figure 23 is a graph showing the measurement of luciferase expression by Fluc LNP-mRNA in primary mouse BMDM cells and various human cell lines (HeLa, THP-1, Jurkat, HEK293T) (top) and representative ISG expression levels (bottom).
도 24는 5′ 말단이 uncapped, cap0, 또는 cap1 상태인 LNP-mRNA (상) 및 Fluc mRNA-2 또는 Rluc mRNA(하)로부터의 루시퍼레이스 발현을 측정한 그래프이다.Figure 24 is a graph measuring luciferase expression from LNP-mRNA (top) and Fluc mRNA-2 or Rluc mRNA (bottom) with the 5′ end in an uncapped, cap0, or cap1 state.
도 25는 HCT116 WT 또는 TRIM25 KO 세포에 Fluc LNP-mRNA를 형질감염한 뒤 수행한 폴리솜 프로파일링 및 리보솜별 분획을 수집하여 추출한 Fluc LNP-mRNA 분포를 RT-qPCR로 측정한 그래프이다.Figure 25 is a graph showing the distribution of Fluc LNP-mRNA extracted by collecting the polysome profiling and ribosome fractions after transfecting HCT116 WT or TRIM25 KO cells with Fluc LNP-mRNA, measured by RT-qPCR.
도 26은 Fluc LNP-mRNA의 세포 내 존재량을 RT-qPCR로 측정한 그래프이다.Figure 26 is a graph measuring the intracellular abundance of Fluc LNP-mRNA by RT-qPCR.
도 27은 Fluc LNP-mRNA 형질감염 후 pulse-chase 실험의 개요 및 단백질 발현량을 루시퍼레이스 분석으로, mRNA 존재량을 RT-qPCR로, poly(A) 길이 분포를 Hire-PAT 분석을 통해 측정한 그래프이다.Figure 27 is a graph outlining the pulse-chase experiment after Fluc LNP-mRNA transfection and measuring protein expression levels by luciferase analysis, mRNA abundance by RT-qPCR, and poly(A) length distribution by Hire-PAT analysis.
도 28은 RT-qPCR을 통해 측정한 LNP-mRNA의 존재량을 측정한 그래프이다. 순서대로 TRIM25가 결핍된 HCT116 GFP-stable 세포 또는 모세포주에서의 EGFP mRNA, HCT116 TRIM25 KO 세포에서의 Fluc mRNA, 및 GFP-stable 세포에서의 Fluc mRNA이다.Figure 28 is a graph measuring the abundance of LNP-mRNA by RT-qPCR. In order, EGFP mRNA in TRIM25-deficient HCT116 GFP-stable cells or parental cell line, Fluc mRNA in HCT116 TRIM25 KO cells, and Fluc mRNA in GFP-stable cells.
도 29는 Fluc 원형 RNA(circular RNA)의 개략도 및 이의 각 실험 단계에서의 완전성을 보여주는 자동 전기영동 결과이다.Figure 29 is a schematic diagram of Fluc circular RNA and the automated electrophoresis results showing its integrity at each experimental step.
도 30은 Fluc 원형 RNA를 LNP를 이용해 HCT116 WT 또는 TRIM25 KO 세포에 형질감염한 후의 ISG의 mRNA 수준, 루시퍼레이스 발현 및 이의 세포내 존재량을 qRT-PCR로 측정한 결과이다.Figure 30 shows the results of measuring the mRNA level of ISG, luciferase expression, and its intracellular abundance by qRT-PCR after transfecting HCT116 WT or TRIM25 KO cells with Fluc circular RNA using LNP.
도 31은 HCT116 WT 세포에서 exonuclease (XRN1) 및 exosome 구성 요소(DIS3, RRP41)를 knockdown한 조건에서의 LNP-mRNA 유래 루시퍼레이스 발현, mRNA 존재량, 및 각 유전자의 knockdown 효율을 RT-qPCR로 측정한 결과이다.Figure 31 shows the results of RT-qPCR measurement of LNP-mRNA-derived luciferase expression, mRNA abundance, and knockdown efficiency of each gene under conditions where exonuclease (XRN1) and exosome components (DIS3, RRP41) were knocked down in HCT116 WT cells.
도 32는 HCT116 WT 세포에서 세포질 선천면역 반응에 관여하는 endoribonuclease(RNASEL) 또는 엔도좀 내 RNA 분해에 관여하는 RNASET2/RNASE4 및 PLD3를 knockdown한 조건에서의 LNP-mRNA 유래 루시퍼레이스 발현, mRNA 존재량, 및 각 유전자의 knockdown 효율을 RT-qPCR로 측정한 결과이다.Figure 32 shows the results of measuring the expression of LNP-mRNA-derived luciferase, mRNA abundance, and knockdown efficiency of each gene by RT-qPCR under conditions in which endoribonuclease (RNASEL) involved in cytoplasmic innate immune response or RNASET2/RNASE4 and PLD3 involved in RNA degradation within endosomes were knocked down in HCT116 WT cells.
도 33은 비변형 IVT mRNA를 사용한 CRISPR KO 스크린에서의 유전자 수준 농축도(좌) 및 인간 TRIM25, N4BP1, KHNYN, ZAP 단백질의 도메인 구조(우)이다.Figure 33 shows gene-level enrichment (left) and domain structures (right) of human TRIM25, N4BP1, KHNYN, and ZAP proteins in a CRISPR KO screen using unmodified IVT mRNA.
도 34는 HCT116 WT 또는 TRIM25 KO 세포에서 N4BP1 siRNA 처리 후 KHNYN/ZAP knockdown과 함께 선형 Fluc LNP-mRNA로부터의 루시퍼레이스 발현을 측정한 그래프(상), N4BP1, KHNYN, ZAP의 knockdown 한 뒤 그 효율을 RT-qPCR로 측정한 결과(중) 및 LNP-mRNA의 존재량을 측정한 결과(하)이다.Figure 34 is a graph measuring luciferase expression from linear Fluc LNP-mRNA along with KHNYN/ZAP knockdown after N4BP1 siRNA treatment in HCT116 WT or TRIM25 KO cells (top), the results of measuring the efficiency of N4BP1, KHNYN, and ZAP knockdown by RT-qPCR (middle), and the results of measuring the abundance of LNP-mRNA (bottom).
도 35는 HCT116 WT 또는 TRIM25 KO 세포에서 N4BP1의 knockout을 확인한 western blot 결과 및 렌티바이러스 KHNYN/ZAP knockdown과 함께 선형 Fluc LNP-mRNA로부터의 루시퍼레이스 발현을 측정한 그래프이다.Figure 35 is a graph showing the results of western blot confirming the knockout of N4BP1 in HCT116 WT or TRIM25 KO cells and measuring luciferase expression from linear Fluc LNP-mRNA along with lentiviral KHNYN/ZAP knockdown.
도 36은 HCT116 WT 또는 TRIM25 KO 세포에서 N4BP1 siRNA 처리 또는 렌티바이러스 sgRNA에 의한 유전자 제거 후 KHNYN/ZAP knockdown과 함께 원형 RNA로부터의 루시퍼레이스 발현을 측정한 그래프이다.Figure 36 is a graph measuring luciferase expression from circular RNA along with KHNYN/ZAP knockdown after N4BP1 siRNA treatment or gene deletion by lentiviral sgRNA in HCT116 WT or TRIM25 KO cells.
도 37은 다양한 염기 변형을 가진 Fluc LNP-mRNA를 TRIM25 knockdown 조건에서 형질감염한 후의 루시퍼레이스 발현을 나타낸 그래프이다.Figure 37 is a graph showing luciferase expression after transfection of Fluc LNP-mRNA with various base modifications under TRIM25 knockdown conditions.
도 38은 TRIM25 RNA 면역침강(RNA-IP) 실험의 개략도 및 RT-qPCR을 통해 측정한 input 대비 상대적 농축도를 나타낸 그래프이다.Figure 38 is a schematic diagram of a TRIM25 RNA immunoprecipitation (RNA-IP) experiment and a graph showing the relative enrichment compared to input as measured by RT-qPCR.
도 39는 m1Ψ 또는 U가 포함된 poly(A)+ UTR-2 RNA 미끼를 oligo-dT 비드에 고정하여 수행한 RNA pulldown 실험 개요 및 내인성 또는 외인성 TRIM25 단백질에 대해 실험을 수행한 결과이다. Figure 39 shows an overview of RNA pulldown experiments performed by immobilizing poly(A)+ UTR-2 RNA bait containing m1Ψ or U onto oligo-dT beads and the results of experiments performed on endogenous or exogenous TRIM25 protein.
도 40는 정제된 재조합 인간 TRIM25 단백질과 poly(A)+ UTR-2 RNA 미끼를 사용한 in vitro RNA 결합 실험, SDS-PAGE 분석 및 크기배제 크로마토그래피 결과이다.Figure 40 shows the results of in vitro RNA binding experiments, SDS-PAGE analysis, and size exclusion chromatography using purified recombinant human TRIM25 protein and poly(A)+ UTR-2 RNA bait.
도 41는 UTR-1 RNA를 미끼로 사용한 RNA pulldown 및 western blot 실험 결과이다.Figure 41 shows the results of RNA pulldown and western blot experiments using UTR-1 RNA as bait.
도 42는 TRIM25 복원 실험 결과 그래프 및 외인성으로 발현된 TRIM25 변이체에 대한 western blot 분석 결과이다.Figure 42 is a graph showing the results of a TRIM25 restoration experiment and a western blot analysis of an exogenously expressed TRIM25 variant.
도 43는 면역침강된 TRIM25를 E3 리가아제로 사용한 in vitro 유비퀴틴화 실험의 개략도 및 Fluc IVT mRNA를 사용하여 TRIM25 WT, 7KA RNA 결합 돌연변이체, 및 R54P E3 돌연변이체를 비교한 in vitro 유비퀴틴화 실험 결과이다.Figure 43 is a schematic diagram of an in vitro ubiquitination experiment using immunoprecipitated TRIM25 as an E3 ligase and the results of an in vitro ubiquitination experiment comparing TRIM25 WT, 7KA RNA binding mutant, and R54P E3 mutant using Fluc IVT mRNA.
도 44는 GFP 안정 발현 세포에서 Fluc LNP-mRNA 형질감염 및 TRIM25 knockdown의 개요도 및 GFP(세포 내 mRNA) 및 루시퍼레이스(LNP-mRNA) 발현을 각각 유세포 분석과 루시퍼레이스 분석으로 측정한 그래프이다.Figure 44 is a schematic diagram of Fluc LNP-mRNA transfection and TRIM25 knockdown in GFP stable expressing cells, and a graph showing GFP (intracellular mRNA) and luciferase (LNP-mRNA) expression measured by flow cytometry and luciferase analysis, respectively.
도 45, 도 46 및 도 47는 HCT116 TRIM25 WT 또는 KO-1 세포에 Fluc mRNA를 형질감염한 후의 루시퍼레이스 발현, GFP 발현 또는 세포 내 존재량을 확인한 결과이다. 도 45는 Lipofectamine, 도 46은 전기천공, 및 도 47는 LNP-2를 통해 Fluc mRNA를 형질감염시켰다.Figures 45, 46, and 47 show the results of confirming luciferase expression, GFP expression, or intracellular abundance after transfection of Fluc mRNA into HCT116 TRIM25 WT or KO-1 cells. Figure 45 shows Fluc mRNA transfection via Lipofectamine, Figure 46 via electroporation, and Figure 47 via LNP-2.
도 48는 지정된 pH에서 30-nt poly(A)+ IVT RNA 미끼를 oligo-dT 비드에 고정한 RNA 품질 및 결합을 확인한 그래프이다.Figure 48 is a graph showing RNA quality and binding of 30-nt poly(A)+ IVT RNA bait immobilized on oligo-dT beads at a specified pH.
도 49는 HCT116 WT 세포 추출물을 이용해 지정된 pH에서 UTR-1 또는 UTR-2를 미끼로 하여 수행한 RNA pulldown 실험 및 그에 따른 western blot 결과이다.Figure 49 shows the RNA pulldown experiment performed at the indicated pH using HCT116 WT cell extracts with UTR-1 or UTR-2 as bait and the corresponding western blot results.
도 50는 CALM1 UTR을 미끼로 하여 수행한 RNA pulldown 실험 및 정제된 TRIM25 단백질을 사용한 in vitro RNA 결합 실험 결과이다.Figure 50 shows the results of an RNA pulldown experiment performed using CALM1 UTR as bait and an in vitro RNA binding experiment using purified TRIM25 protein.
도 51는 정제된 TRIM25 단백질을 사용하여 pH 간격 0.2 단위로 조절된 potassium phosphate buffer(pH별)에서 수행한 in vitro RNA 결합 실험 결과이다.Figure 51 shows the results of an in vitro RNA binding experiment performed in a potassium phosphate buffer (pH-wise) adjusted at pH intervals of 0.2 using purified TRIM25 protein.
도 52는 LNP-mRNA에 대한 세포 내 조절 기작의 제안 모델이다.Figure 52 is a proposed model of the intracellular regulatory mechanism for LNP-mRNA.
이하 실시예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실시예는 하나 이상의 구체예를 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.The following examples are provided for more detailed description. However, these examples are provided solely to illustrate one or more specific examples, and the scope of the present invention is not limited to these examples.
실시예 1. LNP-mRNA의 조절 인자를 확인하기 위한 CRISPR-Cas9 스크리닝Example 1. CRISPR-Cas9 screening to identify regulatory factors of LNP-mRNA.
외인성 mRNA의 조절 기전을 탐색하기 위하여, 단백질 코딩 유전자 19,114개를 대상으로 약 4개의 sgRNA씩 총 77,441개의 sgRNA로 구성된 렌티바이러스 기반의 전체 유전체 CRISPR-Cas9 KO 스크리닝을 수행하였다. sgRNA 라이브러리는 안정적인 이배체 핵형, 효율적인 LNP 전달이 가능한 빠르고 조직화된 엔도시토시스 경로, 외래 핵산에 대한 잘 규명된 인터페론(IFN) 반응을 특징으로 하는 인간 상피세포주인 HCT116에 도입하였다. 7일간의 푸로마이신 선별 후, EGFP를 암호화하는 IVT mRNA를 LNP를 통해 도입하였다(도 1). 도입한 mRNA는 5′ cap1 구조, 120-nt poly(A) 꼬리, 합성된 5′ UTR, 인간 알파글로빈 유래 3′ UTR을 포함하였으며, 이는 mRNA-1273 COVID-19 백신에서 사용된 UTR과 유사하다(도 2). 이러한 mRNA를 m1Ψ 변형이 포함된 형태와 포함되지 않은 형태의 두 가지로 제작하여, 염기 변형에 의존적인 조절 기전을 분석할 수 있도록 하였다. To explore the regulatory mechanisms of exogenous mRNA, we performed a lentiviral-based genome-wide CRISPR-Cas9 KO screen, targeting 19,114 protein-coding genes, comprising approximately 4 sgRNAs per sgRNA, for a total of 77,441 sgRNAs. The sgRNA library was introduced into HCT116, a human epithelial cell line characterized by a stable diploid karyotype, a rapid and organized endocytosis pathway capable of efficient LNP delivery, and a well-characterized interferon (IFN) response to exogenous nucleic acids. After 7 days of puromycin selection, IVT mRNA encoding EGFP was introduced via LNP (Fig. 1). The introduced mRNA contained a 5′ cap1 structure, a 120-nt poly(A) tail, a synthetic 5′ UTR, and a human alpha-globin-derived 3′ UTR, which are similar to the UTR used in the mRNA-1273 COVID-19 vaccine (Fig. 2). These mRNAs were produced in two forms, one with and one without the m1Ψ modification, to enable analysis of the regulatory mechanism dependent on base modification.
그 결과, 예상과 같이, m1Ψ가 포함된 mRNA는 변형되지 않은 mRNA에 비해 더 많은 단백질을 생성하였다(도 3).As a result, as expected, mRNA containing m1Ψ produced more protein than unmodified mRNA (Fig. 3).
상기 IVT mRNA를 BNT162b2 COVID-19 백신에서 사용된 것과 동일한 네 가지 지질 성분(ALC-0315, DSPC, 콜레스테롤, ALC-0159)으로 구성된 LNP에 캡슐화하였다. LNP 제형 및 트랜스펙션 조건은 트랜스펙션 효율의 세포 간 변이를 최소화하도록 최적화하였으며, 이를 통해 GFP 발현 기반 스크리닝이 가능하도록 하였다. 이때, 단일가닥 mRNA에 특이적인 조절 인자를 규명하고자 하였기 때문에 PKR, OAS, TLR3, RIG-I, MDA5 등을 통해 dsRNA 반응을 유도하여 비특이적으로 유전자 조절을 방해할 수 있는 IVT 반응 유래의 dsRNA 부산물을 줄였다. 전사 종결의 정확성을 보장하는 2′-O-메틸기가 말단 및 말단 직전 위치에 도입된 역방향 프라이머를 사용하여 PCR로 IVT DNA 주형을 제작하였다. The IVT mRNA was encapsulated in LNPs composed of the same four lipid components (ALC-0315, DSPC, cholesterol, and ALC-0159) used in the BNT162b2 COVID-19 vaccine. The LNP formulation and transfection conditions were optimized to minimize cell-to-cell variation in transfection efficiency, enabling GFP expression-based screening. Since we aimed to identify single-stranded mRNA-specific regulatory factors, we induced dsRNA responses through PKR, OAS, TLR3, RIG-I, and MDA5 to reduce dsRNA byproducts derived from the IVT reaction that could nonspecifically interfere with gene regulation. The IVT DNA template was constructed by PCR using a reverse primer with a 2′-O-methyl group introduced at the terminal and terminal positions to ensure accurate transcription termination.
그 결과, PKR knockdown은 대량의 dsRNA가 공동 형질감염될 때에만 IVT mRNA로부터의 유전자 발현에 영향을 주었다. LNP 실험에서는 Fluc IVT mRNA를 LNP에 봉입하여 기존 실험과 동일하게 형질감염하였다. Lipofectamine 실험에서는 말단 2'-O-methyl 그룹이 없는 템플릿 DNA(PCR 산물)로부터 Fluc mRNA를 전사하였으며, 이로 인해 dsRNA 부산물이 더 많이 생성되었다(도 4). 아울러, 예상과 같이, 생성된 RNA는 IFN 반응, PKR 인산화, 전반적인 번역 억제를 유도하지 않았다(도 5).As a result, PKR knockdown affected gene expression from IVT mRNA only when large amounts of dsRNA were co-transfected. In LNP experiments, Fluc IVT mRNA was encapsulated in LNPs and transfected in the same manner as in the previous experiments. In Lipofectamine experiments, Fluc mRNA was transcribed from template DNA (PCR product) lacking a terminal 2'-O-methyl group, resulting in increased production of dsRNA byproducts (Fig. 4). Furthermore, as expected, the generated RNA did not induce IFN responses, PKR phosphorylation, or overall translational inhibition (Fig. 5).
이후, 형광활성세포분석법(FACS)을 이용하여, 형광 신호가 가장 낮은(bottom 2.5%, GFPLow) 또는 가장 높은(top 2.5%, GFPHigh) 세포를 각각 분리하였다. 상관관계 분석을 확장하기 위해, 두 번째로 엄격한 기준에 해당하는 세포들(bottom 또는 top 2.55%, 각각 “2nd GFPLow” 또는 “2nd GFPHigh”로 명명)도 함께 수집하였다. 이후 PCR 증폭 및 시퀀싱을 통해 각 군의 sgRNA 분포를 분석하였다. 유의한 후보 유전자는 MAGeCK 알고리즘을 통해 선별하였다. Afterwards, cells with the lowest (bottom 2.5%, GFPLow) or highest (top 2.5%, GFPHigh) fluorescence signal were isolated using fluorescence-activated cell analysis (FACS). To extend the correlation analysis, cells meeting the second most stringent criteria (bottom or top 2.55%, designated “2nd GFPLow” or “2nd GFPHigh,” respectively) were also collected. The distribution of sgRNAs in each group was then analyzed through PCR amplification and sequencing. Significant candidate genes were selected using the MAGeCK algorithm.
그 결과, GFPLow 및 GFPHigh 모두에서 false discovery rate(FDR) < 0.05 기준의 유의미한 히트 유전자가 도출되었으며, sgRNA의 무결성 손실은 관찰되지 않았다(도 6).As a result, significant hit genes were derived with a false discovery rate (FDR) < 0.05 in both GFPLow and GFPHigh, and no loss of integrity of sgRNA was observed (Fig. 6).
이러한 IVT mRNA 기반 스크리닝 외에도, 내인성과 외인성 RNA 간의 조절 기전 차이를 확인하기 위해 GFP를 안정적으로 발현하는 세포주(“GFP-stable cell”)를 제작하여 대조 실험을 수행하였다. 이 세포주에서 생성되는 EGFP mRNA는 polyadenylation 신호로부터 유래된 추가 염기서열을 제외하면, IVT EGFP mRNA와 동일하다. In addition to these IVT mRNA-based screenings, we performed control experiments using a cell line stably expressing GFP (“GFP-stable cell”) to examine differences in regulatory mechanisms between endogenous and exogenous RNAs. EGFP mRNA produced in this cell line is identical to IVT EGFP mRNA, except for additional sequences derived from the polyadenylation signal.
이 GFP-stable 세포주를 대상으로 한 대조 스크리닝에서는 IVT mRNA를 트랜스펙션하지 않았음에도 전사 관련 유전자가 특이적으로 풍부하게 검출되었다. 이는 세포 유래 mRNA와 IVT mRNA 간의 요구 조건이 상이함을 의미한다(도 7).Control screening of this GFP-stable cell line revealed a specific enrichment of transcription-related genes even without transfection with IVT mRNA, suggesting different requirements for cell-derived mRNA and IVT mRNA (Fig. 7).
실시예 2. HSPG 및 V-ATPase의 역할 확인Example 2. Confirmation of the roles of HSPG and V-ATPase
(1) 양성 조절 인자 후보 확인(1) Identification of positive regulatory factor candidates
실시예 1의 IVT mRNA를 이용한 스크리닝에서, 헤파란 설페이트 프로테오 글리칸(heparan sulfate proteoglycan, HSPG) 생합성, 소낭 수송, V-ATPase 구성요소에 관여하는 유전자들을 포함하여 다수의 양성 조절 인자 후보들이 확인되었다(도 8). 이러한 유전자들은 변형되지 않은 mRNA와 m1Ψ 변형된 mRNA 스크리닝 모두에서 공통적으로 풍부하게 나타났으며, 이는 이들의 역할이 RNA 변형 여부와 무관함을 의미한다. Screening using IVT mRNA in Example 1 identified several positive regulatory factor candidates, including genes involved in heparan sulfate proteoglycan (HSPG) biosynthesis, vesicle transport, and V-ATPase components (Fig. 8). These genes were commonly enriched in both unmodified and m1Ψ-modified mRNA screens, suggesting that their roles are independent of RNA modification.
(2) HSPG 및 외인성 RNA 상호작용 확인(2) Confirmation of HSPG and exogenous RNA interaction
이전 연구[SLAS Discov 25, 605-617]에서 서로 다른 이온성 지질(MC3) 및 PEG화 지질(DMG-PEG 2000)을 포함하는 LNP 내 mo5U 변형 mRNA를 이용한 유전체 스크리닝에서 V-ATPase가 LNP 전달에 필수적이라고 보고된 바 있으나, HSPG 생합성 관련 단백질을 포함한 기타 인자들은 이전에 확인되지 않았다. 헤파란 설페이트(Heparan sulfate)는 세포 표면 및 세포외기질에 존재하는 고도로 산성화된 다당류로, 외부 리간드를 끌어당기고 이들의 세포 내 유입을 촉진한다. HSPG 생합성 인자들은 두 가지 엄격한 GFPLow 조건에서 강하게 농축되어 나타났다(도 9). Although a previous study [SLAS Discov 25, 605-617] reported that V-ATPase is essential for LNP delivery in a genome-wide screen using mo5U-modified mRNAs in LNPs containing different ionic lipids (MC3) and PEGylated lipids (DMG-PEG 2000), other factors, including proteins involved in HSPG biosynthesis, had not been previously identified. Heparan sulfate is a highly acidic polysaccharide present on the cell surface and in the extracellular matrix that attracts external ligands and promotes their endocytosis. HSPG biosynthesis factors were strongly enriched under two stringent GFPLow conditions (Fig. 9).
추가 검증을 위해, HCT116 세포에서 EXT1, EXT2, NDST1을 knockdown한 뒤, EGFP 또는 Firefly luciferase(Fluc)를 암호화하는 IVT mRNA를 도입하였다. 이들 인자를 제거하면, 염기 변형 여부와 관계없이 두 mRNA 모두에서 단백질 생성량이 감소하였다(도 10 및 도 11). 이러한 양상은 HEK293T 및 HeLa 세포에서도 동일하게 재현되었다(도 12).For further validation, we knocked down EXT1, EXT2, and NDST1 in HCT116 cells, and then introduced IVT mRNA encoding EGFP or Firefly luciferase (Fluc). Deletion of these factors resulted in a decrease in protein production from both mRNAs, regardless of the presence of base modifications (Figures 10 and 11). This pattern was also replicated in HEK293T and HeLa cells (Figure 12).
또한, 세포 표면의 HSPG와 경쟁하는 heparin을 처리하였을 때, GFP 신호 및 GFP 양성 세포의 비율이 농도 의존적으로 감소하였다. GFP-stable 세포에 Fluc mRNA를 트랜스펙션하였을 때, heparin은 GFP 발현에는 영향을 주지 않으면서 luciferase 발현만을 특이적으로 억제하였다. 이는 HSPG 관련 인자들이 외인성 mRNA에 특이적으로 작용함을 의미한다(도 13).Furthermore, when heparin, which competes with HSPGs on the cell surface, was treated, the GFP signal and the percentage of GFP-positive cells decreased in a concentration-dependent manner. When GFP-stable cells were transfected with Fluc mRNA, heparin specifically inhibited luciferase expression without affecting GFP expression. This indicates that HSPG-related factors act specifically on exogenous mRNA (Fig. 13).
(3) V-ATPase 및 외인성 RNA의 상호작용 확인(3) Confirmation of interaction between V-ATPase and exogenous RNA
다음으로, 엔도솜 내강을 산성화하는 ATP 구동성 양성자 펌프인 V-ATPase의 역할을 확인하였다. Next, we confirmed the role of V-ATPase, an ATP-driven proton pump that acidifies the lumen of the endosomes.
V-ATPase 서브유닛을 고갈시켰을 때, HCT116, HEK293T, HeLa 세포에서 IVT mRNA(EGFP 및 Fluc)로부터의 단백질 생성이 감소하였다(도 10 및 도 11). 또한, V-ATPase를 억제하는 Bafilomycin A1(BafA1)을 처리하였을 때도 IVT mRNA로부터의 GFP 생성이 줄어들었으며, GFP-stable 세포에 Fluc mRNA를 트랜스펙션한 상태에서 BafA1을 처리하였을 경우, BafA1은 외인성 mRNA에 선택적으로 작용하여 luciferase 발현만을 억제하고 GFP에는 영향을 주지 않았다. 또한, clathrin과 dynamin I/II를 각각 표적으로 하는 내포 억제제인 Pitstop-2와 Dynole 또한 LNP-mRNA의 발현을 억제하였다(도 13).Depletion of V-ATPase subunits decreased protein production from IVT mRNA (EGFP and Fluc) in HCT116, HEK293T, and HeLa cells (Figs. 10 and 11). Furthermore, treatment with Bafilomycin A1 (BafA1), which inhibits V-ATPase, also reduced GFP production from IVT mRNA. Furthermore, when GFP-stable cells were transfected with Fluc mRNA and treated with BafA1, BafA1 selectively inhibited exogenous mRNA, suppressing only luciferase expression without affecting GFP. Furthermore, Pitstop-2 and Dynole, which are endocytic inhibitors targeting clathrin and dynamin I/II, respectively, also suppressed LNP-mRNA expression (Fig. 13).
(4) 세포 내 LNP 전달 경로에서 HSPG 및 V-ATPse의 역할 확인(4) Confirmation of the roles of HSPG and V-ATPse in the intracellular LNP delivery pathway
RNA 분석 결과, heparin은 IVT mRNA의 세포 내 존재량을 감소시킨 반면, BafA1은 mRNA 양을 증가시켰다(도 15). 이는 heparin이 LNP의 세포 표면 부착을 방해하여 초기 유입을 저해하는 반면, BafA1은 엔도솜 탈출을 방해하여 mRNA가 엔도솜 내에 축적되게 함을 의미한다.RNA analysis revealed that heparin reduced the intracellular abundance of IVT mRNA, whereas BafA1 increased mRNA levels (Fig. 15). This suggests that heparin inhibits initial uptake by interfering with LNP attachment to the cell surface, whereas BafA1 inhibits endosomal escape, leading to mRNA accumulation within endosomes.
고해상도 poly(A) tail 분석법(Hire-PAT)을 이용한 후속 분석에서는, LNP-mRNA를 24시간 처리한 후 트랜스펙션된 mRNA의 상당수가 deadenylation을 거쳐 poly(A) 길이가 30~50 nt의 분포를 보이는 것으로 나타났다. 이는 IVT mRNA가 세포질로 진입하여 deadenylation에 노출되었음을 의미한다. heparin을 처리한 경우, 전체적인 신호 강도는 감소하였지만 poly(A) 분포는 변화하지 않았다. 이는 heparin이 mRNA 유입은 억제하였지만 이후 단계에는 영향을 미치지 않았음을 의미한다. 반면, BafA1 처리군에서는 대부분의 트랜스펙션된 mRNA가 원래의 poly(A) 길이(~120 nt)를 유지하고 있었으며, 이는 해당 mRNA가 세포질 내 deadenylase에 도달하지 못했음을 보여준다. 대조군인 GFP-stable 세포에서는 heparin과 BafA1 모두 GFP mRNA에 영향을 주지 않았다(도 16).Subsequent analysis using high-resolution poly(A) tail analysis (Hire-PAT) revealed that a significant proportion of transfected mRNAs underwent deadenylation after 24 h of LNP-mRNA treatment, resulting in a poly(A) length distribution of 30–50 nt. This suggests that IVT mRNA enters the cytoplasm and is exposed to deadenylation. When heparin was treated, the overall signal intensity decreased but the poly(A) distribution did not change. This suggests that heparin inhibited mRNA entry but did not affect subsequent steps. In contrast, in the BafA1-treated group, most transfected mRNAs retained their original poly(A) length (~120 nt), indicating that the mRNAs did not reach the cytoplasmic deadenylase. In the control GFP-stable cells, neither heparin nor BafA1 affected GFP mRNA (Fig. 16).
추가적으로, mRNA-1273 COVID-19 백신에 사용된 이온성 지질 SM-102와 PEG화 지질 DMG-PEG 2000으로 구성된 또 다른 유형의 LNP(“LNP-2”)를 사용하여 실험을 진행하였다. Additionally, experiments were conducted using another type of LNP (“LNP-2”) consisting of the ionic lipid SM-102 used in the mRNA-1273 COVID-19 vaccine and the PEGylated lipid DMG-PEG 2000.
그 결과, heparin과 BafA1 모두 LNP-2-mRNA의 발현을 강하게 억제하였으며, 이는 HSPG와 V-ATPase가 LNP 기반 전달 전반에 필수적임을 나타낸다(도 17).As a result, both heparin and BafA1 strongly inhibited the expression of LNP-2-mRNA, indicating that HSPG and V-ATPase are essential for the overall LNP-based delivery (Fig. 17).
실시예 3. TRIM25의 외인성 RNA 억제 확인Example 3. Confirmation of exogenous RNA suppression of TRIM25
(1) 음성 조절 인자 후보 확인(1) Identification of candidate voice modulators
본 스크리닝을 통해 네 개의 음성 조절 인자 후보(GFPHigh, FDR < 0.05)를 확인하였으며, 이들은 TRIM25, WDR77, HNRNPH1, THOC2였다. 이 중 TRIM25는 변형되지 않은 mRNA 스크리닝에서 가장 낮은 FDR 값을 나타내며 가장 강하게 농축되었고, GFP-stable 세포나 m1Ψ 변형 mRNA 스크리닝에서는 농축되지 않았다(도 18). 이는 TRIM25가 변형되지 않은 IVT mRNA를 선택적으로 억제함을 시사한다. TRIM25는 RNA 결합 단백질이자 E3 리가아제로, RNA 바이러스에 대한 항바이러스 활성이 있는 것으로 알려져 있다. 또한 TRIM25는 RIG-I, ZAP과 같은 면역 센서 및 G3BP 단백질과도 상호작용하는 것으로 알려져 있다.This screening identified four negative regulator candidates (GFPHigh, FDR < 0.05), namely TRIM25, WDR77, HNRNPH1, and THOC2. Among them, TRIM25 was the most strongly enriched with the lowest FDR value in the unmodified mRNA screening, but was not enriched in the GFP-stable cells or m1Ψ-modified mRNA screening (Fig. 18). This suggests that TRIM25 selectively represses unmodified IVT mRNA. TRIM25 is an RNA-binding protein and E3 ligase known to have antiviral activity against RNA viruses. TRIM25 is also known to interact with immune sensors such as RIG-I and ZAP, as well as the G3BP protein.
이를 검증하기 위해, 핵 이력을 거치지 않고 세포질에서 작용하는 IVT mRNA의 특성을 고려하여, 세포질 내 존재하는 후보인 TRIM25 및 WDR77을 knockdown하였다. To verify this, considering the characteristics of IVT mRNA that acts in the cytoplasm without going through nuclear history, TRIM25 and WDR77, which are candidates existing in the cytoplasm, were knocked down.
그 결과, TRIM25를 고갈시키면 변형되지 않은 mRNA로부터의 GFP 발현이 증가하였으며, WDR77 knockdown은 유의한 영향을 나타내지 않았다(도 19).As a result, depletion of TRIM25 increased GFP expression from unmodified mRNA, while WDR77 knockdown had no significant effect (Fig. 19).
추가적으로, 서로 다른 sgRNA를 사용하여 TRIM25 KO 세포주(TRIM25 KO-1 및 KO-2)를 두 종류 제작하였다(도 20). 이들 KO 세포에서는 변형되지 않은 mRNA가 모세포주에 비해 더 많은 단백질을 생성하였다(도 21). 특히, TRIM25 KO 세포에서 변형되지 않은 mRNA가 m1Ψ 변형 mRNA에 필적하는 수준의 luciferase가 생성되었다. 이는 TRIM25가 변형되지 않은 mRNA에 대한 핵심 억제 인자임을 보여준다(도 22).Additionally, two TRIM25 knockout (KO) cell lines (TRIM25 KO-1 and KO-2) were generated using different sgRNAs (Fig. 20). In these KO cells, unmodified mRNA produced more protein than the parental cell line (Fig. 21). Notably, in TRIM25 KO cells, unmodified mRNA produced luciferase at levels comparable to those of m1Ψ-modified mRNA. This demonstrates that TRIM25 is a key repressor of unmodified mRNA (Fig. 22).
(2) TRIM25의 세포별 활성 확인(2) Confirmation of TRIM25 activity in each cell
TRIM25의 활성을 다양한 세포 유형에서 탐색한 결과, HeLa(인간 상피세포), THP-1(인간 단핵구), Jurkat(인간 T 림프구), HEK293T(인간 배아 신장세포)에서 TRIM25를 고갈시켰다. 추가로 생쥐 유래 1차 세포인 골수 유래 대식세포(mBMDM)에서도 동일한 실험을 수행하였으며, HCT116 세포에서와 마찬가지로, TRIM25 고갈은 HeLa, THP-1, Jurkat, mBMDM 세포에서 변형되지 않은 mRNA의 발현을 유의하게 증가시켰으나, HEK293T 세포에서는 이러한 효과가 관찰되지 않았다. TRIM25에 의해 매개되는 감시 기작은 세포 유형에 따라 다소 차이를 보이긴 하지만, 상기 기작은 보편적이고 진화적으로 보존된 기전임을 확인하였다(도 23).To investigate the activity of TRIM25 in various cell types, we depleted TRIM25 in HeLa (human epithelial cells), THP-1 (human monocytes), Jurkat (human T lymphocytes), and HEK293T (human embryonic kidney cells). In addition, the same experiment was performed in bone marrow-derived macrophages (mBMDM), a primary cell line derived from mice. Similar to HCT116 cells, TRIM25 depletion significantly increased the expression of intact mRNA in HeLa, THP-1, Jurkat, and mBMDM cells, but not in HEK293T cells. Although the surveillance mechanism mediated by TRIM25 shows some differences depending on the cell type, it was confirmed that this mechanism is universal and evolutionarily conserved (Fig. 23).
(3) TRIM25의 RNA 분자적 특성별 활성 확인(3) Confirmation of TRIM25 activity by RNA molecular characteristics
TRIM25의 특이성을 보다 면밀히 분석하기 위해, 다양한 분자적 특성을 지닌 IVT mRNA를 테스트하였다. To further analyze the specificity of TRIM25, we tested IVT mRNAs with different molecular characteristics.
먼저, cap 구조가 서로 다른 mRNA를 비교하였다. cap0(N7-methylguanosine, m7G) 및 cap1(m7G에 +1 위치의 2′-O-methylation이 추가된 구조)을 가진 mRNA는 각기 다른 캡핑 효율을 가지며, 이를 통해 5′ 말단 구조의 영향을 분석하였다. 그 결과, TRIM25 고갈은 캡 구조와 무관하게 luciferase 발현을 유의하게 증가시켰으며, 이는 TRIM25의 작용이 5′ 말단 구조와 독립적임을 의미한다.First, we compared mRNAs with different cap structures. mRNAs with cap0 (N7-methylguanosine, m7G) and cap1 (m7G plus 2′-O-methylation at the +1 position) exhibited different capping efficiencies, allowing us to analyze the impact of the 5′-terminal structure. As a result, TRIM25 depletion significantly increased luciferase expression regardless of the cap structure, indicating that TRIM25's action is independent of the 5′-terminal structure.
추가적으로, 다양한 mRNA 서열을 가진 경우를 테스트하였다. Fluc 암호화 mRNA의 UTR을 변경한 변형체(Fluc-2) 및 poly(A) 꼬리 길이가 짧은(60 nt) Renilla luciferase(Rluc) 암호화 mRNA를 사용하였다. 모든 경우에서 TRIM25 KO 세포에서는 luciferase 발현이 증가하였으며, 이는 TRIM25 매개 억제가 mRNA 서열이나 poly(A) 꼬리 길이에 의존하지 않음을 보여준다(도 24).Additionally, we tested cases with various mRNA sequences. We used a variant of the Fluc-encoding mRNA with altered UTR (Fluc-2) and a Renilla luciferase (Rluc) mRNA with a short poly(A) tail (60 nt). In all cases, luciferase expression was increased in TRIM25 KO cells, demonstrating that TRIM25-mediated repression is independent of mRNA sequence or poly(A) tail length (Fig. 24).
(4) TRIM25의 억제 기전 확인(4) Confirmation of TRIM25 inhibition mechanism
TRIM25가 IVT mRNA를 어떻게 억제하는지를 이해하기 위해, 먼저 폴리솜 프로파일링을 이용하여 해당 mRNA들의 번역 상태를 분석하였다. 폴리솜 프로파일링은 설탕 구배 상에서 리보솜 결합 수에 따라 번역 중인 mRNA들을 분리하는 방법이다. To understand how TRIM25 represses IVT mRNA, we first analyzed the translational status of these mRNAs using polysome profiling. Polysome profiling is a method for separating mRNAs undergoing translation based on the number of ribosomes bound on a sugar gradient.
그 결과, TRIM25 결손이나 염기 변형은 전체적인 폴리솜 분포에 유의미한 영향을 주지 않았으며, 이는 TRIM25가 전반적인 번역(global translation)에 영향을 미치지 않음을 나타낸다. 또한, RT-qPCR을 통해 구배 분획별 IVT mRNA의 양을 정량하였을 때에도, TRIM25 결손에 따른 뚜렷한 변화는 관찰되지 않았다(도 25). 즉, TRIM25가 번역을 조절할 가능성을 완전히 배제할 수는 없지만, 이는 TRIM25 작용의 주요 기전은 아닐 가능성이 높다.As a result, TRIM25 deletion or nucleotide modification did not significantly affect overall polysome distribution, indicating that TRIM25 does not affect global translation. Furthermore, when the amount of IVT mRNA in each gradient fraction was quantified using RT-qPCR, no significant changes were observed due to TRIM25 deletion (Fig. 25). Thus, while the possibility that TRIM25 regulates translation cannot be completely ruled out, this is unlikely to be the primary mechanism of TRIM25 action.
다음으로, RT-qPCR을 이용하여 TRIM25가 RNA 양에 미치는 영향을 분석하였다. Next, the effect of TRIM25 on RNA amount was analyzed using RT-qPCR.
그 결과 TRIM25가 결핍된 세포에서는 비변형 IVT mRNA의 수준이 급격히 증가하는 것으로 나타났다. 변형된 mRNA는 소폭 증가하는 데 그쳤다. 반면, 엔도솜 탈출을 저해하는 BafA1로 처리한 경우에는 어떠한 유의미한 증가도 관찰되지 않았다. 이는 TRIM25가 mRNA가 세포질로 진입한 이후 주로 mRNA 수준을 감소시키는 방식으로 작용함을 나타낸다(도 26).As a result, TRIM25-deficient cells showed a dramatic increase in the level of unmodified IVT mRNA. The level of modified mRNA increased only slightly. In contrast, no significant increase was observed when treated with BafA1, which inhibits endosomal escape. This indicates that TRIM25 primarily acts by reducing mRNA levels after mRNA enters the cytoplasm (Fig. 26).
또한, LNP-mRNA를 세포에 2시간 동안만 처리하고 세척 후 시간 경과에 따른 변화를 추적하는 타임코스 실험을 수행하였다. 2시간 처리 후 세척 직후에도, 비변형 mRNA의 루시퍼레이스 발현량과 RNA 수준은 이미 m1Ψ 변형 mRNA보다 낮았으며, 이러한 억제는 TRIM25 결손 세포에서는 완화되었다. 이는 TRIM25가 매우 빠른 억제 기전을 작동시킴을 의미한다(도 27). TRIM25 knockdown 또는 결손 세포에서는 EGFP 및 Fluc mRNA 양이 모두 증가하는 현상도 관찰되었다. 이러한 결과는 TRIM25가 mRNA 분해를 촉진한다는 점을 시사한다(도 28).Additionally, a time-course experiment was performed, in which cells were treated with LNP-mRNA for only 2 hours and then washed, followed by tracking changes over time. Even immediately after washing after 2 hours of treatment, luciferase expression and RNA levels of unmodified mRNA were already lower than those of m1Ψ-modified mRNA, and this suppression was alleviated in TRIM25-deficient cells. This indicates that TRIM25 activates a very rapid repression mechanism (Fig. 27). Increases in both EGFP and Fluc mRNA levels were also observed in TRIM25 knockdown or deficient cells. These results suggest that TRIM25 promotes mRNA degradation (Fig. 28).
일반적으로 deadenylation은 mRNA 분해 과정에서 속도 결정 단계로 간주되므로, 2시간 트랜스펙션 및 세척 후 poly(A) tail 길이도 분석하였다. 그 결과, TRIM25 결손은 deadenylation 속도에 유의미한 영향을 미치지 않았으며, 이는 TRIM25가 deadenylation을 가속화하는 방식으로 작용하지는 않는 것을 의미한다(도 27).Since deadenylation is generally considered a rate-determining step in mRNA degradation, we also analyzed the poly(A) tail length after 2 h of transfection and washing. As a result, TRIM25 deletion did not significantly affect the deadenylation rate, indicating that TRIM25 does not act to accelerate deadenylation (Fig. 27).
실시예 4. TRIM25 보조인자로서 N4BP1, KHNYN, 및 ZAP의 확인Example 4. Identification of N4BP1, KHNYN, and ZAP as TRIM25 cofactors.
TRIM25가 endonucleolytic cleavage를 유도하는지를 확인하기 위해 5′ cap과 poly(A) tail 구조가 없는 circular RNA(circRNA)를 사용하여 실험을 수행하였다(도 29). m1Ψ는 원형화 반응과 IRES 활성을 방해하기 때문에, m1Ψ 변형이 도입되지 않은 circRNA를 제작하였다. To determine whether TRIM25 induces endonucleolytic cleavage, we performed experiments using circular RNA (circRNA) lacking a 5′ cap and poly(A) tail (Fig. 29). Because m1Ψ interferes with circularization and IRES activity, we constructed a circRNA without the m1Ψ modification.
그 결과, circRNA는 IFNβ 및 ISG의 발현을 유의미하게 유도하지 않았다. 그러나, TRIM25가 결핍된 조건에서는 circRNA로부터 유래한 루시퍼레이스 발현이 현저히 증가하였다. RNA 수준 역시 TRIM25 결핍 세포에서 대조군보다 더 높게 나타났다. 아울러, 비변형 linear mRNA와 마찬가지로, circRNA 역시 BafA1로 처리한 세포에서는 축적되지 않았다(도 30). 이러한 결과는 TRIM25가 linear mRNA뿐만 아니라 circular RNA도 억제하며, 이 과정에서 exonuclease에 의존하지 않음을 시사한다. 실제로, 5′→3′ exonuclease인 XRN1, 3′→5′ exosome 소단위체인 EXOSC4(RRP41) 및 DIS3(RRP44)를 knockdown 하였을 때에도 유의미한 탈억제 효과는 관찰되지 않았다(도 31).As a result, circRNAs did not significantly induce the expression of IFNβ and ISGs. However, under TRIM25-deficient conditions, circRNA-derived luciferase expression was significantly increased. RNA levels were also higher in TRIM25-deficient cells than in the control. In addition, similar to unmodified linear mRNA, circRNAs did not accumulate in cells treated with BafA1 (Fig. 30). These results suggest that TRIM25 represses not only linear mRNA but also circular RNA, and that this process is independent of exonuclease. In fact, no significant derepression effect was observed when XRN1, a 5′→3′ exonuclease, and EXOSC4 (RRP41) and DIS3 (RRP44), 3′→5′ exosome subunits, were knocked down (Fig. 31).
이러한 결과는 endoribonuclease의 관여 가능성을 강하게 시사한다. 추가적으로, 후보 유전자 접근법을 통해 세포질 선천면역 반응 및 엔도라이소좀 내 RNA 분해에 관여하는 것으로 알려진 일부 RNase들을 제거하였다. 여기에는 endoribonuclease인 RNase L, RNase T2, RNase 4와 함께, 라이소좀의 5′→3′ exonuclease인 PLD3가 포함된다. 그러나 이들에 대한 전장 유전체 스크리닝 및 knockdown 실험에서는 이들의 관여를 뒷받침하는 근거가 확인되지는 않았다(도 32).These results strongly suggest the involvement of endoribonucleases. Additionally, a candidate gene approach was used to eliminate several RNases known to be involved in the cytosolic innate immune response and RNA degradation within endolysosomes. These included the endoribonucleases RNase L, RNase T2, and RNase 4, as well as PLD3, a lysosomal 5′→3′ exonuclease. However, genome-wide screening and knockdown experiments did not confirm their involvement (Fig. 32).
TRIM25 매개 mRNA 분해에 관여하는 RNase를 확인하기 위해, knockout 스크리닝 데이터로 다시 돌아가 통계적 기준에는 미치지 못했지만 발현이 증가한 유전자들을 검토하였다. 그 결과, 비변형 IVT mRNA를 대상으로 한 스크리닝에서 log2FC 0.99, FDR 0.12로 7번째로 높은 순위를 기록한 endoribonuclease인 N4BP1(NEDD4-binding protein 1)을 확인하였다(도 33). N4BP1은 NYN 도메인(endoribonuclease 도메인)과 KH-like 도메인(추정 RNA 결합 도메인), 그리고 유비퀴틴 결합 도메인인 UBA 및 CoCUN 도메인을 포함하고 있다. N4BP1에는 KHNYN이라는 잠재적 paralog가 존재하며, 이 유전자는 비변형 mRNA를 대상으로 한 스크리닝에서 4027번째로 순위가 낮았다. KHNYN은 TRIM25 및 ZAP과 상호작용하며 HIV-1 복제를 억제하는 것으로 알려져 있다.To identify the RNase involved in TRIM25-mediated mRNA degradation, we revisited the knockout screening data and reviewed genes whose expression was increased but did not meet the statistical criteria. We identified N4BP1 (NEDD4-binding protein 1), an endoribonuclease that ranked seventh in the screening of unmodified IVT mRNA with a log2FC of 0.99 and an FDR of 0.12 (Figure 33). N4BP1 contains a NYN domain (endoribonuclease domain), a KH-like domain (putative RNA-binding domain), and ubiquitin-binding domains, UBA and CoCUN. N4BP1 has a potential paralog, KHNYN, which ranked 4027th in the screening of unmodified mRNA. KHNYN interacts with TRIM25 and ZAP and is known to inhibit HIV-1 replication.
N4BP1 또는 KHNYN 각각을 개별적으로 knockdown 하였을 때에는 뚜렷한 효과가 나타나지 않았으나, 두 유전자를 동시에 제거하였을 경우 비변형 mRNA의 단백질 발현량과 RNA 양이 모두 유의미하게 증가하였다. 이는 이들이 해당 감시 기전에서 기능이 중복되는 역할을 한다는 점을 시사한다. TRIM25와 KHNYN 모두 ZAP과 상호작용하는 것으로 알려져 있는 바 ZAP 또한 함께 제거하였으며, 그 결과 루시퍼레이스 발현이 추가적으로 증가하였다. 반면, TRIM25가 결손된 조건에서는 N4BP1, KHNYN, ZAP의 knockdown이 어떠한 효과도 나타내지 않았다 (도 34). 이 결과는 이들 인자들이 TRIM25에 결정적으로 의존하고 있음을 나타낸다.When N4BP1 or KHNYN was knocked down individually, no significant effect was observed. However, when both genes were knocked down simultaneously, both the protein expression level of the intact mRNA and the RNA level significantly increased. This suggests that they play overlapping roles in this surveillance mechanism. Since both TRIM25 and KHNYN are known to interact with ZAP, ZAP was also knocked down, resulting in a further increase in luciferase expression. In contrast, under TRIM25-deficient conditions, knockdown of N4BP1, KHNYN, and ZAP had no effect (Fig. 34). These results indicate that these factors are critically dependent on TRIM25.
추가 검증을 위해, N4BP1 knockout 세포를 생성하였다. N4BP1 단일 결손은 루시퍼레이스 발현의 완만하지만 유의미한 증가를 초래하였으며, 여기에 KHNYN 및 ZAP을 siRNA로 추가 제거하였을 때 억제 효과가 더욱 완화되었다. 반면, TRIM25 결손 세포에서는 어떠한 영향도 나타나지 않았다(도 35). 이러한 조절 효과는 변형된 mRNA보다 비변형 mRNA에서 훨씬 더 강하게 나타났다. For further validation, N4BP1 knockout cells were generated. Single deletion of N4BP1 resulted in a modest but significant increase in luciferase expression, and the inhibitory effect was further alleviated when KHNYN and ZAP were additionally knocked down with siRNA. In contrast, no effect was observed in TRIM25-deficient cells (Fig. 35). This regulatory effect was much stronger with unmodified mRNA than with modified mRNA.
더 나아가, 원형 RNA(circRNA) 역시 N4BP1/KHNYN/ZAP이 제거된 세포에서 TRIM25 의존적으로 탈억제되었다(도 36). 이 모든 결과를 종합하면, N4BP1, KHNYN, ZAP은 TRIM25 매개 외인성 RNA 감시 기전에서 중복적으로 작용하며, 그 기능은 RNA 변형 여부에 따라 조절됨을 의미한다.Furthermore, circular RNAs (circRNAs) were also derepressed in a TRIM25-dependent manner in N4BP1/KHNYN/ZAP-depleted cells (Fig. 36). Taken together, these results suggest that N4BP1, KHNYN, and ZAP function redundantly in TRIM25-mediated exogenous RNA surveillance, and their functions are regulated by RNA modification.
실시예 5. 변형 RNA의 TRIM25 결합 및 활성화 회피 확인Example 5. Confirmation of TRIM25 binding and activation avoidance by modified RNA.
(1) TRIM25 매개 억제에서 염기 변형의 영향 확인(1) Confirmation of the influence of base modification on TRIM25-mediated inhibition
TRIM25 매개 억제에 대한 염기 변형의 영향을 평가하기 위해, Ψ(pseudouridine), mo5U(5-methoxyuridine), m5C(5-methylcytidine)로 변형된 linear mRNA들을 비교하였다(도 37). To assess the effect of base modifications on TRIM25-mediated repression, linear mRNAs modified with Ψ (pseudouridine), mo5U (5-methoxyuridine), and m5C (5-methylcytidine) were compared (Fig. 37).
m1Ψ 변형 mRNA의 경우와 유사하게, Ψ 변형 mRNA는 TRIM25 knockdown 시 발현 증가가 미미하였다. 이는 m1Ψ 및 Ψ 변형 mRNA 모두가 TRIM25를 어느 정도 회피함을 시사하지만, 완전히 회피하지는 못함을 나타낸다. 반면, mo5U 및 m5C 변형 mRNA는 중간 정도 수준의 발현 증가를 나타내었다. 이는 TRIM25가 여전히 이들 mRNA를 타겟팅할 수 있으며, mo5U와 m5C 변형은 TRIM25로부터 부분적인 보호만을 제공함을 의미한다.Similar to the case of m1Ψ variant mRNA, Ψ variant mRNA showed only a minimal increase in expression upon TRIM25 knockdown. This suggests that both m1Ψ and Ψ variant mRNAs evade TRIM25 to some extent, but not completely. In contrast, mo5U and m5C variant mRNAs showed a moderate increase in expression. This suggests that TRIM25 can still target these mRNAs, and that mo5U and m5C variants provide only partial protection from TRIM25.
TRIM25는 RNA와 직접 상호작용하는 것으로 알려져 있으며, RNA 결합에 결함이 있는 TRIM25 돌연변이체는 항바이러스 활성이 결여되어 있다. 따라서 TRIM25가 비변형 mRNA를 차별적으로 인식하고 RNA와의 상호작용을 통해 이를 조절할 수 있을 것이라 가설을 세웠다. 이를 검증하기 위해, LNP329 mRNA를 HCT116 세포에 형질도입한 후, RNA 면역침강 및 RT-qPCR(RIP-qPCR)을 수행하였다(도 38) TRIM25 is known to interact directly with RNA, and TRIM25 mutants defective in RNA binding lack antiviral activity. Therefore, we hypothesized that TRIM25 might differentially recognize unmodified mRNA and regulate it through its interaction with RNA. To verify this, we transfected HCT116 cells with LNP329 mRNA, followed by RNA immunoprecipitation and RT-qPCR (RIP-qPCR) (Figure 38).
그 결과, 내인성 비번역 RNA(18S rRNA), 단백질 암호화 전사체(GAPDH), 또는 m1Ψ 변형 IVT mRNA와는 달리, 비변형 IVT mRNA는 TRIM25 면역침강물에서 강하게 농축되었다. 따라서 TRIM25는 염기 변형이 없는 외인성 RNA와 선호적으로 상호작용하는 것으로 나타났다.As a result, unlike endogenous noncoding RNA (18S rRNA), protein-coding transcript (GAPDH), or m1Ψ-modified IVT mRNA, unmodified IVT mRNA was strongly enriched in TRIM25 immunoprecipitates. Thus, TRIM25 appears to preferentially interact with exogenous RNA without base modifications.
TRIM25의 RNA 결합 특이성은 RNA336 중심의 풀다운 실험을 통해 추가로 입증되었다(도 39). 단일가닥 RNA는 도 24에 제시된 UTR-1 또는 UTR-2의 3′ UTR 말단 100-nt 서열에 30-nt 길이의 poly(A)를 추가하여 구성하였고, 이를 oligo-dT 비드에 고정한 후 HCT116 세포 용해물과 반응시켰다. The RNA-binding specificity of TRIM25 was further demonstrated through pull-down experiments centered on RNA336 (Fig. 39). Single-stranded RNA was constructed by adding a 30-nt poly(A) fragment to the 100-nt sequence at the 3′ UTR of UTR-1 or UTR-2 shown in Fig. 24. This was immobilized on oligo-dT beads and reacted with HCT116 cell lysate.
웨스턴 블롯 결과, TRIM25는 m1Ψ 변형 RNA에 비해 비변형 RNA bait에 선호적으로 결합하는 것으로 나타났으며, 이는 상호작용의 특이성을 뒷받침한다.Western blot results showed that TRIM25 binds preferentially to unmodified RNA bait over m1Ψ-modified RNA, supporting the specificity of the interaction.
TRIM25가 RNA와 직접 상호작용하는지를 확인하기 위해, 재조합 TRIM25 단백질을 정제하였다. 그 결과, 비변형 RNA는 TRIM25 단백질을 효과적으로 풀다운하였으며(도 40), 이는 TRIM25와 RNA 간의 직접적인 상호작용을 의미한다. 반면, m1Ψ 변형 RNA는 TRIM25를 유의미하게 침전시키지 못하였으며, 이는 m1Ψ 변형이 TRIM25의 RNA 상호작용을 저해함을 의미한다.To determine whether TRIM25 directly interacts with RNA, we purified recombinant TRIM25 protein. As a result, unmodified RNA effectively pulled down TRIM25 protein (Figure 40), indicating a direct interaction between TRIM25 and RNA. In contrast, m1Ψ-modified RNA did not significantly precipitate TRIM25, indicating that the m1Ψ modification inhibits TRIM25's interaction with RNA.
(2) RNA-TRIM25 상호작용에서 7K 모티프의 역할 확인(2) Confirmation of the role of the 7K motif in RNA-TRIM25 interaction
TRIM25는 RING 도메인, B-box, coiled-coil 도메인, 그리고 양전하를 띠는 linker 및 그 뒤를 잇는 PRY/SPRY 도메인으로 구성되어 있다. PRY/SPRY 도메인과 함께, linker 내에 존재하는 7개의 라이신 서열(381KKVSKEEKKSKK392, "7K 모티프)은 RNA 결합에 필수적이다. 이 7K 모티프의 라이신 잔기들을 알라닌으로 치환한 돌연변이체("7KA")는 RNA에 대한 TRIM25의 친화도를 감소시키는 것으로 알려져 있다. 실제로, 야생형 TRIM25와 달리 7KA 돌연변이체는 RNA bait와 거의 풀다운되지 않았으며, 이는 RNA-TRIM25 상호작용에서 7K 모티프가 중요함을 의미한다(도 41).TRIM25 is composed of a RING domain, a B-box, a coiled-coil domain, a positively charged linker, and a PRY/SPRY domain. In addition to the PRY/SPRY domain, a seven-lysine sequence (381KKVSKEEKKSKK392, the "7K motif") within the linker is essential for RNA binding. A mutant in which the lysine residues in this 7K motif are substituted with alanines ("7KA") is known to reduce the affinity of TRIM25 for RNA. In fact, unlike wild-type TRIM25, the 7KA mutant showed little pull-down with RNA bait, implying the importance of the 7K motif in the RNA-TRIM25 interaction (Figure 41).
다음으로 TRIM25 결손(KO) 세포에 TRIM25 야생형 또는 7KA RNA 결합 돌연변이체를 이소 발현시켜 rescue 실험을 수행하였다(도 42). 그 결과, 야생형 TRIM25는 비변형 mRNA로부터 발현되는 루시퍼레이스를 억제하였으나, 7KA 돌연변이체는 억제 효과를 나타내지 못하였다. 이는 TRIM25 매개 억제에서 RNA 결합의 중요성을 시사한다. 한편, m1Ψ 변형 mRNA는 어떤 조건에서도 영향을 받지 않았으며, 이는 조절 작용의 특이성을 의미한다.Next, rescue experiments were performed by ectopically expressing TRIM25 wild-type or 7KA RNA-binding mutant in TRIM25 knockout (KO) cells (Fig. 42). As a result, wild-type TRIM25 repressed luciferase expressed from unmodified mRNA, whereas the 7KA mutant did not. This suggests the importance of RNA binding in TRIM25-mediated repression. Meanwhile, m1Ψ-modified mRNA was unaffected under any conditions, indicating the specificity of its regulatory action.
(3) TRIM25 매개 억제에서 E3 리가아제 활성의 역할 확인(3) Confirmation of the role of E3 ligase activity in TRIM25-mediated inhibition
TRIM25는 RNA 결합 단백질로서의 역할 외에도, E3 유비퀴틴 리가아제로 기능한다. TRIM25 매개 억제에서 E3 리가아제 활성이 수행하는 역할을 조사하기 위해, RING 도메인 내 아르지닌 잔기에 돌연변이(R54P)를 도입하였다. 이 돌연변이는 E2 유비퀴틴 연결 효소와의 상호작용을 저해하여 E3 리가아제 활성을 상실시키는 것으로 알려져 있다. Rescue 실험에서, R54P 돌연변이체는 비변형 IVT mRNA의 발현을 억제하지 못하였으며(도 42), 이는 TRIM25 매개 억제에 E3 리가아제 활성이 기여함을 의미한다.In addition to its role as an RNA-binding protein, TRIM25 functions as an E3 ubiquitin ligase. To investigate the role of E3 ligase activity in TRIM25-mediated repression, a mutation (R54P) was introduced at an arginine residue within the RING domain. This mutation is known to impair interaction with the E2 ubiquitin ligase, resulting in loss of E3 ligase activity. In rescue experiments, the R54P mutant failed to repress the expression of unmodified IVT mRNA (Figure 42), indicating that E3 ligase activity contributes to TRIM25-mediated repression.
RNA 결합이 TRIM25의 E3 리가아제 활성을 증가시키는지를 확인하기 위해, in vitro 유비퀴틴화 실험을 수행하였다. 이를 위해, 기질 단백질이 함께 침전될 것으로 예상되는 FLAG-tagged TRIM25를 면역침강한 후, 해당 면역침강물에 E1, E2, 및 유비퀴틴을 첨가하고 IVT mRNA의 존재 또는 부재 하에서 반응시켰다. 그 결과, IVT mRNA를 첨가했을 때 유비퀴틴화된 단백질의 수준이 증가하였으며, 이는 RNA 결합이 TRIM25의 유비퀴틴화 리가아제 활성을 유도함을 나타낸다(도 43). 특히, 비변형 mRNA가 m1Ψ 변형 mRNA보다 더 강한 효과를 나타내었다. 또 다른 종류의 RNA(EGFP mRNA)에서도 유사한 효과가 관찰되었다. RNA 결합 돌연변이체(7KA)는 유비퀴틴화 수준이 감소하였으며, E3 리가아제 활성이 결여된 돌연변이체(R54P)는 음성 대조군으로 사용되었다. 종합하면, m1Ψ 변형은 RNA-TRIM25 상호작용 및 E3 리가아제 활성을 감소시키며, 이를 통해 변형된 RNA가 TRIM25 매개 감시로부터 회피할 수 있도록 돕는다.To determine whether RNA binding enhances the E3 ligase activity of TRIM25, we performed an in vitro ubiquitination experiment. To this end, FLAG-tagged TRIM25, which is expected to co-precipitate with substrate proteins, was immunoprecipitated. E1, E2, and ubiquitin were added to the immunoprecipitate and reacted in the presence or absence of IVT mRNA. The addition of IVT mRNA increased the level of ubiquitinated proteins, indicating that RNA binding induces the ubiquitination ligase activity of TRIM25 (Figure 43). Notably, unmodified mRNA exhibited a stronger effect than m1Ψ-modified mRNA. A similar effect was observed with another type of RNA (EGFP mRNA). An RNA-binding mutant (7KA) showed reduced ubiquitination levels, and a mutant lacking E3 ligase activity (R54P) served as a negative control. In summary, the m1Ψ variant reduces RNA-TRIM25 interaction and E3 ligase activity, thereby helping modified RNAs to evade TRIM25-mediated surveillance.
실시예 6. 산성화된 엔도솜을 통해 유입되는 외인성 RNA에 대한 TRIM25의 타겟팅 확인Example 6. Confirmation of TRIM25 Targeting Exogenous RNA Entering Acidified Endosomes
(1) LNP 전달 RNA에 대한 TRIM25의 타겟팅 확인(1) Confirmation of TRIM25 targeting by LNP delivery RNA
GFP-안정 발현 세포에서 발현된 EGFP mRNA는 IVT EGFP mRNA와 거의 동일함에도 불구하고, TRIM25는 GFP-안정 발현 세포를 이용한 대조 스크리닝에서는 확인되지 않았다. GFP392 안정 발현 세포에서 수행한 knockdown 실험을 통해, TRIM25가 외인성 mRNA와 핵에서 수출된 내인성 mRNA를 구별할 수 있음을 확인하였다(도 44).Although EGFP mRNA expressed in GFP-stabilized cells is nearly identical to IVT EGFP mRNA, TRIM25 was not detected in a control screening using GFP-stabilized cells. Knockdown experiments performed in GFP392 stably expressed cells confirmed that TRIM25 can distinguish between exogenous mRNA and endogenous mRNA exported from the nucleus (Fig. 44).
이러한 결과를 바탕으로, TRIM25의 RNA 타겟팅이 RNA가 세포질로 유입되는 경로에 의해 영향을 받는지를 조사하였다. 이를 확인하기 위해, 양이온성 지질 기반의 트랜스펙션 시약인 Lipofectamine을 사용하였다. Lipofectamine은 생리학적 pH에서 지속적인 양전하를 띠므로 형질도입 물질을 원형질막과 초기 엔도솜을 통해 전달하는 것으로 알려져 있다. Based on these results, we investigated whether TRIM25's RNA targeting is influenced by the RNA entry pathway into the cytoplasm. To confirm this, we used Lipofectamine, a cationic lipid-based transfection reagent. Lipofectamine, which maintains a persistent positive charge at physiological pH, is known to transport transfection agents across the plasma membrane and early endosomes.
그 결과, Lipofectamine을 통해 전달된 Fluc mRNA는 TRIM25 KO 세포에서 루시퍼레이스 발현 수준이 약간 증가하는 정도에 그쳤다. 또 다른 mRNA인 EGFP mRNA 역시 Lipofectamine을 통해 전달했을 때, 모세포와 KO 세포 간에 유의미한 차이를 보이지 않았다. 이상과 같은 결과는 TRIM25는 양이온성 지질을 통해 전달된 mRNA를 효과적으로 타겟팅하지 못함을 의미한다(도 45).As a result, Fluc mRNA delivered via Lipofectamine only slightly increased luciferase expression levels in TRIM25 KO cells. Another mRNA, EGFP mRNA, also showed no significant difference between parental and KO cells when delivered via Lipofectamine. These results indicate that TRIM25 does not effectively target mRNA delivered via cationic lipids (Figure 45).
이어서 전기천공법(electroporation)을 사용하여 RNA를 세포질로 유입시켰다. 전기천공법은 원형질막에 일시적인 구멍을 형성하여 mRNA가 세포 내로 진입할 수 있도록 하는 방법이다. 전기천공법을 사용한 경우, TRIM25의 결손 또는 knockdown은 리포터 mRNA에 유의미한 영향을 미치지 않았으며, 이는 TRIM25가 원형질막을 직접 통과하여 전달된 mRNA를 억제하지 못함을 의미한다(도 46). 반면, mRNA-1273에서와 같이 SM-102(이온화 지질), DMG-PEG 2000, DSPC 및 콜레스테롤로 구성된 또 다른 유형의 LNP를 사용하였을 때에는, TRIM25의 결핍이 Fluc 발현에 강한 영향을 미치는 것으로 나타났다(도 47). 이상과 같은 결과는 TRIM25는 LNP를 통해 전달된 외인성 mRNA를 선택적으로 타겟팅할 수 있음을 의미한다.Next, RNA was introduced into the cytoplasm using electroporation. Electroporation is a method that creates temporary pores in the plasma membrane to allow mRNA to enter the cell. When electroporation was used, deletion or knockdown of TRIM25 did not significantly affect reporter mRNA, indicating that TRIM25 does not inhibit mRNA delivered directly through the plasma membrane (Figure 46). In contrast, when another type of LNP consisting of SM-102 (ionized lipid), DMG-PEG 2000, DSPC, and cholesterol was used, as in mRNA-1273, TRIM25 deficiency strongly affected Fluc expression (Figure 47). These results indicate that TRIM25 can selectively target exogenous mRNA delivered via LNPs.
(2) TRIM25의 pH 감응적 RNA 결합 특성 확인(2) Confirmation of the pH-sensitive RNA binding properties of TRIM25
LNP 내의 이온화 지질은 엔도솜 성숙 과정 중의 산성 환경에서 양성자로 프로톤화되어 양전하를 띠게 되며, 이로 인해 엔도솜 파열이 유도된다. 엔도솜 막을 사이에 둔 높은 프로톤 농도 기울기(약 10~100배)를 고려할 때, 산성화된 엔도솜(pH 5.4-5.8)의 파열은 프로톤 이온을 방출하고 파열 부위 인근의 pH를 일시적으로 저하시키는 것으로 예상된다. 이에 따라, TRIM25의 RNA 결합 활성이 pH 변화에 의해 영향을 받는지 검증하였다. 구체적으로, IVT RNA를 oligo-dT 비드에 고정하고 세포 용해물을 사용하여 pH 6.5~8.0 범위에서 RNA 결합 실험을 수행하였다. During endosomal maturation, ionized lipids within LNPs are protonated and positively charged in the acidic environment, inducing endosome rupture. Given the high proton gradient (approximately 10- to 100-fold) across the endosomal membrane, rupture of acidified endosomes (pH 5.4-5.8) is expected to release protons and temporarily lower the pH near the rupture site. Therefore, we examined whether the RNA-binding activity of TRIM25 is affected by pH changes. Specifically, IVT RNA was immobilized on oligo-dT beads, and RNA-binding experiments were performed using cell lysates at pH 6.5-8.0.
그 결과, TRIM25는 약산성 조건(pH 6.5)에서 UTR-1 RNA에 강하게 결합하였으며, pH 6.5와 7.0 사이에서 결합 강도에 급격한 변화가 나타났다. 유사한 pH 감수성은 또 다른 RNA(UTR-2)를 사용한 유사한 RNA 풀다운 실험에서도 관찰되었다(도 48 및 도 49).As a result, TRIM25 strongly bound to UTR-1 RNA under slightly acidic conditions (pH 6.5), and the binding strength showed a sharp change between pH 6.5 and 7.0. Similar pH sensitivity was also observed in similar RNA pull-down experiments using another RNA (UTR-2) (Figs. 48 and 49).
추가적으로, 생화학적으로 합성한 RNA를 biotin-avidin 결합을 통해 고정한 후, 서로 다른 두 종류의 완충용액 조건에서 RNA 결합 실험을 수행하여 TRIM25의 pH 감응적 RNA 결합 특성을 추가로 확인하였다. TRIM25와 달리, hnRNPA1 및 G3BP1과 같은 다른 RNA 결합 단백질들은 pH 변화에 의해 거의 영향을 받지 않았다. 정제된 재조합 TRIM25 단백질을 이용한 RNA 결합 실험에서는, TRIM25가 산성 조건에서 RNA와의 직접적인 상호작용이 증가함을 보여주었다(도 50). Additionally, the pH-sensitive RNA-binding properties of TRIM25 were further confirmed by performing RNA-binding experiments in two different buffer conditions after immobilizing biochemically synthesized RNA via biotin-avidin conjugation. Unlike TRIM25, other RNA-binding proteins, such as hnRNPA1 and G3BP1, were hardly affected by pH changes. RNA-binding experiments using purified recombinant TRIM25 protein showed that TRIM25's direct interaction with RNA increased under acidic conditions (Figure 50).
pH 감수성의 범위를 보다 정밀하게 파악하기 위해, 0.2 단위 간격으로 pH를 변화시키며 결합 실험을 반복하였다. TRIM25의 RNA 결합 친화도는 생리적 세포질 pH인 pH 7.2와 pH 7.0 사이에서도 유의미하게 증가하였으며, 더 낮은 pH에서 추가적으로 증가하였다. 이는 세포 내에서의 미세한 pH 변화만으로도 TRIM25-RNA 상호작용이 유도될 수 있음을 의미한다(도 51). 주목할 점은, m1Ψ가 도입된 RNA는 모든 실험 조건에서 비변형 RNA에 비해 TRIM25에 대한 결합력이 낮았다는 점이다. 이는 m1Ψ 변형이 산성 조건에서도 TRIM25 결합을 저해함을 나타낸다. 이러한 결과들은, 프로톤 이온이 TRIM25의 RNA 결합 활성을 증가시킴으로써, 산성화된 엔도솜에서 탈출하는 외부 RNA를 선택적으로 포획할 수 있는 가능성을 제공함을 의미한다(도 52).To more precisely determine the range of pH sensitivity, binding experiments were repeated while varying the pH in 0.2-unit intervals. The RNA-binding affinity of TRIM25 significantly increased between pH 7.2 and pH 7.0, which are physiological cytoplasmic pHs, and further increased at lower pHs. This suggests that even subtle pH changes within cells can induce TRIM25-RNA interactions (Fig. 51). Notably, RNAs with m1Ψ introduced showed lower binding affinity to TRIM25 than unmodified RNA under all experimental conditions. This indicates that the m1Ψ modification inhibits TRIM25 binding even under acidic conditions. These results suggest that proton ions may selectively capture foreign RNAs escaping from acidified endosomes by enhancing the RNA-binding activity of TRIM25 (Fig. 52).
참조예Reference example
참조예 1. 플라스미드 제작Reference Example 1. Plasmid Production
인간 TRIM25 발현 플라스미드를 제작하기 위해, TRIM25의 참조 코딩서열(RefSeq NM_005082) 중 단일 SNP(dbSNP rs205498, G/A 변이로 P358L 점돌연변이에 해당하며, 정제된 대립유전자에서 77% 이상에서 발견됨)을 포함한 서열을 PCR 증폭하여, N말단에 FLAG 태그가 부착된 pCK 벡터에 삽입하여 외인성 발현용 플라스미드를 제작하였다. 단백질 정제를 위한 용도로는 동일 서열을 His10-eYFP-SUMOstar-Strep 태그가 N말단에 부착된 pX 벡터에 서브클로닝하였다. TRIM25의 기타 변이체들, 예를 들어 7KA RNA 결합 결손 변이체 및 R54P E3 리가아제 결손 변이체는 부위특이적 돌연변이 유도법(site-directed mutagenesis)을 통해 제작하였다. rescue 실험 및 RNA pulldown 실험에 사용하기 위해 CMV 프로모터는 PGK 프로모터로 교체하였고, 안정적인 발현을 확보하기 위해 FLAG 태그는 제거하였다. 본 연구에 사용된 플라스미드 목록은 표 1에 정리되어 있다.To construct a human TRIM25 expression plasmid, a sequence containing a single SNP (dbSNP rs205498, a G/A mutation corresponding to the P358L point mutation, found in more than 77% of purified alleles) in the reference coding sequence of TRIM25 (RefSeq NM_005082) was PCR amplified and inserted into the pCK vector with a FLAG tag at the N terminus to construct a plasmid for exogenous expression. For protein purification, the same sequence was subcloned into the pX vector with a His10-eYFP-SUMOstar-Strep tag at the N terminus. Other mutants of TRIM25, such as the 7KA RNA-binding defective mutant and the R54P E3 ligase defective mutant, were constructed by site-directed mutagenesis. For rescue and RNA pulldown experiments, the CMV promoter was replaced with the PGK promoter, and the FLAG tag was removed to ensure stable expression. The list of plasmids used in this study is summarized in Table 1.
참조예 2. 세포 배양 및 세포주의 구축Reference Example 2. Cell culture and cell line establishment
(1) HCT116 야생형 모세포주(1) HCT116 wild-type parental cell line
HCT116 야생형 모세포주 및 이로부터 유래된 세포주들은 9.1% 태아소 혈청(FBS) (Welgene, S001-01)이 첨가된 McCoy's 5A 배지(Welgene, LM 005-01)에서 배양하였다. HEK293T, lenti-X HEK293T, HeLa, MEF 세포는 9.1% FBS가 포함된 DMEM(Welgene, LM 001-05)에서 배양하였으며, 부유성 HEK293E는 5% FBS와 50 μg/mL G418 황산염(Gibco, 10131-027)이 포함된 DMEM(Welgene, LM001-170)에서 배양하였다. THP-1 세포는 9.1% FBS 및 0.05mM β-mercaptoethanol(Thermo Scientific, 35602BID)이 첨가된 RPMI 1640(Welgene, LM 011-03)에서 배양하였고, Jurkat 세포는 9.1% FBS가 첨가된 동일한 RPMI 1640 배지에서 배양하였다. 모든 세포주는 마이코플라스마 음성으로 확인되었다.HCT116 wild-type parental cell line and cell lines derived from it were cultured in McCoy's 5A medium (Welgene, LM 005-01) supplemented with 9.1% fetal bovine serum (FBS) (Welgene, S001-01). HEK293T, lenti-X HEK293T, HeLa, and MEF cells were cultured in DMEM (Welgene, LM 001-05) containing 9.1% FBS, and suspended HEK293E cells were cultured in DMEM (Welgene, LM001-170) containing 5% FBS and 50 μg/mL G418 sulfate (Gibco, 10131-027). THP-1 cells were cultured in RPMI 1640 (Welgene, LM 011-03) supplemented with 9.1% FBS and 0.05 mM β-mercaptoethanol (Thermo Scientific, 35602BID), and Jurkat cells were cultured in the same RPMI 1640 medium supplemented with 9.1% FBS. All cell lines were confirmed to be mycoplasma-negative.
(2) 마우스 유래 골수 대식세포(mBMDMs)(2) Mouse-derived bone marrow macrophages (mBMDMs)
마우스 유래 골수 대식세포(mBMDMs) 배양을 위해 6주령의 야생형 암컷 C57BL/6J 마우스의 대퇴골 및 경골에서 골수 세포를 분리하였다. 세포는 다음이 포함된 DMEM에서 배양하였다: 10% 열불활성화 FBS(Biowest, S1620), 1% 비필수 아미노산(Thermo Scientific, 11140-050), 1% penicillin/streptomycin(Thermo Scientific, 15070-063), 및 M-CSF 공급원으로서 20% L929 조건 배지. L929 조건 배지는 Bowdish Lab(McMaster University) 프로토콜에 따라 L929 세포(ATCC, CCL-1)를 이용해 제조하였으며, IMDM 배지(Thermo Scientific, 12440053)에 10% FBS 및 1% penicillin/streptomycin을 첨가하여 배양하였다. 3일째와 5일째에 M-CSF가 포함된 신선한 배지 10 mL를 추가하였고, 6일째에 세포를 부드럽게 긁어 수확하였다. 이후 모든 실험에서는 항생제 비첨가 배지를 사용하였다.For the culture of mouse-derived bone marrow macrophages (mBMDMs), bone marrow cells were isolated from the femurs and tibias of 6-week-old wild-type female C57BL/6J mice. Cells were cultured in DMEM containing 10% heat-inactivated FBS (Biowest, S1620), 1% nonessential amino acids (Thermo Scientific, 11140-050), 1% penicillin/streptomycin (Thermo Scientific, 15070-063), and 20% L929 conditioned medium as a source of M-CSF. L929 conditioned medium was prepared using L929 cells (ATCC, CCL-1) according to the Bowdish Lab (McMaster University) protocol and cultured in IMDM medium (Thermo Scientific, 12440053) supplemented with 10% FBS and 1% penicillin/streptomycin. On days 3 and 5, 10 mL of fresh medium containing M-CSF was added, and on day 6, cells were gently scraped and harvested. In all subsequent experiments, antibiotic-free medium was used.
(3) HCT116 GFP-stable 세포주(3) HCT116 GFP-stable cell line
HCT116 GFP-stable 세포주를 만들기 위해, 최소 CMV 프로모터, CRISPR 스크리닝에 사용된 in vitro 전사 RNA의 5′ 및 3′ 비번역 영역(UTR)을 포함하는 EGFP 코딩 서열, 및 하위 SV40 poly(A) 시그널을 포함한 플라스미드를 렌티바이러스 백본 벡터(Addgene #60954)를 사용해 구성하였다. 렌티바이러스는 lenti-X HEK293T 세포에 렌티바이러스 패키징 플라스미드를 포함한 플라스미드들을 공형질감염하여 생산하였고, 생성된 바이러스는 0.45μm 필터(Millipore, SLHVR33RS)를 통해 여과하였다. 수확한 렌티바이러스를 8 μg/mL polybrene(Millipore, TR-1003-G)과 함께 HCT116 모세포주에 감염시켰다. GFP 양성 세포는 유세포분석기(FACS)를 이용해 선별하고 단일 세포로 분리하였다. 클론을 확장 배양한 뒤, GFP 형광이 가장 균일하게 나타나는 단일 클론을 후속 실험에 사용하였다.To establish the HCT116 GFP-stable cell line, a plasmid containing the EGFP coding sequence including the minimal CMV promoter, the 5′ and 3′ untranslated regions (UTRs) of the in vitro-transcribed RNA used for CRISPR screening, and the downstream SV40 poly(A) signal was constructed using a lentiviral backbone vector (Addgene #60954). Lentivirus was produced by cotransfecting lenti-X HEK293T cells with the plasmids containing the lentiviral packaging plasmid, and the produced virus was filtered through a 0.45 μm filter (Millipore, SLHVR33RS). The harvested lentivirus was then used to infect the HCT116 parental cell line together with 8 μg/mL polybrene (Millipore, TR-1003-G). GFP-positive cells were selected using flow cytometry (FACS) and isolated as single cells. After expansion of the clones, the single clone with the most uniform GFP fluorescence was used in subsequent experiments.
(4) HCT116 TRIM25 knockout 세포주(4) HCT116 TRIM25 knockout cell line
HCT116 TRIM25 knockout 세포주는 표준 TRIM25 mRNA 아이소폼(RefSeq NM_005082.4)을 표적하는 sgRNA를 Cas9 발현 백본 플라스미드(Addgene #48148)에 서브클로닝하였다. 두 개의 독립 sgRNA는 Brunello 인간 CRISPR knockout pooled library(Addgene #73179)에서 선정하였다: 엑손 3을 표적하는 sgRNA(sgTRIM25-1, 본 연구에서 TRIM25 KO-1 클론에 사용, GCAGCTACAACAAGAATACA, 서열번호 165), 및 엑손 7을 표적하는 sgRNA(sgTRIM25-2, TRIM25 KO-2 클론에 사용, TGTTCCGGGGCTCCAAACGT, 서열번호 166). HCT116 야생형 모세포주에 Cas9과 sgRNA를 발현하는 플라스미드를 공형질감염하고, 제한 희석을 통해 단일 세포를 분리하였다. TRIM25의 결실 여부는 Sanger 염기서열 분석과 western blotting으로 확인하였다.The HCT116 TRIM25 knockout cell line was constructed by subcloning sgRNAs targeting the canonical TRIM25 mRNA isoform (RefSeq NM_005082.4) into a Cas9 expression backbone plasmid (Addgene #48148). Two independent sgRNAs were selected from the Brunello human CRISPR knockout pooled library (Addgene #73179): an sgRNA targeting exon 3 (sgTRIM25-1, used in the TRIM25 KO-1 clone in this study; GCAGCTACAACAAGAATACA, SEQ ID NO: 165), and an sgRNA targeting exon 7 (sgTRIM25-2, used in the TRIM25 KO-2 clone; TGTTCCGGGGCTCCAAACGT, SEQ ID NO: 166). HCT116 wild-type parental cells were cotransfected with plasmids expressing Cas9 and sgRNA, and single cells were isolated by limiting dilution. TRIM25 deletion was confirmed by Sanger sequencing and Western blotting.
(5) HCT116 pooled N4BP1 knockout 세포주(5) HCT116 pooled N4BP1 knockout cell line
HCT116 pooled N4BP1 knockout 세포주는 렌티바이러스 감염을 통해 제작되었다. Cas9과 비표적 대조 sgRNA 또는 N4BP1 표적 sgRNA를 발현하는 렌티바이러스 벡터(Addgene #52961)를 클로닝하였으며, 사용된 sgRNA 서열은 다음과 같다: sgCtrl-1 (CTATATTGTCGCGCAGTGGA, 서열번호 1), sgCtrl-2 (CTCCCTGCCGGCCGGGTTAG, 서열번호 2), sgN4BP1-1 (AGAAAGAGAATGTTACCCCA, 서열번호 3), sgN4BP1-2 (TTACACAGAATGCTGCCACA, 서열번호 4). 이 sgRNA들은 Brunello 인간 CRISPR knockout pooled library(Addgene #73179)에서 선정되었다. CRISPR 렌티바이러스는 lenti-X HEK293T 세포에 렌티바이러스 패키징 플라스미드(Addgene #12259, pMD2.G 및 #12260, psPAX2)와 각 CRISPR 렌티바이럴 백본 플라스미드를 트랜스펙션한 후 생성 및 수집하였다. 생성된 각 렌티바이러스는 8 μg/mL polybrene을 포함한 조건에서 HCT116 야생형 또는 TRIM25 결손 세포에 1일간 감염시켰고, 다음 날 신선한 배지로 교체하였다. 감염된 세포는 1 μg/mL puromycin 하에서 5일간 선별하였으며, N4BP1 결손 여부는 웨스턴 블롯을 통해 검증하였다.The HCT116 pooled N4BP1 knockout cell line was generated via lentiviral infection. Lentiviral vectors expressing Cas9 and a non-targeting control sgRNA or an N4BP1-targeting sgRNA were cloned (Addgene #52961). The sgRNA sequences used were as follows: sgCtrl-1 (CTATATTGTCGCGCAGTGGA, SEQ ID NO: 1), sgCtrl-2 (CTCCCTGCCGGCCGGGTTAG, SEQ ID NO: 2), sgN4BP1-1 (AGAAAGAGAATGTTACCCCA, SEQ ID NO: 3), and sgN4BP1-2 (TTACACAGAATGCTGCCACA, SEQ ID NO: 4). These sgRNAs were selected from the Brunello human CRISPR knockout pooled library (Addgene #73179). CRISPR lentiviruses were generated and harvested by transfecting lentiviral packaging plasmids (Addgene #12259, pMD2.G and #12260, psPAX2) and each CRISPR lentiviral backbone plasmid into lenti-X HEK293T cells. Each generated lentivirus was used to infect HCT116 wild-type or TRIM25-deficient cells for 1 day under conditions containing 8 μg/mL polybrene, and the medium was replaced with fresh medium the following day. Infected cells were selected for 5 days under 1 μg/mL puromycin, and N4BP1 deletion was verified by Western blotting.
참조예 3. In vitro transcription (IVT)Reference Example 3. In vitro transcription (IVT)
(1) IVT 템플릿 설계(1) IVT template design
진핵생물 mRNA를 모사한 mRNA의 in vitro transcription(IVT)을 위해, 본 연구에 사용된 리포터 유전자들(Enhanced green fluorescent protein, EGFP; Firefly luciferase, Fluc; Renilla luciferase, Rluc)의 코딩 서열(CDS)을 5′ 및 3′ 비번역구간(UTR)으로 둘러싸인 mRNA 생산용 플라스미드에 서브클로닝하였다. 개시 코돈을 포함하는 open reading frame으로는 표준 EGFP 서열을 사용하였으며, Fluc/Rluc의 경우 이전 연구에서 사용된 pmirGLO-3XmiR-1 듀얼 루시퍼레이스 플라스미드(Promega의 백본 플라스미드)에서 유래된 서열을 사용하였다. UTR 구성은 CRISPR 스크리닝 및 검증 실험에서 Moderna의 선행 연구에서 보고된 조합[Cell 168, 1114-1125.e10]을 따랐다. 5′ UTR은 Moderna COVID-19 mRNA 백신(mRNA-1273)의 최초 41뉴클레오타이드 합성 서열(GenBank: OR134578.1)로, Kozak 서열이 포함되어 있으며, 3′ UTR은 인간 알파글로빈(alpha-globin) mRNA에서 유래된 서열을 사용하였다. 도 24에 사용된 Fluc mRNA-2 및 Rluc mRNA의 경우, pmirGLO-3XmiR-1 듀얼 루시퍼레이스 벡터에서 발현될 것으로 예측되는 서열을 기반으로 UTR 구성이 달라졌다.For in vitro transcription (IVT) of mRNAs mimicking eukaryotic mRNAs, the coding sequences (CDSs) of the reporter genes used in this study (Enhanced green fluorescent protein, EGFP; Firefly luciferase, Fluc; Renilla luciferase, Rluc) were subcloned into mRNA production plasmids flanked by 5′ and 3′ untranslated regions (UTRs). The standard EGFP sequence was used as the open reading frame including the initiation codon, and for Fluc/Rluc, the sequence derived from the pmirGLO-3XmiR-1 dual luciferase plasmid (backbone plasmid from Promega) used in a previous study was used. The UTR composition followed the combination reported in a previous study by Moderna [Cell 168, 1114-1125.e10] in CRISPR screening and validation experiments. The 5′ UTR is the original 41-nucleotide synthetic sequence of Moderna COVID-19 mRNA vaccine (mRNA-1273) (GenBank: OR134578.1), which includes the Kozak sequence, and the 3′ UTR uses a sequence derived from human alpha-globin mRNA. For Fluc mRNA-2 and Rluc mRNA used in Figure 24, the UTR configuration was changed based on the sequence predicted to be expressed in the pmirGLO-3XmiR-1 dual luciferase vector.
(2) IVT mRNA 합성 및 변형(2) IVT mRNA synthesis and modification
IVT용 템플릿은 T7 프로모터 서열이 포함된 forward primer와 poly(T) 서열이 포함된 reverse primer를 이용하여 PCR 증폭을 통해 제작하였다. 이들 프라이머는 각각 목표하는 5′ 및 3′ UTR을 표적하도록 설계되었으며, reverse primer의 5′ 말단 2뉴클레오타이드는 2′-O-메틸화 처리되어 있어 전사 종결의 정확도를 높이고 부산물 생성을 방지한다. 도 4에서 사용된 non-optimal RNA를 생성할 때는 2′-O-메틸화가 없는 reverse primer를 사용하였다. PCR로 증폭한 IVT DNA 템플릿은 아가로스 겔 전기영동 후 겔 추출(Qiagen, 28704)하였고, 이후 추가로 올리고 정제(Zymo Research, D4061)를 거쳐 mMESSAGE mMACHINE T7 전사 키트(Invitrogen, AM1344)를 사용하여 IVT mRNA를 합성하였다. The IVT template was generated by PCR amplification using a forward primer containing the T7 promoter sequence and a reverse primer containing a poly(T) sequence. These primers were designed to target the 5′ and 3′ UTRs of interest, respectively, and the 5′-terminal two nucleotides of the reverse primer were 2′-O-methylated to increase the accuracy of transcription termination and prevent byproduct formation. When generating the non-optimal RNA used in Figure 4, a reverse primer without 2′-O-methylation was used. The PCR-amplified IVT DNA template was subjected to agarose gel electrophoresis, gel-extracted (Qiagen, 28704), and further oligo-purified (Zymo Research, D4061) to synthesize IVT mRNA using the mMESSAGE mMACHINE T7 Transcription Kit (Invitrogen, AM1344).
mRNA의 캡핑(capping)과 폴리어데닐화(polyadenylation)는 전사와 동시에 수행되었다. 캡핑에는 대표적인 cap1 아날로그(CleanCap AG (3′-OMe), TriLink, N-7413)를 사용하였다. 캡 구조에 따른 차이를 비교하기 위한 실험(도 24)에서는 1세대 cap0 아날로그(mCAP, TriLink, N-7001)와 이후 개발된 cap0 아날로그(ARCA, TriLink, N-7003)를 사용하였다. cap1 아날로그의 삽입률을 높이기 위해, IVT 템플릿용 PCR에서 +1 위치의 염기를 G 대신 A로 시작하는 forward primer를 사용하는 변형된 설계가 적용되었으며, 해당 조건은 다음 그림에 사용된 mRNA 합성에 적용되었다: 도 4, 도 17, 도 23, 도 24, 도 26, 도 27, 도 31, 도 32, 도 34, 도 35, 도 43, 도 45, 도 46, 도 47. mRNA의 poly(A) 꼬리는 템플릿에 삽입된 poly(T) 서열을 전사하여 생성하였으며, 대부분의 IVT mRNA에서는 120 nt의 고정된 길이를 사용하였고, 도 4의 Fluc mRNA와 도 24의 Rluc mRNA에서는 60 nt 길이를 사용하였다.mRNA capping and polyadenylation were performed concurrently with transcription. Representative cap1 analogs (CleanCap AG (3′-OMe), TriLink, N-7413) were used for capping. Experiments comparing differences in cap structure (Fig. 24) used first-generation cap0 analogs (mCAP, TriLink, N-7001) and later-developed cap0 analogs (ARCA, TriLink, N-7003). To increase the insertion rate of cap1 analogs, a modified design was applied using a forward primer starting with A instead of G at the +1 position in the PCR for IVT templates, and the conditions were applied to the mRNA synthesis used in the following figures: Fig. 4, Fig. 17, Fig. 23, Fig. 24, Fig. 26, Fig. 27, Fig. 31, Fig. 32, Fig. 34, Fig. 35, Fig. 43, Fig. 45, Fig. 46, Fig. 47. The poly(A) tail of the mRNA was generated by transcribing the poly(T) sequence inserted into the template, and a fixed length of 120 nt was used for most IVT mRNAs, while a length of 60 nt was used for Fluc mRNA in Fig. 4 and Rluc mRNA in Fig. 24.
(3) 염기 변형 mRNA 및 circRNA의 합성(3) Synthesis of base-modified mRNA and circRNA
도 37에서 사용된 염기 변형 mRNA를 합성하기 위해, IVT 반응에서 각 NTP는 원하는 변형된 NTP로 전량 치환하여 사용하였다. 사용된 변형 NTP는 다음과 같다:To synthesize the base-modified mRNA used in Figure 37, each NTP in the IVT reaction was completely replaced with the desired modified NTP. The modified NTPs used were as follows:
N1-methylpseudoUTP (TriLink, N-1081), pseudoUTP (TriLink, N-1019), 5-methoxyUTP (TriLink, N-1093), 5-methylCTP (TriLink, N-1014).N1-methylpseudoUTP (TriLink, N-1081), pseudoUTP (TriLink, N-1019), 5-methoxyUTP (TriLink, N-1093), 5-methylCTP (TriLink, N-1014).
전사 후, RNase-free DNase(Takara, 2270A)로 DNA 템플릿을 제거하고, 생성된 mRNA는 Qiagen의 RNeasy Mini Kit(74106)를 사용하여 RNA 정제 프로토콜에 따라 정제하였으며, 추가적인 on-column DNase 처리(Qiagen, 79254)를 수행하였다. NTP의 잔류를 최소화하고 RNA의 순도를 극대화하기 위해, 정제된 RNA는 RNA cleanup 정제를 한 차례 더 수행하였다.After transcription, the DNA template was removed with RNase-free DNase (Takara, 2270A), and the generated mRNA was purified according to the RNA purification protocol using Qiagen's RNeasy Mini Kit (74106), with additional on-column DNase treatment (Qiagen, 79254). To minimize NTP residues and maximize RNA purity, the purified RNA was further purified using RNA cleanup.
도 29, 30, 및 36에서 사용된 원형 RNA(circRNA)의 경우, Fluc 서열을 포함하는 플라스미드로부터 PCR을 통해 IVT 템플릿을 제작하였다. 이 템플릿은 circRNA-synIRES-R25-NanoLuc (Addgene #188116)에서 유래되었으며, 본 연구에서는 NanoLuc CDS를 Fluc CDS로 대체하였다. IVT 템플릿 제작과 circRNA 합성은 기존 보고된 방법[Nat. Biotechnol. 41, 262-272 및 Nat. Commun. 9, 2629] 약간 수정하여 수행하였다. 간단히 설명하면, circRNA는 HiScribe™ T7 High Yield RNA Synthesis Kit (NEB, E2040S)를 사용하여 in vitro 전사하였고, 전사 후 DNase 처리로 DNA 템플릿을 제거하였다. RNA 환형화는 2 mM GTP를 첨가하고 55°C에서 15분간 반응시켜 촉진하였다. 환형화된 RNA는 컬럼 정제를 거친 뒤, RNase R(Abm, E049)을 사용하여 37°C에서 15분간 처리하여 직선형 RNA를 제거하였고, 이후 다시 한 번 컬럼 정제를 수행하였다. 최종적으로, circRNA는 Zymoclean Gel RNA Recovery Kit (Zymo Research, R1011)를 사용한 아가로스 겔 전기영동 정제를 통해 추가 정제하였다.For the circular RNAs (circRNAs) used in Figures 29, 30, and 36, IVT templates were generated by PCR from a plasmid containing the Fluc sequence. This template was derived from circRNA-synIRES-R25-NanoLuc (Addgene #188116), and in this study, the NanoLuc CDS was replaced with the Fluc CDS. IVT template generation and circRNA synthesis were performed according to previously reported methods [Nat. Biotechnol. 41, 262-272 and Nat. Commun. 9, 2629] with slight modifications. Briefly, circRNAs were transcribed in vitro using the HiScribe™ T7 High Yield RNA Synthesis Kit (NEB, E2040S), and the DNA template was removed by DNase treatment after transcription. RNA circularization was promoted by adding 2 mM GTP and incubating at 55°C for 15 min. Circular RNA was purified by column chromatography, then treated with RNase R (Abm, E049) at 37°C for 15 min to remove linear RNA, followed by another column chromatography purification. Finally, circRNA was further purified by agarose gel electrophoresis using the Zymoclean Gel RNA Recovery Kit (Zymo Research, R1011).
m6A 변형 circRNA의 경우, IVT 반응에서 N6-methylATP(TriLink, N-1013)와 ATP를 지정된 비율로 혼합하여 사용하였다.For m6A-modified circRNAs, N6-methylATP (TriLink, N-1013) and ATP were mixed in the specified ratio in the IVT reaction.
(4) 최종 품질 분석(4) Final quality analysis
최종 IVT RNA의 완전성은 자동 전기영동 장비(Agilent TapeStation, RNA ScreenTape 분석)를 이용해 평가하였으며, LNP 포뮬레이션에 앞서 RNA 농도는 분광광도계를 통해 정량하였다. 본 연구에 사용된 IVT 템플릿 제작용 플라스미드 및 PCR 프라이머 서열, 그리고 IVT mRNA의 CDS 및 UTR 서열에 관한 정보는 표 1 및 표 2에 표기하였다.The integrity of the final IVT RNA was assessed using an automated electrophoresis device (Agilent TapeStation, RNA ScreenTape analysis), and RNA concentration was quantified spectrophotometrically prior to LNP formulation. The sequences of the plasmids and PCR primers used for constructing the IVT templates used in this study, as well as the CDS and UTR sequences of the IVT mRNA, are listed in Tables 1 and 2.
참조예 4. Dot blot 분석Reference Example 4. Dot blot analysis
Dot blot 분석은 [Mol. Ther. Nucleic Acids 15, 26-35]에 제시된 방법에 따라 수행하였다. 말단 2′-O-메틸화 여부에 따라 제작된 Fluc IVT mRNA를 최종 농도 8, 40, 200 ng/μL로 희석하였고, 각 농도에서 5 μL(총량: 40, 200, 1000 ng)를 양전하 나일론 멤브레인(Sigma, GERPN203B)에 점적하였다. 양성 대조군으로는 dsRNA ladder(NEB, N0363S)를 0.8, 4, 20 ng/μL로 희석하여 동일한 방식으로 5 μL(총량: 4, 20, 100 ng)를 점적하였다. 샘플 점적 후, 멤브레인을 60°C에서 20분간 건조시키고, UV-C (12 mJ/cm², 2회 조사)를 이용해 RNA를 멤브레인에 고정하였다. 이후 멤브레인은 5% 탈지우유 용액에서 블로킹하였으며, dsRNA는 α-dsRNA J2 단클론 항체(English and Scientific Consulting Kft, 1:1000)와 HRP-conjugated 2차 항체를 이용한 화학발광법으로 검출하였다.Dot blot analysis was performed according to the method presented in [Mol. Ther. Nucleic Acids 15, 26-35]. Fluc IVT mRNAs produced according to terminal 2′-O-methylation were diluted to final concentrations of 8, 40, and 200 ng/μL, and 5 μL (total volume: 40, 200, and 1000 ng) at each concentration was spotted onto a positively charged nylon membrane (Sigma, GERPN203B). As a positive control, dsRNA ladder (NEB, N0363S) was diluted to 0.8, 4, and 20 ng/μL, and 5 μL (total volume: 4, 20, and 100 ng) was spotted in the same manner. After sample application, the membrane was dried at 60°C for 20 min, and RNA was immobilized on the membrane using UV-C (12 mJ/cm², irradiation twice). The membrane was then blocked in 5% skim milk solution, and dsRNA was detected by chemiluminescence using α-dsRNA J2 monoclonal antibody (English and Scientific Consulting Kft, 1:1000) and HRP-conjugated secondary antibody.
참조예 5. Lipid nanoparticle (LNP) 포뮬레이션 및 형질감염Reference Example 5. Lipid nanoparticle (LNP) formulation and transfection
LNP 포뮬레이션은 이전 보고[Sci. Adv. 7, eabf4398]를 기반으로, mRNA 포뮬레이션에 맞게 일부 수정하여 수행하였다. 우선, LNP는 유기상에, 목적하는 IVT mRNA는 수상에 각각 용해시켜 1:3의 부피 비율로 혼합하였다. 본 연구에서 사용된 LNP는 Pfizer-BioNTech에서 개발한 BNT162b2 COVID-19 mRNA 백신의 조성을 모방하였으며[Nature 595, 572-577 및 Int. J. Pharm. 601, 120586], 다음과 같은 네 가지 지질 성분을 포함하였다: 이온화 지질 (ALC-0315; Echelon Biosciences, N-1020), 인지질 (DSPC; Avanti, 850365), 콜레스테롤 (Sigma, C8667), PEG 결합 지질 (ALC-0159; Echelon Biosciences, N-2010).The LNP formulation was based on a previous report [Sci. Adv. 7, eabf4398] with some modifications to suit the mRNA formulation. First, LNPs were dissolved in the organic phase, and the target IVT mRNA was dissolved in the aqueous phase, and mixed in a volume ratio of 1:3. The LNP used in this study mimicked the composition of the BNT162b2 COVID-19 mRNA vaccine developed by Pfizer-BioNTech [Nature 595, 572-577 and Int. J. Pharm. 601, 120586] and contained the following four lipid components: ionizable lipid (ALC-0315; Echelon Biosciences, N-1020), phospholipid (DSPC; Avanti, 850365), cholesterol (Sigma, C8667), and PEG-linked lipid (ALC-0159; Echelon Biosciences, N-2010).
mRNA 포뮬레이션 전, 지질들은 에탄올(Sigma, E7023)에 몰 비율 46.3:9.4:42.7:1.6으로 용해하여 lipid master mix를 만들었다. 정제된 mRNA는 pH 3의 10 mM 시트르산 완충용액(Sigma, 854)에 희석하였고, 여기에 lipid master mix를 수상:유기상 = 3:1 비율로 첨가하였다.Before mRNA formulation, lipids were dissolved in ethanol (Sigma, E7023) at a molar ratio of 46.3:9.4:42.7:1.6 to prepare a lipid master mix. The purified mRNA was diluted in 10 mM citric acid buffer (Sigma, 854), pH 3, and the lipid master mix was added at a ratio of aqueous phase:organic phase = 3:1.
도 47에서 mRNA-1273 COVID-19 백신의 LNP 조성(LNP-2)을 모방하기 위해, 이온화 지질로는 SM-102(MedChemExpress, HY-134541), PEGylated 지질로는 DMG-PEG 2000(Avanti, 880151P)을 사용하였다. 포뮬레이션된 LNP-mRNA 혼합물은 PBS로 희석하였고, 형질감염 전 Quant-iT RiboGreen RNA Assay(Invitrogen, R11491)를 통해 캡슐화 효율을 측정하였다. 측정된 캡슐화 효율을 기반으로, 12-well 스케일에서 세포당 10-50 ng의 LNP-mRNA를 형질감염하였으며, 유세포 분석을 통해 95% 이상의 형질감염 효율 및 균일한 발현 수준을 확인하였다. 실험 규모 및 세포 유형에 따라 상향 또는 하향 스케일 조정은 비례적으로 수행하였다.To mimic the LNP composition (LNP-2) of the mRNA-1273 COVID-19 vaccine in Figure 47, SM-102 (MedChemExpress, HY-134541) was used as the ionizable lipid, and DMG-PEG 2000 (Avanti, 880151P) was used as the PEGylated lipid. The formulated LNP-mRNA mixture was diluted with PBS, and the encapsulation efficiency was measured using the Quant-iT RiboGreen RNA Assay (Invitrogen, R11491) prior to transfection. Based on the measured encapsulation efficiency, 10-50 ng of LNP-mRNA per cell was transfected in a 12-well plate, and flow cytometry confirmed a transfection efficiency of greater than 95% and a uniform expression level. Up- or down-scaling was performed proportionally depending on the experimental scale and cell type.
참조예 6. 전장 유전체 CRISPR-Cas9 knockout 스크리닝Reference Example 6. Whole-genome CRISPR-Cas9 knockout screening
전장 유전체 수준의 CRISPR-Cas9 knockout 스크리닝을 위해, 본 연구에서는 Brunello 인간 CRISPR knockout pooled library(Addgene #73179)를 사용하였다. 해당 라이브러리는 총 19,114개의 단백질 코딩 유전자를 표적하는 77,441개의 sgRNA(유전자당 약 4개의 독립 sgRNA 포함)와, 정규화를 위한 1,000개의 비표적 sgRNA를 포함하고 있다. 스크리닝은 생물학적 반복 2회로 수행되었다. Cas9 및 sgRNA 라이브러리를 포함한 플라스미드는 기존 보고된 방법[Cell 186, 3291-3306/e21]에 따라 증폭하였고, 스크리닝 전에 sgRNA의 무결성은 차세대 염기서열 분석(NGS)을 통해 검증하였다. CRISPR 렌티바이러스는 lenti-X HEK293T 세포에서 생성 및 수확하였으며, 다양한 농도의 푸로마이신 하에서의 세포 생존율 측정을 통해 감염도(MOI, multiplicity of infection)를 계산하였다.For genome-wide CRISPR-Cas9 knockout screening, the Brunello human CRISPR knockout pooled library (Addgene #73179) was used in this study. The library contains 77,441 sgRNAs (approximately four independent sgRNAs per gene) targeting 19,114 protein-coding genes, plus 1,000 non-targeting sgRNAs for normalization. Screening was performed in two biological replicates. Plasmids containing Cas9 and the sgRNA library were amplified according to a previously reported method [Cell 186, 3291-3306/e21], and the integrity of the sgRNAs was verified by next-generation sequencing (NGS) before screening. CRISPR lentiviruses were produced and harvested from lenti-X HEK293T cells, and the multiplicity of infection (MOI) was calculated by measuring cell viability under various concentrations of puromycin.
Knockout을 위해 HCT116 세포를 신선한 배지에 8 μg/mL polybrene과 pooled CRISPR 렌티바이러스를 포함하여 제한된 MOI(약 0.3) 조건으로 시드하여, 세포당 하나의 유전자만 침묵되는 단일 입자 감염 세포를 생성하였다. 1일간 감염 후 신선한 배지로 교체하였고, 2일째에 세포를 계대 배양한 뒤 1 μg/mL 푸로마이신이 포함된 배지에서 7일간 선별하였다. CRISPR 라이브러리의 최소 300배 커버리지를 확보하기 위해 푸로마이신 처리 기간 동안 3×107개 이상의 세포를 유지하였다. 이후 변형되지 않은 또는 N1-메틸슈도유리딘화된 EGFP mRNA 1 μg을 LNP 포뮬레이션하여, 접시당 1.6×107개의 감염된 세포에 형질감염하였고, 반복당 총 4개의 접시(총 6.4×107개 이상의 세포)를 준비하여 충분한 sgRNA 커버리지를 확보하였다. LNP-mRNA를 pooled knockout 세포에 처리한 지 1일 후, 세포를 트립신 처리하여 분리하고, 3% FBS가 포함된 PBS에 1.2-1.5×107 cells/mL 농도로 재부유시킨 뒤, 35 μm cell strainer를 통과시켜 BD FACS Aria III (BD Biosciences)를 이용한 FACS 분석을 수행하였다. GFP 형광 세기가 가장 높은 상위 0-2.5% 및 2.5-5% 구간(GFPHigh), 또는 가장 낮은 하위 0-2.5% 및 2.5-5% 구간(GFPLow)의 세포를 분리하였으며, 각 조건에서 6.25×105개의 세포가 수집될 때까지 sorting을 수행하였다. 비정렬된 대조군(unsorted control)으로는 2.5×107개의 살아있는 세포를 확보하여 최소 300배 커버리지를 확보하였다. GFP-stable 세포주 스크리닝에서는 HCT116 GFP-stable 세포에 CRISPR 감염만 수행하였고, EGFP mRNA의 LNP 전달은 생략하였다.For knockout, HCT116 cells were seeded in fresh medium containing 8 μg/mL polybrene and pooled CRISPR lentivirus at a limited MOI (approximately 0.3) to generate single-particle infected cells with only one gene silenced per cell. After 1 day of infection, the medium was replaced with fresh medium, and on day 2, cells were passaged and selected in medium containing 1 μg/mL puromycin for 7 days. To ensure at least 300-fold coverage of the CRISPR library, cells were maintained at a density of at least 3 × 107 cells during the puromycin treatment period. 1 μg of unmodified or N1-methylpseudouridinated EGFP mRNA was then formulated in LNPs to transfect 1.6 × 107 infected cells per dish, with a total of four dishes per replicate (totaling at least 6.4 × 107 cells) to ensure sufficient sgRNA coverage. One day after LNP-mRNA treatment of pooled knockout cells, cells were trypsinized, resuspended in PBS containing 3% FBS at a concentration of 1.2–1.5 × 10 7 cells/mL, and passed through a 35 μm cell strainer for FACS analysis using a BD FACS Aria III (BD Biosciences). Cells were isolated from the top 0–2.5% and 2.5–5% ranges with the highest GFP fluorescence intensity (GFPHigh), or the bottom 0–2.5% and 2.5–5% ranges with the lowest GFP fluorescence intensity (GFPLow), and sorting was performed until 6.25 × 10 5 cells were collected for each condition. 2.5 × 10 7 viable cells were collected as an unsorted control to ensure at least 300x coverage. In the GFP-stable cell line screening, only CRISPR infection was performed on HCT116 GFP-stable cells, and LNP delivery of EGFP mRNA was omitted.
세포 분리 후, 정렬된 세포와 비정렬 대조군 세포로부터의 유전체 DNA(gDNA)는 MasterPure Complete DNA and RNA Purification Kit (Lucigen, MC85200)를 사용하여 제조사의 프로토콜에 따라 추출하였다. 이후, 두 단계의 PCR 반응을 통해 시퀀싱 라이브러리를 제작하였으며, Herculase II Fusion DNA polymerase(Agilent, 600677)를 사용하여 기존 보고[Nature 542, 197-202 및 Science. 370(6523):eabc9546]와 동일한 방식으로 수행하였다. 비정렬 대조군의 경우, PCR 반응 1회당 100 μL 반응액에 6.6 μg의 gDNA를 사용하였으며, 총 24개의 PCR 반응을 수행하여 300배 커버리지를 확보하였다. 정렬된 상위 및 하위 GFP 형광 세포의 경우, 추출한 모든 gDNA를 나누어 각각 100 μL 반응으로 2회 PCR을 수행하였다. 1차 증폭에서는 보편적 forward 및 reverse primer를 사용하여 18회 사이클의 PCR을 수행하였다. 이후, 병합된 PCR 반응액 중 5 μL를 template로 하여, barcoded indexing primer를 사용한 50 μL 규모의 2차 PCR을 수행하였고, 이때 8~10회 확장 사이클을 적용하였다. PCR 생성물은 Agencourt AMPure XP (Beckman Coulter Life Sciences, A63880)를 사용하여 정제하였으며, 시퀀싱 라이브러리의 품질은 Agilent TapeStation(DNA ScreenTape)을 이용한 자동 전기영동 분석으로 확인하였다. NEBNext Library Quant Kit for Illumina (NEB, E7630L)를 사용하여 정량한 뒤, Illumina NovaSeq 6000 플랫폼으로 시퀀싱을 수행하였다. 사용된 primer 서열에 관한 정보는 표 3에 나타내었다.After cell separation, genomic DNA (gDNA) from sorted and unsorted control cells was extracted using the MasterPure Complete DNA and RNA Purification Kit (Lucigen, MC85200) according to the manufacturer's protocol. Subsequently, a two-step PCR reaction was performed to prepare a sequencing library using Herculase II Fusion DNA polymerase (Agilent, 600677) in the same manner as previously reported [Nature 542, 197-202 and Science. 370(6523):eabc9546]. For the unsorted control, 6.6 μg of gDNA was used per 100 μL reaction, and a total of 24 PCR reactions were performed to ensure 300-fold coverage. For the sorted upper and lower GFP fluorescent cells, all extracted gDNA was divided into two 100 μL reactions, and PCR was performed twice. In the first amplification, 18 cycles of PCR were performed using universal forward and reverse primers. Subsequently, 5 μL of the merged PCR reaction mixture was used as a template for a 50 μL second PCR using barcoded indexing primers, with 8–10 extension cycles. The PCR products were purified using Agencourt AMPure XP (Beckman Coulter Life Sciences, A63880), and the quality of the sequencing library was confirmed by automated electrophoresis using an Agilent TapeStation (DNA ScreenTape). After quantification using the NEBNext Library Quant Kit for Illumina (NEB, E7630L), sequencing was performed on the Illumina NovaSeq 6000 platform. Information on the primer sequences used is shown in Table 3.
번호number
시퀀싱 데이터 처리를 위해, 디멀티플렉싱된 FASTQ 파일로부터 각 리드당 20nt의 sgRNA 서열을 추출하였고, CRISPR 라이브러리 정보에 기반해 Addgene에서 구축한 sgRNA 참조 서열에 Bowtie 2를 이용하여 unique alignment 및 mismatch 허용 없음 조건으로 정렬하였다. 유일하게 정렬된 리드를 집계한 결과, 총 3×107개 이상의 시퀀싱 리드를 확보하였으며, 최소 300배의 sgRNA 커버리지를 달성하였다.For sequencing data processing, 20 nt sgRNA sequences per read were extracted from demultiplexed FASTQ files and aligned to the sgRNA reference sequence constructed by Addgene based on CRISPR library information using Bowtie 2 under unique alignment and no mismatch tolerance conditions. A total of more than 3× 107 sequencing reads were secured by counting the uniquely aligned reads, achieving at least 300x sgRNA coverage.
이후 sgRNA별 리드 카운트를 RPM(Rows Per Million) 방식으로 정규화한 뒤, MAGeCK 통계 분석 도구(v0.5.9.4)를 사용하여 분석을 수행하였다. 각 정렬 조건에서 비정렬 대조군 대비 유의하게 농축된 sgRNA는 CRISPR 라이브러리 내 비표적 sgRNA의 분포를 기준으로 판별하였다. 각 조건에 대한 MAGeCK 출력 결과와, 스크리닝의 유효성을 검토하기 위한 필수 유전자 표적 sgRNA 목록은 gene-level 통계로 정리하였다.After normalizing the read counts for each sgRNA using the RPM (Rows Per Million) method, analysis was performed using the MAGeCK statistical analysis tool (v0.5.9.4). Significantly enriched sgRNAs compared to the non-aligned control in each alignment condition were identified based on the distribution of off-target sgRNAs within the CRISPR library. The MAGeCK output results for each condition and the list of essential gene-targeting sgRNAs to verify the validity of the screening were organized using gene-level statistics.
참조예 7. Lipofectamine 기반 형질감염 및 저해제 처리Reference Example 7. Lipofectamine-based transfection and inhibitor treatment
Lipofectamine을 이용한 mRNA 형질감염은 제조사 지침에 따라 Lipofectamine MessengerMAX (Invitrogen, LMRNA015)를 사용하여 IVT mRNA를 다양한 농도로 세포에 1일간 처리하여 수행하였다. TRIM25의 외인성 발현이 필요한 rescue 실험, RNA pulldown, in vitro 유비퀴틴화 실험에서는 FuGENE HD 형질감염 시약(Promega, E2312)을 제조사 지침에 따라 사용하여 TRIM25 발현 플라스미드를 세포에 전달하였다.mRNA transfection using Lipofectamine MessengerMAX (Invitrogen, LMRNA015) was performed by treating cells with IVT mRNA at various concentrations for 1 day using Lipofectamine MessengerMAX (Invitrogen, LMRNA015) according to the manufacturer's instructions. For rescue experiments, RNA pulldown, and in vitro ubiquitination experiments requiring exogenous expression of TRIM25, the TRIM25 expression plasmid was delivered to cells using FuGENE HD transfection reagent (Promega, E2312) according to the manufacturer's instructions.
개별 유전자의 knockdown을 위해, 세포에 50 nM의 siRNA를 Lipofectamine RNAiMAX 시약(Invitrogen, 13778075)을 사용하여 제조사 지침에 따라 2일간 처리하였다. Jurkat 세포 및 mBMDM에서는 높은 knockdown 효율을 얻기 위해, siRNA를 전기천공 방식으로 도입하였으며, 구체적인 방법은 아래 Electroporation 항목에 기술되어 있다. 본 연구에 사용된 siRNA는 ON-TARGETplus SMARTpool siRNA(Dharmacon) 또는 Invitrogen Block-iT RNAi Designer(Invitrogen)를 이용해 개별 설계한 siRNA를 사용하였다. 음성 대조군으로는 ON-TARGETplus Non-targeting control(Dharmacon) 또는 AccuTarget Negative Control siRNA(Bioneer)를 사용하였다. 사용된 siRNA의 서열에 관한 정보는 표 4에 나타내었다.For individual gene knockdown, cells were treated with 50 nM siRNA using Lipofectamine RNAiMAX reagent (Invitrogen, 13778075) according to the manufacturer's instructions for 2 days. To achieve high knockdown efficiency in Jurkat cells and mBMDM, siRNA was introduced via electroporation, as described in the Electroporation section below. The siRNAs used in this study were either ON-TARGETplus SMARTpool siRNA (Dharmacon) or individually designed siRNAs using Invitrogen Block-iT RNAi Designer (Invitrogen). ON-TARGETplus Non-targeting Control (Dharmacon) or AccuTarget Negative Control siRNA (Bioneer) served as negative controls. The sequences of the siRNAs used are presented in Table 4.
Orthogonal loss-of-function 실험을 위해, HCT116 야생형 또는 GFP-stable 세포주에 LNP 포뮬레이션된 mRNA를 형질감염하면서, 세포 내 positive regulator 후보들에 대한 저해제를 1일간 처리하였다. EGFP mRNA를 이용한 농도 의존성 실험에서는 HCT116 야생형 세포에 대해 총 6가지 농도의 heparin(0, 0.1, 0.2, 0.5, 2, 10 μg/mL) 또는 Bafilomycin A1(0, 0.1, 0.2, 0.3, 0.5, 1 nM)을 처리하였다. Fluc mRNA를 GFP-stable 세포에 형질감염한 실험에서는 다음의 조건을 사용하였다: 0.2 μg/mL heparin(Sigma, H3393), 0.2 nM Bafilomycin A1(Sigma, 19-148), 20 nM Pitstop-2(Sigma, SML1169), 및 50 nM Dynole(Abcam, ab120463).For orthogonal loss-of-function experiments, HCT116 wild-type or GFP-stable cell lines were transfected with LNP-formulated mRNA, followed by treatment with inhibitors of intracellular positive regulator candidates for 1 day. In a concentration-dependent experiment using EGFP mRNA, HCT116 wild-type cells were treated with six concentrations of heparin (0, 0.1, 0.2, 0.5, 2, and 10 μg/mL) or Bafilomycin A1 (0, 0.1, 0.2, 0.3, 0.5, and 1 nM). The following conditions were used in experiments transfecting Fluc mRNA into GFP-stable cells: 0.2 μg/mL heparin (Sigma, H3393), 0.2 nM Bafilomycin A1 (Sigma, 19-148), 20 nM Pitstop-2 (Sigma, SML1169), and 50 nM Dynole (Abcam, ab120463).
참조예 8. ElectroporationReference Example 8. Electroporation
Electroporation은 Neon NxT System(Invitrogen, MPK5000)을 사용하여 제조사 지침에 따라 수행하였다. 도 46에서의 IVT mRNA 형질감염 실험을 위해, HCT116 세포를 트립신 처리한 후 DPBS로 세척하였다. 이후 세포를 5×106 cells/mL 농도로 R buffer(Invitrogen, MPK1096) 50 μL에 재부유시켰고, 여기에 Fluc IVT mRNA 100 ng을 첨가하여 잘 혼합하였다. 세포-RNA 혼합물 중 10 μL를 펄스당 사용하였으며, 샘플당 총 40 μL를 네 번의 펄스를 통해 처리하였다. 전기천공은 다음 조건으로 수행하였다: 1530 V, 20 ms, 1 펄스/10 μL. Electroporation 직후, 세포는 12-well plate의 1 mL pre-warmed McCoy’s 배지로 즉시 옮겼다.Electroporation was performed using the Neon NxT System (Invitrogen, MPK5000) according to the manufacturer's instructions. For the IVT mRNA transfection experiment in Figure 46, HCT116 cells were trypsinized and washed with DPBS. Cells were then resuspended in 50 μL of R buffer (Invitrogen, MPK1096) at a concentration of 5 × 10 6 cells/mL, and 100 ng of Fluc IVT mRNA was added and mixed well. Ten μL of the cell-RNA mixture was used per pulse, and a total of 40 μL was processed per sample over four pulses. Electroporation was performed under the following conditions: 1530 V, 20 ms, 1 pulse/10 μL. Immediately after electroporation, cells were immediately transferred to 1 mL of pre-warmed McCoy's medium in a 12-well plate.
도 23에서의 Jurkat 또는 mBMDM에서의 TRIM25 knockdown 실험에서는, 세포를 DPBS로 세척한 뒤 각각 2×107 cells/mL(Jurkat), 1.2×107 cells/mL(mBMDM)의 농도로 T buffer(Invitrogen, MPK10096)에 재부유시켰다. mBMDM의 경우, 재부유 전에 부드럽게 긁어서 수집하였다. 이후 siRNA를 최종 농도 1 μM이 되도록 첨가하여 잘 혼합하였다. 세포-siRNA 혼합물은 100 μL씩 electroporation에 사용하였고, 조건은 다음과 같다: Jurkat: 1350 V, 10 ms, 3 펄스, mBMDM: 1500 V, 20 ms, 1 펄스.In the TRIM25 knockdown experiment in Jurkat or mBMDM in Figure 23, cells were washed with DPBS and resuspended in T buffer (Invitrogen, MPK10096) at a concentration of 2 × 10 7 cells/mL (Jurkat) and 1.2 × 10 7 cells/mL (mBMDM), respectively. mBMDM were collected by gentle scraping before resuspension. siRNA was then added to a final concentration of 1 μM and mixed well. 100 μL of the cell-siRNA mixture was used for electroporation under the following conditions: Jurkat: 1350 V, 10 ms, 3 pulses; mBMDM: 1500 V, 20 ms, 1 pulse.
Electroporation 직후, Jurkat은 6-well plate에 1.9 mL의 RPMI 1640 배지에, mBMDM은 12-well plate에 2×105개씩 분주하여 1 mL의 L929 조건화 DMEM에 옮긴 뒤, 2시간 후 DMEM 1 mL를 추가하였다. 다음 날, siRNA가 도입된 세포에 LNP를 통해 mRNA를 형질감염하였다.Immediately after electroporation, Jurkat cells were seeded in 1.9 mL of RPMI 1640 medium in 6-well plates, and mBMDM cells were seeded in 2 × 105 cells in 12-well plates, transferred to 1 mL of L929-conditioned DMEM, and 1 mL of DMEM was added 2 h later. The following day, mRNA was transfected into the siRNA-transduced cells via LNP.
참조예 9. Flow cytometryReference Example 9. Flow cytometry
GFP-stable 세포 및 EGFP mRNA로 형질감염된 야생형 세포 등 살아있는 세포에서의 GFP 형광은 BD Accuri C6 Plus Flow Cytometer(BD Biosciences)를 사용하여 측정하였다. FACS 분리와 유사하게, 세포는 트립신 처리 후 재부유시키고, 35 μm cell strainer 캡이 포함된 튜브를 통해 여과하였다. 여과된 세포는 분석기에 주입하여 적절한 형광 채널(FITC-A)에서 살아있는 세포의 GFP 형광을 측정하였다.GFP fluorescence in live cells, including GFP-stable cells and wild-type cells transfected with EGFP mRNA, was measured using a BD Accuri C6 Plus Flow Cytometer (BD Biosciences). Similar to FACS separation, cells were trypsinized, resuspended, and filtered through a tube containing a 35 μm cell strainer cap. The filtered cells were injected into the analyzer, and GFP fluorescence in live cells was measured in the appropriate fluorescence channel (FITC-A).
참조예 10. Luciferase assayReference Example 10. Luciferase assay
Luciferase 분석은 세포를 1X passive lysis buffer로 용해한 후, 제조사 지침에 따라 Dual-Luciferase Reporter 1000 Assay System (Promega, E1910)를 사용하여 발광을 측정하였다. LNP-mRNA 실험에서는 Fluc 또는 Rluc를 발현하는 mRNA를 형질감염하였고, firefly luciferase 활성을 측정할 때는 Luciferase Assay Reagent II (LAR II)를, renilla luciferase 활성을 측정할 때는 LAR II와 Stop & Glo Reagent를 함께 사용하였다.Luciferase assays were performed by lysing cells with 1X passive lysis buffer and measuring luminescence using the Dual-Luciferase Reporter 1000 Assay System (Promega, E1910) according to the manufacturer's instructions. In LNP-mRNA experiments, mRNA expressing Fluc or Rluc was transfected. Luciferase Assay Reagent II (LAR II) was used for measuring firefly luciferase activity, and LAR II and Stop & Glo Reagent were used together for measuring renilla luciferase activity.
참조예 11. 정량적 실시간 PCR (RT-qPCR)Reference Example 11. Quantitative real-time PCR (RT-qPCR)
총 RNA는 RNeasy Mini Kit (Qiagen, 74106)를 사용하여 추출하였고, on-column RNase-free DNase (Qiagen, 79254)로 처리하였다. 이후, Superscript IV (Invitrogen, 18090200)를 사용하여 random hexamer primer로 역전사 반응을 수행하였다. 정량적 PCR은 Power SYBR Green PCR Master Mix (Applied Biosystems, A25778)를 사용하여 수행하였다. 사용된 primer 서열에 관한 정보는 표 5에 나타내었다.Total RNA was extracted using the RNeasy Mini Kit (Qiagen, 74106) and treated with on-column RNase-free DNase (Qiagen, 79254). Reverse transcription was then performed using Superscript IV (Invitrogen, 18090200) with random hexamer primers. Quantitative PCR was performed using Power SYBR Green PCR Master Mix (Applied Biosystems, A25778). Information on the primer sequences used is shown in Table 5.
참조예 12. 고해상도 poly(A) tail 분석 (Hire-PAT)Reference Example 12. High-resolution poly(A) tail analysis (Hire-PAT)
Hire-PAT 분석 및 모세관 전기영동 데이터의 신호 처리는 다음과 같이 수행하였다. 우선 총 RNA에 G/I tailing을 수행한 후, universal primer를 이용하여 역전사하였다. 형광 신호 검출을 위해 6-FAM으로 표지된 universal reverse primer 및 유전자 특이적 forward primer를 사용하여 PCR 증폭하였다. 사용된 primer 서열에 관한 정보는 표 6에 나타내었다.Hire-PAT analysis and signal processing of capillary electrophoresis data were performed as follows. First, total RNA was subjected to G/I tailing, followed by reverse transcription using a universal primer. For fluorescent signal detection, PCR amplification was performed using a 6-FAM-labeled universal reverse primer and a gene-specific forward primer. The primer sequences used are presented in Table 6.
참조예 13. 웨스턴 블롯팅Reference Example 13. Western Blotting
세포는 protease inhibitor(Millipore, 535140) 및 phosphatase inhibitor cocktail(AG Scientific, P-1518)이 포함된 0.5% NP-40 lysis buffer(20 mM Tris-HCl pH 7.5, 100 mM KCl, 0.2 mM EDTA, 0.5% NP-40) 또는 1X passive lysis buffer(Promega, E1910)로 용해하였고, 용해액의 단백질 농도는 BCA assay(Pierce, 23227)를 사용하여 측정하였다. Cells were lysed with 0.5% NP-40 lysis buffer (20 mM Tris-HCl pH 7.5, 100 mM KCl, 0.2 mM EDTA, 0.5% NP-40) containing protease inhibitor (Millipore, 535140) and phosphatase inhibitor cocktail (AG Scientific, P-1518) or 1X passive lysis buffer (Promega, E1910), and the protein concentration of the lysate was measured using the BCA assay (Pierce, 23227).
단백질 샘플은 Thermo Scientific의 단백질 마커(26616)와 함께 8-16% Novex Tris-Glycine 단백질 겔(Invitrogen, XP08162BOX)에 로딩한 뒤, 메탄올로 활성화한 PVDF 멤브레인(Millipore)으로 단백질을 전이하였다. 전이된 멤브레인은 5% 탈지우유 또는 BSA가 첨가된 PBS-T 용액으로 블로킹하였으며, 1차 항체와 HRP-conjugated 2차 항체를 이용해 면역탐지하였다. Chemiluminescence는 SuperSignal West Pico 또는 Femto 시약(Thermo Scientific, 34580 또는 34095)을 사용하여 수행하였으며, ChemiDoc XRS+ System(Bio-Rad)으로 신호를 검출하였다.Protein samples were loaded onto 8–16% Novex Tris-Glycine protein gels (Invitrogen, XP08162BOX) along with Thermo Scientific protein marker (26616), and then transferred to PVDF membranes (Millipore) activated with methanol. The transferred membranes were blocked with 5% skim milk or BSA-supplemented PBS-T solution, and immunoblotted with primary antibodies and HRP-conjugated secondary antibodies. Chemiluminescence was performed using SuperSignal West Pico or Femto reagents (Thermo Scientific, 34580 or 34095), and signals were detected with a ChemiDoc XRS+ System (Bio-Rad).
사용된 1차 항체는 다음과 같다: α-TRIM25 (Abcam, ab167154, 1:1000), α-Fluc (Invitrogen, PA5-32209, 1:1000), α-RIG-I (Cell Signaling, 3743S, 1:1000), α-eIF2α (Cell Signaling, 5324S, 1:1000), α-phospho-eIF2α (Cell Signaling, 3398S, 1:1000), α-PKR (Cell Signaling, 12297S, 1:1000), α-phospho-PKR (Thermo Scientific, MA5-38282, 1:500), α-Ubiquitin (Abcam, ab7254, 1:2000), α-hnRNPA1 (Santa Cruz, sc-32301, 1:1000), α-α-Tubulin (Abcam, ab52866, 1:1000), α-GAPDH (Santa Cruz, sc-32233, 1:1000).Primary antibodies used were as follows: α-TRIM25 (Abcam, ab167154, 1:1000), α-Fluc (Invitrogen, PA5-32209, 1:1000), α-RIG-I (Cell Signaling, 3743S, 1:1000), α-eIF2α (Cell Signaling, 5324S, 1:1000), α-phospho-eIF2α (Cell Signaling, 3398S, 1:1000), α-PKR (Cell Signaling, 12297S, 1:1000), α-phospho-PKR (Thermo Scientific, MA5-38282, 1:500), α-Ubiquitin (Abcam, ab7254, 1:2000), α-hnRNPA1 (Santa Cruz, sc-32301, 1:1000), α-α-Tubulin (Abcam, ab52866, 1:1000), α-GAPDH (Santa Cruz, sc-32233, 1:1000).
참조예 14. Polysome profilingReference Example 14. Polysome profiling
Polysome profiling은 다음과 같이 수행하였다. 100 μg/mL cycloheximide (CHX)(Sigma, C4859)가 포함된 10-50% sucrose gradient 용액(Acros Organics, AC419760050)은 Gradient Master(Biocomp, B108-2)를 사용하여 준비하였다. 프로파일링 전, LNP 포뮬레이션된 mRNA를 TRIM25 knockdown 또는 knockout HCT116 세포주에 1일간 처리하였다. 세포는 수확 전에 100 μg/mL CHX가 첨가된 냉 PBS로 수집하였고, 이후 CHX, protease inhibitor, phosphatase inhibitor, RNase inhibitor(Ambion, AM2696)가 포함된 polysome-extraction buffer (PEB: 50 mM Tris-HCl pH 7.5, 100 mM KCl, 5 mM MgCl₂, 0.5 mM TCEP, 0.5% NP-40)로 용해하였다.Polysome profiling was performed as follows. A 10–50% sucrose gradient solution (Acros Organics, AC419760050) containing 100 μg/mL cycloheximide (CHX) (Sigma, C4859) was prepared using Gradient Master (Biocomp, B108-2). Prior to profiling, LNP-formulated mRNA was treated for 1 day in TRIM25 knockdown or knockout HCT116 cells. Cells were collected with cold PBS containing 100 μg/mL CHX before harvesting, and then lysed with polysome-extraction buffer (PEB: 50 mM Tris-HCl pH 7.5, 100 mM KCl, 5 mM MgCl₂, 0.5 mM TCEP, 0.5% NP-40) containing CHX, protease inhibitor, phosphatase inhibitor, and RNase inhibitor (Ambion, AM2696).
수득된 용해액은 조심스럽게 sucrose gradient 위에 적층한 뒤, 초원심분리기(Beckman Coulter Ultracentrifuge Optima XE, SW41Ti rotor)를 사용하여 36,000 rpm에서 2시간 동안 원심분리하였다. 이후 분획된 샘플은 Bio-Rad EM-1 Econo UV detector를 통해 분석하고, Bio-Rad 7318303을 이용하여 1 mL씩 수집한 후 RT-qPCR 분석을 진행하였다.The obtained lysate was carefully layered onto a sucrose gradient and centrifuged at 36,000 rpm for 2 h using an ultracentrifuge (Beckman Coulter Ultracentrifuge Optima XE, SW41Ti rotor). The fractionated samples were then analyzed using a Bio-Rad EM-1 Econo UV detector, and 1 mL of each sample was collected using Bio-Rad 7318303 and subjected to RT-qPCR analysis.
RT-qPCR을 위해, 각 분획에서 수집한 125 μL(전체의 1/8 부피)로부터 RNA를 Direct-zol RNA Miniprep Kit(Zymo Research, R2052)를 사용해 추출하였다. 이때 정량화를 위한 spike-in RNA 1 ng을 첨가하였다. 추출한 RNA는 on-column DNase 처리 후 역전사 및 정량 PCR을 통해 각 분획 내 RNA 비율(%RNA portion)을 계산하였다.For RT-qPCR, RNA was extracted from 125 μL (1/8 total volume) collected from each fraction using the Direct-zol RNA Miniprep Kit (Zymo Research, R2052). 1 ng of spike-in RNA was added for quantification. The extracted RNA was treated with on-column DNase, reverse-transcribed, and quantitative PCR was performed to calculate the percentage of RNA (%RNA portion) in each fraction.
참조예 15. RNA 면역침강 분석 (RIP)Reference Example 15. RNA Immunoprecipitation Analysis (RIP)
RNA immunoprecipitation(RIP)은 다음과 같이 수행하였다. Salmon sperm DNA(Invitrogen, 15632011)로 전처리된 protein A sepharose 비드(GE Healthcare, 17-5138-01)에 정상 토끼 IgG(Cell Signaling, 2792S) 또는 TRIM25에 특이적인 monoclonal rabbit 항체(Abcam, ab167154)를 결합시켰다. RIP 실험에서는 m1Ψ 또는 U가 포함된 Fluc mRNA를 LNP 포뮬레이션하여 HCT116 세포에 형질감염하였고, 1일간 배양하였다. 이후 세포를 protease inhibitor, phosphatase inhibitor, RNase inhibitor가 포함된 0.2% NP-40 RIP buffer로 용해하였다. 용해액은 항체가 결합된 비드와 함께 4°C에서 2시간 동안 반응시킨 뒤, 총 5회 세척하였다. 마지막 세척 단계에서 일부 비드는 TRIM25 면역침강 여부를 확인하기 위한 western blotting용으로 따로 보관하였고, 나머지 비드에서 얻은 공침된 RNA 및 입력 샘플(input)은 TRIzol을 이용해 추출하였다. 이때 정량화를 위해 spike-in RNA 1 ng을 첨가하였다. 추출된 RNA는 역전사 후 RT-qPCR을 통해 정량하였으며, spike-in으로 정규화된 RNA 수준을 기준으로 입력 대비 상대적 농축도를 계산하였다.RNA immunoprecipitation (RIP) was performed as follows. Normal rabbit IgG (Cell Signaling, 2792S) or a monoclonal rabbit antibody specific for TRIM25 (Abcam, ab167154) was conjugated to protein A sepharose beads (GE Healthcare, 17-5138-01) pretreated with salmon sperm DNA (Invitrogen, 15632011). In RIP experiments, HCT116 cells were transfected with LNP formulations containing Fluc mRNA containing m1Ψ or U and cultured for 1 day. Cells were then lysed in 0.2% NP-40 RIP buffer containing protease inhibitors, phosphatase inhibitors, and RNase inhibitors. The lysates were incubated with antibody-conjugated beads at 4°C for 2 h and then washed five times. In the final washing step, some beads were reserved for Western blotting to confirm TRIM25 immunoprecipitation, while the coprecipitated RNA and input samples from the remaining beads were extracted using TRIzol. For quantification, 1 ng of spike-in RNA was added. The extracted RNA was reverse-transcribed and quantified by RT-qPCR, and the relative enrichment compared to the input was calculated based on the spike-in-normalized RNA level.
참조예 16. Recombinant TRIM25 정제Reference Example 16. Recombinant TRIM25 tablets
His10-eYFP-SUMOstar-Strep 태그가 N-말단에 부착된 재조합 TRIM25 단백질은 서스펜션 배양된 HEK293E 세포에서 이종 발현하였다. 세포는 약 7×105 cells/mL의 밀도로 일시적 형질감염을 하였으며, 0.5 L 배양 시 0.15 mg의 플라스미드 DNA와 1.5 mg의 선형 polyethylenimine (PEI)을 1% DMSO 존재 하에 혼합하여 사용하였다. 형질감염 후 세포는 33 °C에서 72시간 동안 배양하였다.Recombinant TRIM25 protein with a His10-eYFP-SUMOstar-Strep tag at its N-terminus was heterologously expressed in suspension-cultured HEK293E cells. Cells were transiently transfected at a density of approximately 7 × 10 5 cells/mL, and 0.15 mg of plasmid DNA and 1.5 mg of linear polyethylenimine (PEI) were mixed in the presence of 1% DMSO per 0.5 L culture. After transfection, cells were cultured at 33 °C for 72 h.
모든 정제 과정은 4 °C에서 수행하였다. 세포는 2,000 g로 10분간 원심분리하여 수확하였고, 냉 PBS로 세척한 후 buffer A(50 mM HEPES pH 8.0, 300 mM NaCl, 2 mM β-mercaptoethanol(BME))에 재부유시켰다. 이때 10% 글리세롤, EDTA-free protease inhibitor (Thermo Fisher Scientific, A32955), 20 μg/mL micrococcal nuclease, 5 mM CaCl₂를 첨가하였다. 세포는 초음파로 분해한 뒤 35,000 g에서 30분간 원심분리하였다. 상등액은 Ni-NTA Superflow resin(Qiagen, 1018142)에 로딩하였고, 20 mM imidazole(Sigma-Aldrich, I202)이 포함된 buffer A로 세척한 뒤, 200 mM imidazole이 포함된 buffer A로 단백질을 용출하였다. 이어서, SUMOstar protease(LifeSensors, 4110) 및 Benzonase nuclease(Sigma-Aldrich, E1014)와 함께 하룻밤 반응시켰다. 그 후, 시료는 Strep-Tactin Superflow resin(IBA Lifesciences, 4-4030-025)에 로딩한 뒤 buffer A로 세척하였고, 50 mM biotin(IBA Lifesciences, 2-1016-005)이 포함된 buffer A로 용출하였다. 용출된 단백질은 0.22 μm 멤브레인 필터를 이용하여 응집체를 제거한 뒤, HiPrep 26/10 Desalting column(GE Healthcare, 17-5087-01)을 사용하여 buffer 교환을 수행하였다. 최종 정제된 TRIM25 단백질은 5 mM Tris-HCl pH 7.0, 150 mM NaCl, 0.5 mM TCEP가 포함된 buffer에 평형시킨 후, 액체질소로 급속 동결하여 -80 °C에서 보관하였다.All purification procedures were performed at 4 °C. Cells were harvested by centrifugation at 2,000 g for 10 min, washed with cold phosphate-buffered saline (PBS), and resuspended in buffer A (50 mM HEPES, pH 8.0, 300 mM NaCl, 2 mM β-mercaptoethanol (BME)). 10% glycerol, EDTA-free protease inhibitor (Thermo Fisher Scientific, A32955), 20 μg/mL micrococcal nuclease, and 5 mM CaCl₂ were added. Cells were lysed by sonication and centrifuged at 35,000 g for 30 min. The supernatant was loaded onto Ni-NTA Superflow resin (Qiagen, 1018142), washed with buffer A containing 20 mM imidazole (Sigma-Aldrich, I202), and eluted with buffer A containing 200 mM imidazole. The mixture was then incubated overnight with SUMOstar protease (LifeSensors, 4110) and Benzonase nuclease (Sigma-Aldrich, E1014). The sample was then loaded onto Strep-Tactin Superflow resin (IBA Lifesciences, 4-4030-025), washed with buffer A, and eluted with buffer A containing 50 mM biotin (IBA Lifesciences, 2-1016-005). The eluted protein was filtered through a 0.22 μm membrane filter to remove aggregates, and then buffer exchange was performed using a HiPrep 26/10 Desalting column (GE Healthcare, 17-5087-01). The final purified TRIM25 protein was equilibrated in a buffer containing 5 mM Tris-HCl pH 7.0, 150 mM NaCl, and 0.5 mM TCEP, rapidly frozen in liquid nitrogen, and stored at -80 °C.
참조예 17. Poly(A)+ 또는 Biotin 표지 RNA Pulldown/Binding AssayReference Example 17. Poly(A)+ or Biotin-Labeled RNA Pulldown/Binding Assay
RNA pulldown 실험은 하기와 같이 수행하였다. 간단히, 3′ 말단에 30-nt poly(A)를 갖는 94-nt 길이의 UTR1 또는 UTR2 RNA를 T7 중합효소 기반 in vitro 전사를 통해 합성하였고, 이는 총 130-nt 길이의 RNA로 구성되며, 5′ 말단에 6-nt T7 프로모터 서열, UTR 본체(94-nt), 그리고 3′ 말단의 30-nt poly(A)로 구성된다. Poly(A)+ IVT RNA bait는 Oligo d(T)25 자성 비드(NEB, S1419S)에 25 °C, 1,400 rpm 조건으로 Thermomixer에서 1시간 반응시킨 후, 4 °C에서 하룻밤 동안 회전시켜 결합시켰다. Biotin 표지 RNA 실험의 경우, 말단에 biotin이 부착된 20-nt CALM1 UTR RNA(Dharmacon)를 합성하여, Streptavidin 자성 비드(Thermo Scientific, 88816)에 4 °C에서 하룻밤 회전 반응시켜 결합시켰다. 모든 실험에서는 Tris-HCl 기반 RNA pulldown buffer(50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 5% glycerol, 0.5 mM TCEP, 1% Triton X-100)를 사용하였으며, 단, 도 51에서는 pH 조절을 위해 Tris-HCl 대신 potassium phosphate buffer를 사용하였다. 모든 buffer는 실험 전 지정된 pH 조건에 맞추어 protease inhibitor, phosphatase inhibitor, RNase inhibitor를 첨가하였다. 사용된 RNA bait의 서열에 관한 정보는 표 7에 나타내었다.RNA pulldown experiments were performed as follows. Briefly, 94-nt long UTR1 or UTR2 RNAs with a 30-nt poly(A) at the 3′ end were synthesized through T7 polymerase-based in vitro transcription, resulting in a total RNA length of 130 nt, consisting of a 6-nt T7 promoter sequence at the 5′ end, the UTR body (94 nt), and a 30-nt poly(A) at the 3′ end. Poly(A)+ IVT RNA bait was bound to Oligo d(T)25 magnetic beads (NEB, S1419S) by incubation at 25 °C and 1,400 rpm in a thermomixer for 1 h, followed by overnight rotation at 4 °C. For biotin-labeled RNA experiments, 20-nt CALM1 UTR RNA (Dharmacon) with biotin attached to the terminal was synthesized and bound to Streptavidin magnetic beads (Thermo Scientific, 88816) by rotation overnight at 4 °C. In all experiments, Tris-HCl-based RNA pulldown buffer (50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 5% glycerol, 0.5 mM TCEP, 1% Triton X-100) was used, except that in Figure 51, potassium phosphate buffer was used instead of Tris-HCl for pH adjustment. Protease inhibitors, phosphatase inhibitors, and RNase inhibitors were added to all buffers according to the designated pH conditions before the experiment. Information on the sequences of the RNA baits used is shown in Table 7.
번호number
다음으로, 지정된 pH의 RNA pulldown buffer로 용해한 세포 용해액을 RNA가 결합된 비드와 함께 4 °C에서 2시간 반응시켰다. 이후 비드를 5회 세척하였고, 마지막 세척 단계에서 오염을 최소화하기 위해 새로운 단백질 low-bind 튜브(Eppendorf, Z666505)로 옮겼다. 침강된 단백질 시료는 50 mM TCEP(Thermo Scientific, 77720)이 포함된 샘플 버퍼(Bio-Rad, 161-0747)를 이용해 용출한 뒤, western blotting을 통해 분석하였다. 내인성 TRIM25 분석에는 HCT116 야생형 또는 TRIM25 knockout 세포를 사용하였고, 외인성 TRIM25 분석에는 TRIM25 knockout 세포에 GFP, TRIM25 야생형 또는 TRIM25 7KA RNA 결합 돌연변이 발현 플라스미드를 형질감염한 뒤 용해하였다.Next, cell lysates dissolved in RNA pulldown buffer at the indicated pH were reacted with RNA-bound beads at 4 °C for 2 hours. The beads were then washed five times and transferred to new protein low-binding tubes (Eppendorf, Z666505) to minimize contamination in the final wash step. The precipitated protein samples were eluted with sample buffer (Bio-Rad, 161-0747) containing 50 mM TCEP (Thermo Scientific, 77720) and analyzed by western blotting. For endogenous TRIM25 analysis, HCT116 wild-type or TRIM25 knockout cells were used, and for exogenous TRIM25 analysis, TRIM25 knockout cells were transfected with GFP, TRIM25 wild-type, or TRIM25 7KA RNA-binding mutant expression plasmids and then lysed.
재조합 TRIM25 단백질 실험에서는, 정제된 단백질을 RNA가 결합된 비드와 함께 4 °C에서 600 rpm 조건으로 Thermomixer에서 1시간 동안 반응시켰다. pH 의존적인 RNA 결합 친화성을 확인하기 위한 실험에서는 도 50에서는 4 °C에서 10분간, 도 51에서는 25 °C에서 2분간 반응하도록 제한하였다. 반응 후, 단백질이 결합된 비드는 3회 세척한 뒤, 50 mM TCEP(Thermo Scientific, 77720)이 포함된 샘플 버퍼(Bio-Rad, 161-0747)에 재현탁하였다. 이후 SDS-PAGE를 수행하고, InstantBlue Coomassie Protein Stain(Abcam, ab119211)으로 염색하여 분석하였다.In experiments with recombinant TRIM25 protein, purified protein was reacted with RNA-bound beads in a thermomixer at 4 °C and 600 rpm for 1 hour. For experiments to determine pH-dependent RNA binding affinity, the reaction was limited to 10 minutes at 4 °C in Figure 50 and 2 minutes at 25 °C in Figure 51. After the reaction, the protein-bound beads were washed three times and resuspended in sample buffer (Bio-Rad, 161-0747) containing 50 mM TCEP (Thermo Scientific, 77720). SDS-PAGE was then performed, and the samples were stained with InstantBlue Coomassie Protein Stain (Abcam, ab119211) for analysis.
참조예 18. In vitro 유비퀴틴화 분석Reference Example 18. In vitro ubiquitination assay
In vitro 유비퀴틴화 실험을 위해, FLAG 태그가 부착된 TRIM25 야생형, 7KA 또는 R54P 돌연변이 단백질을 HCT116 TRIM25 knockout 세포주에 외인성으로 발현시켰다. 플라스미드를 형질감염한 지 2일 후, 세포를 용해하고 α-FLAG M2 자성 비드(Millipore, M8823)와 반응시켜 FLAG-tagged TRIM25 단백질을 pull-down하였다.For in vitro ubiquitination experiments, FLAG-tagged TRIM25 wild-type, 7KA, or R54P mutant proteins were exogenously expressed in HCT116 TRIM25 knockout cells. Two days after plasmid transfection, cells were lysed and FLAG-tagged TRIM25 proteins were pulled down by reaction with α-FLAG M2 magnetic beads (Millipore, M8823).
단백질의 양은 SDS-PAGE 겔에서 BSA 기반 정량으로 측정하였으며, 단백질당 4.5 pmol에 해당하는 양의 비드를 in vitro 유비퀴틴화 반응에 사용하였다. 반응은 최종 부피 50 μL의 in vitro 유비퀴틴화 버퍼(50 mM Tris pH 7.5, 150 mM NaCl, 5 mM MgCl₂, 2 mM ATP, 0.5 mM TCEP)에 다음 구성 성분을 첨가하여 수행하였다: E1 효소: UBE1 125 ng (UBPBio, B1101), E2 효소: 6XHis-UbE2D3 250 ng (UBPBio, C1601), 유비퀴틴: 500 ng (UBPBio, E1100), m1Ψ 또는 U IVT RNA: 5 pmolThe amount of protein was measured by BSA-based quantification on SDS-PAGE gels, and beads corresponding to 4.5 pmol per protein were used for the in vitro ubiquitination reaction. The reaction was performed in a final volume of 50 μL of in vitro ubiquitination buffer (50 mM Tris pH 7.5, 150 mM NaCl, 5 mM MgCl₂, 2 mM ATP, 0.5 mM TCEP) containing the following components: E1 enzyme: UBE1 125 ng (UBPBio, B1101), E2 enzyme: 6XHis-UbE2D3 250 ng (UBPBio, C1601), ubiquitin: 500 ng (UBPBio, E1100), m1Ψ or U IVT RNA: 5 pmol
혼합물은 Thermomixer에서 1,300 rpm으로 흔들어주며 37 °C에서 30분간 반응시켰다. 반응은 50 mM TCEP가 포함된 샘플 버퍼를 첨가하고 가열 처리하여 종료하였으며, 이후 α-TRIM25 및 α-Ubiquitin 항체를 이용한 western blotting으로 분석하였다.The mixture was shaken at 1,300 rpm in a thermomixer and incubated at 37 °C for 30 min. The reaction was terminated by adding sample buffer containing 50 mM TCEP and heating, and then analyzed by Western blotting using α-TRIM25 and α-Ubiquitin antibodies.
참조예 19. ImagingReference Example 19. Imaging
내인성 G3BP1 및 TRIM25를 관찰하기 위한 이미징 실험에서는, LNP 형질감염 하루 전에 HCT116 야생형 세포를 지름 18 mm의 현미경용 커버글라스(Marienfeld, HSU-0111580)에 시드하였다. 다음 날, m1Ψ 또는 U가 포함된 Fluc mRNA를 LNP 포뮬레이션하여 두 가지 농도로 6시간 동안 형질감염하였다. 저농도는 10 ng/mL로 본 연구의 표준 조건이며, 고농도는 표준 조건의 50배인 500 ng/mL이다.For imaging experiments to observe endogenous G3BP1 and TRIM25, HCT116 wild-type cells were seeded on 18-mm diameter microscope coverslips (Marienfeld, HSU-0111580) one day before LNP transfection. The following day, Fluc mRNA containing m1Ψ or U was formulated into LNPs and transfected for 6 h at two concentrations. The low concentration was 10 ng/mL, which is the standard condition for this study, and the high concentration was 500 ng/mL, which is 50 times the standard condition.
음성 대조군으로는 LNP-mRNA 형질감염에 사용된 것과 동일한 부피의 PBS 또는 LNP만을 각각 처리하였다. G3BP focus 형성을 유도하는 스트레스 조건 대조군으로는, 세포에 0.5 mM sodium arsenite(NaAsO₂)(Sigma, S7400)를 45분간 처리하거나, poly(I:C) 2 μg/mL를 Lipofectamine RNAiMAX를 이용해 12시간 동안 형질감염하였다.As negative controls, cells were treated with PBS or LNP alone in the same volume as that used for LNP-mRNA transfection. As a stress condition control that induces G3BP focus formation, cells were treated with 0.5 mM sodium arsenite (NaAsO₂) (Sigma, S7400) for 45 min or transfected with 2 μg/mL of poly(I:C) for 12 h using Lipofectamine RNAiMAX.
표시된 처리가 완료된 후, 세포는 PBS로 한 번 세척한 뒤, PBS에 녹인 4% 메탄올 무함유 paraformaldehyde(EMS, 15714)로 30분간 고정하였다. 고정된 세포는 PBS로 세 번 세척한 뒤, 0.5% Triton X-100(Promega, H5141)이 포함된 PBS로 7분간 투과 처리하였다. 투과된 각 샘플은 0.1% Tween-20이 포함된 PBS(PBS-T)로 세척한 뒤, 1% BSA가 포함된 PBS로 1시간 동안 blocking하였다. 그 후, 1차 항체인 α-G3BP1(BD Bioscience, 611127, 1:200) 또는 α-TRIM25(Abcam, ab167154, 1:1000)를 4 °C에서 하룻밤 동안 반응시켰다. 항체 반응 후, 커버글라스는 PBS-T로 6회 세척하고, 해당 항체에 대응하는 Alexa Fluor 형광 2차 항체로 염색하였다: α-G3BP1에 대해 Alexa Fluor 488(α-Mouse, Invitrogen, A-21202, 1:400), α-TRIM25에 대해 Alexa Fluor 594(α-Rabbit, Invitrogen, A-21207, 1:400)을 사용하였다. 2차 항체 반응 후 PBS-T로 6회 세척한 뒤, 핵은 DAPI(VECTASHIELD® Antifade Mounting Medium with DAPI, Vector Laboratories, H-1200)로 염색하였다. 형광 신호는 현미경 슬라이드(Marienfeld, 1000612)에 장착된 샘플을 Nikon Eclipse Ti2 현미경으로 관찰하였고, 이미지는 ImageJ(v1.54g)를 이용해 분석하였다.After the indicated treatments were completed, cells were washed once with PBS and fixed with 4% methanol-free paraformaldehyde (EMS, 15714) in PBS for 30 min. The fixed cells were washed three times with PBS and then permeabilized with PBS containing 0.5% Triton X-100 (Promega, H5141) for 7 min. Each permeabilized sample was washed with PBS containing 0.1% Tween-20 (PBS-T) and blocked with PBS containing 1% BSA for 1 h. Then, the primary antibody, α-G3BP1 (BD Bioscience, 611127, 1:200) or α-TRIM25 (Abcam, ab167154, 1:1000), was reacted overnight at 4 °C. After antibody reaction, the coverslips were washed six times with PBS-T and stained with Alexa Fluor fluorescent secondary antibodies corresponding to the corresponding antibodies: Alexa Fluor 488 (α-Mouse, Invitrogen, A-21202, 1:400) for α-G3BP1 and Alexa Fluor 594 (α-Rabbit, Invitrogen, A-21207, 1:400) for α-TRIM25. After secondary antibody reaction, the coverslips were washed six times with PBS-T, and the nuclei were stained with DAPI (VECTASHIELD® Antifade Mounting Medium with DAPI, Vector Laboratories, H-1200). Fluorescent signals were observed with a Nikon Eclipse Ti2 microscope on samples mounted on microscope slides (Marienfeld, 1000612), and images were analyzed using ImageJ (v1.54g).
참조예 20. Gene Ontology (GO) enrichment 분석Reference Example 20. Gene Ontology (GO) enrichment analysis
FDR < 0.05의 유의미한 후보 유전자들에 대해 gene ontology enrichment 분석을 수행하기 위해 g:Profiler(v1.0.0)을 사용하였다. 후보 유전자의 알려진 생물학적 기능 및 세포 내 위치를 평가하기 위해 GO:BP(Biological Process) 및 GO:CC(Cellular Components) 항목을 기준으로 분석을 진행하였다.We used g:Profiler (v1.0.0) to perform gene ontology enrichment analysis on candidate genes with significant FDR < 0.05. To assess the known biological functions and subcellular localization of candidate genes, we analyzed them based on GO:BP (Biological Process) and GO:CC (Cellular Components) categories.
참조예 21. 단백질-단백질 상호작용 네트워크 분석Reference Example 21. Protein-protein interaction network analysis
LNP-mRNA의 핵심 positive regulator 후보 유전자에 대한 단백질-단백질 상호작용 네트워크 분석은 STRING (v12.0)을 사용하여 수행하였다. 네트워크에 포함된 노드 간 연결선은 STRING 소프트웨어에서 계산된 상호작용 근거의 강도를 나타내며, 기본 설정으로 시각화하였다. GFPLow 조건에서 신뢰할 수 있는 후보 유전자는 다음 두 가지 기준을 모두 만족하는 경우로 정의하였다: (1) 가장 엄격한 조건에서 FDR < 0.05, (2) 쌍으로 대응되는 binning 조건에서 상위 1%에 해당하는 유전자 (예: 0-2.5% GFPLow에서 FDR < 0.05이면서, 2.5-5% GFPLow에서 상위 1%에 해당하는 유전자). 네 가지 GFPLow 조건([m1Ψ 0-2.5% 및 2.5-5%], [U 0-2.5% 및 2.5-5%])에서 선별된 신뢰성 높은 후보 유전자들을 STRING 분석의 입력값으로 사용하여 단백질 상호작용 네트워크를 구성하였다.Protein-protein interaction network analysis of candidate genes for key positive regulators of LNP-mRNA was performed using STRING (v12.0). The connection lines between nodes in the network represent the strength of the interaction evidence calculated by the STRING software and were visualized with default settings. Reliable candidate genes under the GFPLow condition were defined as those that met both of the following two criteria: (1) FDR < 0.05 under the most stringent condition and (2) genes in the top 1% of pairwise binning conditions (e.g., genes with FDR < 0.05 at 0-2.5% GFPLow and in the top 1% at 2.5-5% GFPLow). High-confidence candidate genes selected under four GFPLow conditions ([m1Ψ 0-2.5% and 2.5-5%], [U 0-2.5% and 2.5-5%]) were used as inputs for STRING analysis to construct protein interaction networks.
전술한 본 발명의 설명은 예시를 위한 것이며, 본 발명이 속하는 기술분야의 통상의 지식을 가진 자는 본 발명의 기술적 사상이나 필수적인 특징을 변경하지 않고서 다른 구체적인 형태로 쉽게 변형이 가능하다는 것을 이해할 수 있을 것이다. 그러므로 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적이 아닌 것으로 이해해야만 한다.The foregoing description of the present invention is provided for illustrative purposes only. Those skilled in the art will readily appreciate that the present invention can be readily modified into other specific forms without altering the technical spirit or essential characteristics of the present invention. Therefore, the embodiments described above should be understood as illustrative in all respects and not restrictive.
Claims (14)
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2024-0068773 | 2024-05-27 | ||
| KR20240068773 | 2024-05-27 | ||
| KR10-2025-0011841 | 2025-01-24 | ||
| KR20250011841 | 2025-01-24 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025249887A1 true WO2025249887A1 (en) | 2025-12-04 |
Family
ID=97870873
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2025/007217 Pending WO2025249887A1 (en) | 2024-05-27 | 2025-05-27 | Composition for increasing stability or expression of exogenous rna in cells, comprising inhibitor of trim25 expression or activity |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR20250170532A (en) |
| WO (1) | WO2025249887A1 (en) |
-
2025
- 2025-05-27 KR KR1020250069193A patent/KR20250170532A/en active Pending
- 2025-05-27 WO PCT/KR2025/007217 patent/WO2025249887A1/en active Pending
Also Published As
| Publication number | Publication date |
|---|---|
| KR20250170532A (en) | 2025-12-05 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Napier et al. | ATRX represses alternative lengthening of telomeres | |
| WO2010090452A2 (en) | Small interference rna complex with increased intracellular transmission capacity | |
| WO2016021973A1 (en) | Genome editing using campylobacter jejuni crispr/cas system-derived rgen | |
| WO2010056043A2 (en) | Cell-penetrating, sequence-specific and nucleic acid-hydrolyzing antibody, method for preparing the same and pharmaceutical composition comprising the same | |
| WO2015053523A1 (en) | Bicistronic expression vector for antibody expression and method for producing antibody using same | |
| WO2017188797A1 (en) | Method for evaluating, in vivo, activity of rna-guided nuclease in high-throughput manner | |
| WO2018030874A1 (en) | Manipulated immunoregulatory element and immunity altered thereby | |
| Campos et al. | Mapping of pseudouridine residues on cellular and viral transcripts using a novel antibody-based technique | |
| WO2012115454A2 (en) | Method for concentrating cells that are genetically altered by nucleases | |
| WO2011081465A9 (en) | Pharmaceutical composition for suppression of vascularization | |
| WO2025249887A1 (en) | Composition for increasing stability or expression of exogenous rna in cells, comprising inhibitor of trim25 expression or activity | |
| WO2021225423A1 (en) | Novel nucleic acid ligand, and method for identifyng same | |
| WO2023043278A1 (en) | Method for evaluating efficacy of anticancer agent or screening anticancer agent | |
| WO2015126033A1 (en) | Composition for controlling stem cell pluripotency, containing lin28a methylation inhibitor, and method for screening for lin28a methylation inhibitor | |
| WO2024096642A1 (en) | Dna aptamer for diagnosis or treatment of colorectal cancer | |
| WO2023101505A1 (en) | Composition for inhibiting immune checkpoints comprising wnk3 inhibitor as active ingredient | |
| WO2020071641A1 (en) | Pharmaceutical composition for preventing or treating fabry disease, containing tsp1 protein inhibitor as active ingredient | |
| WO2011118994A2 (en) | Nlk as a marker for diagnosis of liver cancer and as a therapeutic agent thereof | |
| WO2022092995A2 (en) | Rna interactome capturing protocol and antiviral composition discovered using same | |
| WO2021145700A1 (en) | Cells having high adaptability under hypoxic conditions, and use thereof | |
| WO2024262942A1 (en) | Corona virus packaging signal element and use thereof | |
| WO2020242278A1 (en) | Method for inhibiting infection and activation of virus | |
| WO2021107747A1 (en) | Rna interference-inducing nucleic acid comprising 8-oxoguanine, modified nucleic acid binding to microrna comprising 8-oxoguanine, and uses thereof | |
| WO2021251656A1 (en) | Technique for diagnosing stress by using exosome-derived mirna | |
| WO2024005574A1 (en) | Method for screening regulatory element for increasing mrna translation, novel regulatory element resulting from method, and use thereof |