WO2022124760A1 - Composition for inducing or enhancing generation of cones or cone-like rods - Google Patents

Composition for inducing or enhancing generation of cones or cone-like rods Download PDF

Info

Publication number
WO2022124760A1
WO2022124760A1 PCT/KR2021/018460 KR2021018460W WO2022124760A1 WO 2022124760 A1 WO2022124760 A1 WO 2022124760A1 KR 2021018460 W KR2021018460 W KR 2021018460W WO 2022124760 A1 WO2022124760 A1 WO 2022124760A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cone
inducing
composition
nucleic acid
Prior art date
Application number
PCT/KR2021/018460
Other languages
French (fr)
Korean (ko)
Inventor
정혜원
채재병
장효익
Original Assignee
올릭스 주식회사
건국대학교 글로컬산학협력단
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by 올릭스 주식회사, 건국대학교 글로컬산학협력단 filed Critical 올릭스 주식회사
Publication of WO2022124760A1 publication Critical patent/WO2022124760A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0621Eye cells, e.g. cornea, iris pigmented cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/312Phosphonates
    • C12N2310/3125Methylphosphonates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/318Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
    • C12N2310/3181Peptide nucleic acid, PNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/40Nucleotides, nucleosides, bases

Definitions

  • the present invention relates to a composition for inducing or enhancing the production of cone cells or cone-like rod cells, and more particularly, to a composition comprising an asymmetric RNAi-inducing nucleic acid molecule that inhibits the expression of a Neural retina leucine zipper (NRL) gene. It relates to a composition for inducing or enhancing the production of cone cells or cone cell-like rod cells.
  • NRL Neural retina leucine zipper
  • Rod cells are very sensitive to light, so they can detect small amounts of light in dim conditions. Conversely, cone cells are less sensitive, so they can process large amounts of light during the day and continuously transmit visual signals. It is known that in many mammalian species, including mice and humans, the number of rods mediating vision under dim light greatly exceeds the number of cones. However, in an industrialized world where lighting forces cones to work all day, rod-mediated vision is less important. A number of patients have been identified who lack rod cell function from birth, and there is a report that they were virtually unable to recognize their abnormal visual acuity (Dryja, 2000). Conversely, when there is a dysfunction of the cone cells, the patient always shows symptoms and often suffers from visual handicap depending on the degree of their cone dysfunction.
  • cone cells are lost or dysfunction occurs, and rods remain relatively conserved.
  • achromatopsia a case of severe hereditary retinal dystrophy (retinal dystrophy) having a normal rod function despite the complete absence of cone function from birth has been reported (Hess et al, 1986) (Hess et al, 1986) ; Nishiguchi et al, 2005).
  • AMD age related macular degeneration
  • Korean Patent Laid-Open No. 10-2018-0012737 discloses a method for improving visual acuity by extending the function of rod cells using a nucleic acid encoding a gene product regulating endogenous photosensitive signal transduction in visual cells.
  • NRL transcriptional activator was identified in Swaroop et al. 266-270 (1992)). To date, there are various research results showing that NRL plays a very important function in the differentiation process of rod and cone cells.
  • the inventors of the present invention developed a nucleic acid molecule for inducing asymmetric RNAi that inhibits the expression of the NRL gene, and the nucleic acid molecule for inducing asymmetric RNAi is a cone cell or cone-like rod cell.
  • One object of the present invention is to provide a composition for inducing or enhancing the production of cones or cone-like rods, which includes a nucleic acid molecule for inducing asymmetric RNAi that suppresses the expression of NRL gene.
  • Another object of the present invention is to provide a nucleic acid molecule for inducing asymmetric RNAi or a pharmaceutical composition for improving eyesight, a pharmaceutical composition for improving or treating ocular or retinal diseases, or a composition for cell culture comprising the composition.
  • Another object of the present invention is to provide a method for inducing or enhancing the generation of cone cells or cone-like rod cells using the asymmetric RNAi-inducing nucleic acid molecule or the composition, or a method for improving or treating ocular or retinal diseases. do.
  • Another object of the present invention is the use of the nucleic acid molecule for inducing asymmetric RNAi or a composition comprising the same, for inducing or enhancing the production of cone cells or cone-like rods, for improving visual acuity, or for ocular or
  • An object of the present invention is to provide a use for improving or treating retinal diseases.
  • the present invention provides a composition for inducing or enhancing the production of cones or cone-like rods, comprising a nucleic acid molecule for inducing asymmetric RNAi that inhibits the expression of NRL gene.
  • the present invention also provides a nucleic acid molecule for inducing asymmetric RNAi or a pharmaceutical composition for improving eyesight, or a pharmaceutical composition for improving or treating ocular or retinal diseases, comprising the composition.
  • the present invention also provides a composition for cell culture for inducing or enhancing the production of the asymmetric RNAi-inducing nucleic acid molecule or a cone or cone-like rod comprising the composition.
  • the present invention also provides a method for inducing or enhancing the production of cone cells or cone-like rods using the nucleic acid molecule for inducing asymmetric RNAi or the composition.
  • the present invention also provides a method for improving or treating an ocular or retinal disease, comprising administering the nucleic acid molecule for inducing asymmetric RNAi or the composition to a subject.
  • the present invention also relates to the use of the nucleic acid molecule for inducing asymmetric RNAi or a composition comprising the same, for inducing or enhancing the production of cone cells or cone-like rods, for improving visual acuity, or for ocular or retinal diseases.
  • RNAi RNA interference
  • dsRNA double-stranded RNA
  • Nucleic acid molecules inducing RNAi refers to any nucleic acid molecule capable of inhibiting or down-regulating gene expression or viral replication by mediating said RNA interference in a sequence-specific manner.
  • the term may refer to both an individual nucleic acid molecule, a plurality of said nucleic acid molecules, or a pool of said nucleic acid molecules.
  • the nucleic acid molecule for inducing RNAi may be siRNA.
  • RNA Small interfering RNA
  • dsRNA short double-stranded RNA
  • an “antisense strand” is a polynucleotide substantially or 100% complementary to a target nucleic acid of interest, for example, messenger RNA (mRNA), a non-mRNA RNA sequence (e.g., microRNA, piwiRNA). , tRNA, rRNA, and hnRNA), or coding or non-coding DNA sequences, in whole or in part.
  • mRNA messenger RNA
  • non-mRNA RNA sequence e.g., microRNA, piwiRNA
  • tRNA, rRNA, and hnRNA coding or non-coding DNA sequences, in whole or in part.
  • ense strand is a polynucleotide having the same nucleic acid sequence as a target nucleic acid, wherein the mRNA (messenger RNA), non-mRNA RNA sequence (e.g., microRNA, piwiRNA, tRNA, rRNA, and hnRNA), or coding or refers to a polynucleotide identical in whole or in part to a non-coding DNA sequence.
  • mRNA messenger RNA
  • non-mRNA RNA sequence e.g., microRNA, piwiRNA, tRNA, rRNA, and hnRNA
  • coding refers to a polynucleotide identical in whole or in part to a non-coding DNA sequence.
  • a “gene” is to be taken in its broadest sense and may encode a structural protein or a regulatory protein.
  • the regulatory protein includes a transcription factor, a heat shock protein, or a protein involved in DNA/RNA replication, transcription, and/or translation.
  • the target gene to be suppressed is inherent in the viral genome, and may be integrated into an animal gene or exist as an extrachromosomal component.
  • the gene of interest may be a gene on the HIV genome.
  • the siRNA molecule is useful for inactivating translation of an HIV gene in a mammalian cell.
  • NRL Neuronal retina leucine zipper
  • Cone cell refers to photoreceptor cells in the retina of the eye, which are densely packed in the center of the macula even in the retina and have a plump shape.
  • the cone cells can sense bright light of about 0.1 Lux or more, can distinguish colors, and are also referred to as cone cells.
  • Rod cell refers to a photoreceptor cell in the retina of the eye, which is widely distributed in the periphery of the retina and has an elongated shape sensitive to weak light.
  • the rod cells sense the light and shade and the shape of the object, but cannot distinguish the color, and can sense a weak light of about 0.1 Lux or less.
  • Cone cell-like rod cell refers to a rod cell having the properties of a cone cell due to loss or reduction in the original properties of the rod cell.
  • the cone cell-like rod cells may be generated during the production process of rod cells, or may be generated when completely generated rod cells change their properties and are converted into cone cell-like rod cells.
  • Color blindness refers to a genetic trait that does not properly distinguish colors due to abnormalities in the photoreceptor cells of the retina, and is also called color vision abnormality. In many cases, color blindness is due to congenital causes, but it can also be acquired. Most color blindness is caused by abnormalities in the cone cells that can distinguish colors. Specifically, in patients with complete color blindness (achromatopsia), the case of severe hereditary retinal dystrophy (retinal dystrophy) with a complete absence of cone function from birth but normal rod function has been reported (Hess et al, 1986; Nishiguchi et al, 2005). Therefore, there is a possibility to improve or treat the symptoms or diseases of color blindness by inducing or enhancing the production of cone cells or cone cell-like rod cells.
  • the nucleic acid molecule for inducing RNAi may be characterized in that the sense strand has a length of 15 to 17 nt and the antisense strand has a length of 16 nt or more.
  • the antisense strand may be characterized as having a length of 16 to 31 nt, preferably having a length of 19 to 26 nt. More preferably, the length of the sense strand is 16 nt, and the length of the antisense strand complementary thereto may be characterized in that it is 19 nt, 24 nt, 25 nt, or 26 nt, but is not limited thereto.
  • the 3' end of the sense strand and the 5' end of the antisense strand form a blunt end.
  • the 3' end of the antisense strand may include, for example, an overhang of 1 to 16 nt.
  • the sense strand or the antisense strand of the nucleic acid molecule for inducing RNAi may include one or more chemical modifications.
  • siRNA cannot pass through the cell membrane for reasons such as high negative charge and high molecular weight due to the phosphate backbone structure, and it is rapidly degraded and removed from the blood, so it is difficult to deliver a sufficient amount for RNAi induction to the actual target site.
  • many high-efficiency delivery methods using cationic lipids and cationic polymers have been developed, but in vivo, it is difficult to deliver siRNA as high as in vitro, and it is difficult to There is a problem in that the siRNA delivery efficiency is reduced due to the interaction.
  • the present inventors introduced a chemical modification to the asymmetric siRNA structure to develop an asiRNA construct (cp-asiRNA) with self-delivery ability that can be effectively and intracellularly delivered without a separate carrier.
  • the chemical modification in the sense strand or the antisense strand may include one or more selected from the group consisting of:
  • -OH group at the 2' carbon position of the sugar structure in the nucleotide is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl-O-propyl ( propyl), -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; replacement of oxygen in the nucleotide structure with sulfur; nucleotide linkages are modified with phosphorothioate, boranophosphate, or methyl phosphonate; transformation into peptide nucleic acid (PNA), locked nucleic acid (LNA), or unlocked nucleic acid (UNA) form; and a phosphate group, a lipophilic compound, or a cell penetrating peptide bond.
  • PNA peptide nucleic acid
  • LNA locked nucleic acid
  • UNA unlocked nucle
  • the lipophilic compound is cholesterol, tocopherol, stearic acid, retinoic acid, DHA (docosahexaenoic acid), palmitic acid, linoleic acid ), linolenic acid, and any one or more selected from the group consisting of long-chain fatty acids having 10 or more carbon atoms.
  • it may be characterized as cholesterol, but is not limited thereto.
  • the sense strand may comprise one or more chemical modifications selected from: 2 to 4 nucleotide bonds adjacent to the 3' end are phosphorothioate, boranophosphate, or modified with methyl phosphonate; -OH group at the 2' carbon position of the sugar structure within 2 or more nucleotides is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl-O substituted with -propyl, -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; and binding of a lipophilic compound or a cell penetrating peptide to the 3' end.
  • the antisense strand may contain any one or more chemical modifications selected from the following: 3 to 5 nucleotide bonds adjacent to the 3' end are phosphorothioate, boranophosphate ), or modified with methyl phosphonate; -OH group at the 2' carbon position of the sugar structure within 2 or more nucleotides is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl -O substituted with -propyl, -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; and binding of a phosphate group or a cell penetrating peptide to the 5' end.
  • the -OH group at the 2' carbon position of the sugar structure within two or more nucleotides of the sense strand or the antisense strand is replaced with -OCH 3 (methoxy) or -F (fluorine) being transformed; at least 10% of the nucleotide linkages in the sense strand or the antisense strand are modified to phosphorothioate; cholesterol binding to the 3' end of the sense strand; and one or more modifications selected from the group consisting of a phosphate group bond to the 5' end of the antisense strand.
  • Exemplary nucleic acid molecules for inducing RNAi may include the following sense strands:
  • exemplary nucleic acid molecules for inducing RNAi may include the following antisense strands:
  • * is a phosphorothioate bond (phosphorothioated bond) modification
  • m is 2'-O-methyl (Methyl) substitution
  • 2'-F- is 2'-Fluoro substitution
  • -chol denotes a bond with 3'-cholesterol
  • P denotes a bond with a 5'-phosphate group.
  • one to three phosphate groups may be bonded to the 5' end of the antisense strand, but the present invention is not limited thereto.
  • the nucleic acid molecule for inducing RNAi may be characterized in that it inhibits the expression of the NRL gene.
  • the nucleic acid molecule for inducing RNAi may be characterized in that it inhibits the expression of the NRL gene by complementary binding to the mRNA encoding the NRL.
  • the NRL gene for example, the mRNA encoding the NRL may include a target sequence targeted by the RNAi-inducing nucleic acid molecule.
  • the present invention relates to a composition for inducing or enhancing the production of cones or cone-like rods comprising the nucleic acid molecule for inducing RNAi.
  • the RNAi-inducing nucleic acid molecule can induce a decrease in the number of rod cells and an increase in the number of cone cells among photoreceptor cells.
  • the RNAi-inducing nucleic acid molecule can suppress the expression of the NRL gene, and thus, the original properties of rod cells are lost or reduced in the process of rod cell generation, and thus cone cell-like properties with cone-like properties. It can induce the production of rod cells.
  • the RNAi-inducing nucleic acid molecule can inhibit the expression of the NRL gene, thereby changing the properties of previously generated rod cells to those of cone cells, thereby converting rod cells into cone-like rod cells.
  • RNAi-inducing nucleic acid molecule can inhibit the expression of the NRL gene, thereby inducing a decrease in the number of rod cells and an increase in the number of cone cells.
  • the decrease in the number of rod cells may be due to reduced differentiation into rod cells or promotion of rod cell death.
  • the increase in the number of the cone cells may be due to promotion of differentiation or proliferation into cone cells.
  • the composition comprising the RNAi-inducing nucleic acid molecule can induce or enhance the production of cone cells or cone-like rod cells.
  • the composition can inhibit the expression of the NRL gene, whereby the original properties of the rod cells are lost or reduced in the process of generating rod cells, thereby preventing the production of cone-like rod cells having cone-like properties. induce; converting rod cells into cone-like rod cells by changing the properties of previously generated rod cells to those of cone cells; It can induce an increase in the number of cone cells themselves.
  • the RNAi-inducing nucleic acid molecule or a composition comprising the same induces or enhances the production of cone cells or cone-like rod cells in a state in which cone cells are lost or dysfunctional, thereby supplementing the dysfunction of cone cells. Since it can contribute, it can be utilized as an active ingredient of a pharmaceutical composition for improving eyesight, or a pharmaceutical composition for improving or treating eye or retinal diseases.
  • the present invention relates to a pharmaceutical composition for improving eyesight comprising the RNAi-inducing nucleic acid molecule, and inducing or enhancing the production of cone cells or cone-like rod cells.
  • the present invention relates to a pharmaceutical composition for improving or treating ocular or retinal diseases, including the RNAi-inducing nucleic acid molecule, and inducing or enhancing the production of cone cells or cone-like rod cells.
  • the ocular or retinal disease is a condition in which cone cells are lost or malfunctioning, by inducing or enhancing the production of cone cells or cone cell-like rod cells, thereby compensating or supplementing the functional abnormality of cone cells. It may include any disease that can be treated. Preferably, the ocular or retinal disease may be a disease in which cone cells are lost or malfunctioning, while rod cells are normally present.
  • the ocular or retinal disease may be a disease accompanying color blindness or color blindness.
  • the color blindness may be congenital or acquired.
  • the color blindness may be complete color blindness, partial color blindness, or color weakness.
  • the complete color blindness refers to a case in which cone cells that discriminate colors do not exist in the retina at all in achromatopsia, and refers to a symptom or disease that cannot distinguish colors at all, unlike other color blindness.
  • the partial color blindness is caused by a lack of cone cells that recognize the three primary colors of red, green, and blue in the retina, and refers to a symptom or disease in which some colors cannot be distinguished.
  • the partial color blindness may be at least one selected from the group consisting of a first color vision or more, a second color vision or more, and a third color vision or more.
  • the first color vision abnormality refers to a symptom or disease in which there is no L cone cell ( ⁇ cell) for recognizing red, red and green cannot be distinguished, and red is recognized as a dark color.
  • the second color vision abnormality does not have M cone cells ( ⁇ cells) that recognize green and cannot distinguish red from green like the first color vision abnormality, but in this case, unlike the first color vision abnormality, green is recognized as a dark color. means a symptom or disease.
  • the third color vision abnormality refers to a symptom or disease in which S cone cells ( ⁇ cells) that recognize blue do not exist, cannot distinguish between blue and yellow, and recognize blue as dark green and yellow as pale red.
  • the color weakness refers to a symptom or disease in which a low-saturation color cannot be recognized or distinguished in a short time.
  • the pharmaceutical composition may be prepared by including one or more pharmaceutically acceptable carriers in addition to the nucleic acid molecule for inducing RNAi as an active ingredient.
  • a pharmaceutically acceptable carrier must be compatible with the active ingredient of the present invention, and saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or two or more of these ingredients may be used by mixing, and other conventional additives such as antioxidants, buffers, and bacteriostats may be added as needed.
  • diluents such as an aqueous solution, suspension, emulsion, or the like.
  • an injectable formulation such as an aqueous solution, suspension, emulsion, or the like.
  • a method commonly known in the art to which the present invention pertains may be used, and a stabilizer for freeze-drying may be added.
  • the method of administering the pharmaceutical composition may be determined by a person skilled in the art based on the symptoms of the patient and the severity of the disease.
  • it may be formulated in various forms such as powders, tablets, capsules, liquids, injections, ointments, syrups, etc., and may be provided in unit-dose or multi-dose containers, such as sealed ampoules and bottles. .
  • the pharmaceutical composition of the present invention can be administered orally or parenterally.
  • the route of administration of the composition according to the present invention is not limited thereto, but for example, oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intestinal, sublingual , or topical administration is possible.
  • the dosage of the composition according to the present invention varies according to the patient's weight, age, sex, health status, diet, administration time, method, excretion rate, or severity of disease, etc., and a person of ordinary skill in the art can be easily determined.
  • the composition of the present invention may be formulated into a suitable dosage form for clinical administration using known techniques.
  • the present invention relates to a cell culture composition for inducing or enhancing the production of the RNAi-inducing nucleic acid molecule or cone cells or cone-like rod cells comprising the composition.
  • the cell culture composition comprises culturing any one or more cells selected from the group consisting of stem cells, retinal progenitor cells, retinal organoid cells, photoreceptor progenitor cells, cone cells, or rod cells to obtain cone cells or cone cells. - It is for inducing or enhancing the production of rod-like cells, and may be provided in the form of a medium composition or a medium raw material composition.
  • the stem cells may be induced pluripotent stem cells (iPS cells).
  • the retinal precursor cells may refer to cells just before stem cells are differentiated into retinal cells.
  • the retinal organoid cells may refer to cells in which stem cells are grown in vitro to implement a tissue such as a human retinal structure.
  • the photoreceptor progenitor cells may refer to cells just before stem cells are differentiated to become photoreceptor cells, that is, cone cells or rod cells.
  • the composition for cell culture is a medium containing components capable of differentiating or proliferating cells, but is not limited thereto, but is not limited thereto, but is not limited thereto, but is not limited thereto, but is not limited thereto, but is DMEM (Dulbecco's Modified Eagle's Medium), 80% knockout DMEM, MEM (Minimal Essential Medium), BME (Basal).
  • DMEM Dulbecco's Modified Eagle's Medium
  • MEM Minimum Essential Medium
  • BME Base
  • composition for cell culture is 20% knockout serum replacer (Gibco), glutamine, mercaptoethanol, non-essential amino acids, basic fibroblast growth factor (bFGF), vitronectin recombinant human protein (Vitronectin Recombinant Human Protein) ), N2 Supplement, heparin, fetal bovine serum, taurine, glutamax, blebbistatin, and the like may be additionally included.
  • the composition for cell culture for inducing or enhancing the production of the cone cells or cone cell-like rod cells contains about 0.01 to 100 ⁇ M, about 0.01 to 50 ⁇ M, about 0.01 to 10 ⁇ M, about the RNAi-inducing nucleic acid molecule. It may be included in a concentration of 0.01 to 5 ⁇ M, about 0.01 to 1 ⁇ M, about 0.01 to 0.5 ⁇ M, about 0.05 to 0.5 ⁇ M, or about 0.1 to 0.2 ⁇ M.
  • composition for cell culture for inducing or promoting the production of the cone cells or cone cell-like rod cells contains the RNAi-inducing nucleic acid molecule at a concentration outside the numerical range, cone cells or The effect of inducing or enhancing the production of cone-like rods may be reduced.
  • the present invention relates to a method for inducing or enhancing the production of cone cells or cone-like rod cells using the RNAi-inducing nucleic acid molecule or the composition.
  • the method may be performed in vitro.
  • the RNAi-inducing nucleic acid molecule or the composition is added to any one or more cell culture medium selected from the group consisting of stem cells, retinal progenitor cells, retinal analogue cells, photoreceptor progenitor cells, cone cells, and rod cells in vitro. to do; Or in vitro, any one or more cells selected from the group consisting of stem cells, retinal progenitor cells, retinal analog cells, photoreceptor progenitor cells, cone cells, or rod cells in a medium containing the RNAi-inducing nucleic acid molecule or the composition It may include the step of culturing.
  • the RNAi-inducing nucleic acid molecule in the step of adding the RNAi-inducing nucleic acid molecule or the composition to the cell culture medium, the RNAi-inducing nucleic acid molecule is added to the cell culture medium at about 0.01 to 100 ⁇ M, about 0.01 to 50 ⁇ M, about 0.01 It may be added at a concentration of 10 ⁇ M to 10 ⁇ M, about 0.01 to 5 ⁇ M, about 0.01 to 1 ⁇ M, about 0.01 to 0.5 ⁇ M, about 0.05 to 0.5 ⁇ M, or about 0.1 to 0.2 ⁇ M.
  • the RNAi-inducing nucleic acid molecule in the step of culturing the cell in a medium containing the RNAi-inducing nucleic acid molecule or the composition, is added to the cell culture medium at about 0.01 to 100 ⁇ M, about 0.01 to 50 It may be included in a concentration of ⁇ M, about 0.01 to 10 ⁇ M, about 0.01 to 5 ⁇ M, about 0.01 to 1 ⁇ M, about 0.01 to 0.5 ⁇ M, about 0.05 to 0.5 ⁇ M, or about 0.1 to 0.2 ⁇ M.
  • the effect of inducing or enhancing the production of cone cells or cone-like rods may decrease.
  • the present invention relates to a method of improving or treating an ocular or retinal disease, comprising administering the RNAi-inducing nucleic acid molecule or the composition to a subject.
  • the present invention provides a nucleic acid molecule for inducing RNAi or a composition comprising the same for inducing or enhancing the production of cone cells or cone-like rod cells, use for improving visual acuity, or ocular or It relates to use for improving or treating retinal diseases.
  • a cone cell or cone cell-like rod cell is generated using a nucleic acid molecule for inducing asymmetric RNAi that can efficiently suppress the expression of the NRL gene, which plays a very important function in the differentiation process of rod cells and cone cells. can induce or promote
  • the nucleic acid molecule for inducing asymmetric RNAi may be usefully utilized as a therapeutic agent capable of improving vision or improving or treating ocular or retinal diseases by supplementing the loss or dysfunction of cone cells.
  • rhodosin Rhodosin
  • L/M-opsin L/M-opsin antibodies
  • FIG. 1 a portion expressed in a relatively low brightness indicates a cell nucleus stained by Hoechst 33342, and a portion expressed in a relatively high brightness indicates a cell fluorescently stained by a rhodopsin antibody or an L/M-opsin antibody.
  • FIG. 2 is a schematic diagram of each photoreceptor cell fluorescently stained with rhodopsin antibody and L/M-opsin antibody from immunofluorescent staining images for retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. It is a graph showing the analysis result.
  • Figure 3 shows rhodopsin, L/M-opsin, and S-opsin, which are proteins extracted from the retinal analogs treated with cp-asiNRL and the control retinal analogs not treated with cp-asiNRL, Western blot analysis. It is a drawing showing the result.
  • Example 1 Preparation of a nucleic acid molecule for inducing asymmetric RNAi having cell-penetrating ability to target NRL
  • cp-asiNRL which is a nucleic acid molecule for inducing asymmetric RNAi, which has cell-penetrating ability to target NRL, has already been disclosed in WO2020/179702, and its preparation was also prepared as disclosed in the same patent document.
  • Table 1 shows the base sequence of asiNRL targeting NRL.
  • 2'-F- means, for example, in the case of 2'-F-G, the 2'-OH of the existing G (guanine) is substituted with fluorine (Fluoro), and "-Chol” denotes a form in which cholesterol is added to the 3'-terminus, and "P” denotes a form in which the 5'-terminus is phosphorylated (eg, phosphate modification or phosphorothioate modification is added to the 5' end of the antisense strand) will do
  • Cholesterol attached to the 3' end of the sense strand allows it to penetrate the cell membrane.
  • substitution of phosphorothioate for the phosphate backbone close to the 3' end of the sense and antisense strands increases resistance to exohydrolase, cellular uptake and in vivo bioavailability.
  • resistance to nucleases is increased, and siRNA immunogenicity and off-target effect are reduced.
  • the 2' modification of some sugars with fluorine stabilizes the double-stranded RNA duplex, increases stability in seurm, and enables efficient silencing in vitro and in vivo.
  • RISC RNA-induced silencing complex
  • cp-asiNRL a nucleic acid molecule for inducing asymmetric RNAi having cell-penetrating ability to target NRL, was obtained in this example.
  • iPS cells Induced pluripotent stem cells
  • iPS cells were prepared from mononuclear cells isolated from the blood of a 43- or 62-year-old normal human. After culturing about 0.5 x 10 6 mononuclear cells in StemSpanTM SFEM II (Stemcell Technologies) medium containing SFEM II Supplement (Stemcell Technologies) for about 7 days, plasmid pCXLE-hOCToct3/4-shp53 with reprogramming genes inserted -F, pCXLEe-hSK, and pCXLEe-hUL, and pCXLEe-hOEBNA1, which promote gene expression, were injected into cells using electroporation.
  • a reprogramming medium (StemFit ® Basic02, AJINOMOTO) in a culture dish coated with i-MATIRX 511 (Takara), and induced pluripotent stem cell colony was formed.
  • the stem cell culture medium (Essential 8TM Medium, ThermoFisher scientific) was replaced and cultured for about 10 to 12 days, and then the cell population was collected.
  • the collected induced pluripotent stem cells were passaged about 20 to 24 times in a culture dish coated with Vitronectin Recombinant Human Protein (ThermoFisher scientific), and then used to prepare organoids.
  • the embryoid bodies of about 20 pieces/cm 2 were cultured in a culture dish coated with MatrigelTM GFR Membrane Matrix (Corning) together with a nerve induction medium for about 15 days. About 13 to 15 days after the culture in which the neural structure is formed, it was replaced with a neural retinal differentiation medium (DMEM/F12 (3:1), B27, MEM Non-Essential Amino Acids Solution) and cultured. , Cell bodies showing neural retinal structures were collected about 25 to 30 days after culture. After culturing the collected cell bodies in an ultra-low attachment dish containing neural retina medium for about 5 days, the medium was cultured with about 10% Fetal bovine serum, about 100 ⁇ M Taurine, and 1X Glutamax. (Glutamax) was replaced with the added neural retinal medium and then cultured for about 160 days to obtain a retinal analogue.
  • DMEM/F12 3:1
  • B27 MEM Non-Essential Amino Acids Solution
  • cp-asiNRL was added to a culture medium containing retinal analogues at about 48 hour intervals. was treated about 3 times. At this time, it was treated so that the final concentration of cp-asiNRL contained in the culture medium containing the retinal analogue was about 0.1 or 0.2 ⁇ M. About 30 retinal analogues were used to evaluate the effectiveness of cp-asiNRL.
  • tissue sections were fixed in about 4% PFA for about 15 minutes, softened in about 0.5% Triton X-100, washed with PBS, and then washed with about 2% bovine serum albumin. (bovine serum albumin) (Gibco, Dublin, Ireland) and about 5% normal goat serum (Jackson Immunoresearch, PA, USA) were blocked at about 4°C for about 12 hours. Thereafter, the tissue sections were reacted with rabbit rhodopsin (Millipore) and mouse L/M opsin (Millipore) antibodies at about 4° C. for about 12 hours, and then washed with PBS about 3 times. .
  • bovine serum albumin rabbit rhodopsin
  • mouse L/M opsin mouse L/M opsin
  • the protein was extracted from the retinal analog using RIPA buffer. Equal amounts of protein (about 20 ⁇ g) were separated by SDS-PAGE (Sodium Dodecyl Sulfate-Poly Acrylamide Gel Electrophoresis) and blotted with polyvinylidene fluoride (PVDF) membrane (Millipore, Billerica, MA).
  • SDS-PAGE Sodium Dodecyl Sulfate-Poly Acrylamide Gel Electrophoresis
  • PVDF polyvinylidene fluoride
  • the membrane immunoactive protein was chemically reacted using a luminescence substrate (ClarityTM Western ECL Substrate; Bio-Rad Laboratories Inc, Hercules, CA), and the results were recorded with a digital imaging system (ChemiDoc Touch Imager; Bio -Rad) was used for visualization.
  • a luminescence substrate ClarityTM Western ECL Substrate
  • Bio-Rad Laboratories Inc Hercules, CA
  • a digital imaging system ChemiDoc Touch Imager; Bio -Rad
  • the retinal analogues prepared from induced pluripotent stem cells were treated with cp-asiNRL at a concentration of about 0.1 ⁇ M.
  • the cp-asiNRL treatment was performed at about 142 days after culturing the retinal analog, which is a time point at which rod cells and cone cells were already formed (refer to Reference Example 1-2 above).
  • About 18 days after cp-asiNRL administration (about 160 days after culturing retinal analogues), the production patterns of rods and cones were analyzed using immunofluorescence staining.
  • Cells fluorescently stained with rhodopsin antibody in retinal analogues represent rod cells, and cells fluorescently stained with L/M-opsin antibody represent cone cells.
  • retinal analogues not treated with cp-asiNRL were used as a control. Specific test materials and test methods were described in detail in the reference example above.
  • a portion expressed in a relatively low brightness indicates a cell nucleus stained by Hoechst 33342
  • a portion expressed in a relatively high brightness indicates a cell fluorescently stained by a rhodopsin antibody or an L/M-opsin antibody.
  • FIG. 2 is a schematic diagram of each photoreceptor cell fluorescently stained with rhodopsin antibody and L/M-opsin antibody from immunofluorescent staining images for retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. It is a graph showing the analysis result.
  • the number of rod cells stained by the rhodopsin antibody in the retinal analogs treated with cp-asiNRL showed a decrease in the number of 19 ⁇ 1.85 per 0.2 mm 2 compared to the control, whereas by the L/M-opsin antibody It was confirmed that the number of stained cone cells showed an increase in the number of 19 ⁇ 1.85 per 0.2 mm 2 compared to the control.
  • the protein was extracted from the retinal analog at about 18 days (160 days after retinal analog culture). That is, proteins from 7 retinal analogues treated with cp-asiNRL at a concentration of about 0.1 ⁇ M, 7 retinal analogues treated with cp-asiNRL at a concentration of about 0.2 ⁇ M, and 7 control retinal analogues not treated with cp-asiNRL were isolated. extracted. Western blot analysis was performed on the extracted proteins using antibodies against rhodopsin, L/M-opsin, and S-opsin. Specific test materials and test methods were described in detail in the reference example above.
  • FIG. 3 is a view showing the results of western blot analysis for rhodopsin, L/M-opsin, and S-opsin, which are proteins extracted from retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. .
  • cp-asiNRL is a cone-like rod having cone-like properties rather than the original properties of rod cells in the process of rod cell generation.
  • rods can be converted into cone-like rods, or the proliferation of healthy cones that have already been generated can be increased. This suggests that it can induce a decrease in the number of rod cells and an increase in the number of cone cells. That is, cp-asiNRL suggests that it can enhance the production of cone cells or induce or enhance the production of cone-like rod cells.
  • cp-asiNRL can contribute to replenishing the dysfunction of cone cells by inducing or enhancing the production of cone cells or cone-like rods in a state in which cone cells are lost or dysfunctional. It can be used as an active ingredient of a pharmaceutical composition for improving or treating eye or retinal diseases for improving eyesight.

Abstract

Provided is a composition for inducing or enhancing the generation of cones or cone-like rods, the composition comprising an asymmetric RNAi-inducing nucleic acid molecule to inhibit the expression of the neural retina leucine zipper (NRL) gene.

Description

원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 조성물Compositions for inducing or enhancing the production of cone cells or cone-like rod cells
본 발명은 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 조성물에 관한 것으로, 더욱 상세하게는 NRL (Neural retina leucine zipper) 유전자의 발현을 억제하는 비대칭 RNAi 유도용 핵산 분자를 포함하는 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물에 관한 것이다. 본 특허출원은 2020년 12월 07일에 대한민국 특허청에 제출된 대한민국 특허출원 제10-2020-0169854호에 대하여 우선권을 주장하며, 상기 특허출원의 개시 사항은 본 명세서에 참조로서 삽입된다.The present invention relates to a composition for inducing or enhancing the production of cone cells or cone-like rod cells, and more particularly, to a composition comprising an asymmetric RNAi-inducing nucleic acid molecule that inhibits the expression of a Neural retina leucine zipper (NRL) gene. It relates to a composition for inducing or enhancing the production of cone cells or cone cell-like rod cells. This patent application claims priority to Korean Patent Application No. 10-2020-0169854 filed with the Korean Intellectual Property Office on December 07, 2020, the disclosure of which is incorporated herein by reference.
간상세포 (rod cell)는 빛에 매우 민감하므로, 이들은 흐릿한 상태에서 소량의 빛을 감지할 수 있다. 반대로, 원추세포 (cone cell)는 덜 민감하므로, 낮에 다량의 빛을 처리하고, 연속적으로 시각적 신호를 전달할 수 있다. 마우스 및 인간을 포함하는 많은 포유동물 종에서, 흐릿한 빛 아래에서 시력을 매개하는 간상세포의 수는 원추세포의 수를 크게 상회한다고 알려져 있다. 그러나, 조명이 원추세포를 하루 종일 작동하도록 하는 산업화된 세계에서, 간상세포-매개 시력은 덜 중요하다. 출생시부터 간상세포 기능이 부재한 많은 환자들이 확인되었고, 사실상 이들의 비정상적인 시력을 인식할 수 없었다는 보고가 있다 (Dryja, 2000). 반대로, 원추세포에 기능 이상 (dysfunction)이 있는 경우, 환자는 항상 증상을 보이고, 종종 이들의 원추세포 기능 이상의 정도에 따라 시각 장애 (visual handicap)를 겪는다. 또한, 일부 사례에서는, 원추세포가 손실되거나 또는 기능 이상이 발생하고, 간상세포가 상대적으로 보존된 상태로 유지되기도 한다. 예를 들면, 완전 색맹 (achromatopsia) 환자에 있어서, 출생시부터 원추세포 기능이 완전히 부재하지만 정상의 간상세포 기능을 갖는 중증 유전성 망막이영양증 (retinal dystrophy)인 경우가 보고되고 있다 (Hess et al, 1986; Nishiguchi et al, 2005). 나이 관련 황반 변성 (age related macular degeneration: AMD)의 경우에도, 중심 황반 (central macula)에서 원추세포-풍부 망막중심오목 (cone-rich fovea)의 변성에 의해 주로 시력 손상이 유발된다. 그러므로, 환자는 중심 시력 (vision)과 시각 (acuity)을 잃지만, 종종 주변 황반 (peripheral macula)은 비교적 잘 보존되어서, 원추세포의 부족으로 제한되긴 하나, 상기 망막중심오목 밖의 약간의 유용한 잔존 시력 (residual vision)을 갖는다. 대한민국 공개특허 제 10-2018-0012737 호에서는, 시각세포에서 내인성 감광성 신호전달을 조절하는 유전자 산물을 코딩하는 핵산을 이용하여 간상세포의 기능을 확장하여 시력을 향상시키는 방법에 대하여 개시하고 있다.Rod cells are very sensitive to light, so they can detect small amounts of light in dim conditions. Conversely, cone cells are less sensitive, so they can process large amounts of light during the day and continuously transmit visual signals. It is known that in many mammalian species, including mice and humans, the number of rods mediating vision under dim light greatly exceeds the number of cones. However, in an industrialized world where lighting forces cones to work all day, rod-mediated vision is less important. A number of patients have been identified who lack rod cell function from birth, and there is a report that they were virtually unable to recognize their abnormal visual acuity (Dryja, 2000). Conversely, when there is a dysfunction of the cone cells, the patient always shows symptoms and often suffers from visual handicap depending on the degree of their cone dysfunction. Also, in some cases, cone cells are lost or dysfunction occurs, and rods remain relatively conserved. For example, in patients with complete color blindness (achromatopsia), a case of severe hereditary retinal dystrophy (retinal dystrophy) having a normal rod function despite the complete absence of cone function from birth has been reported (Hess et al, 1986) (Hess et al, 1986) ; Nishiguchi et al, 2005). Even in the case of age related macular degeneration (AMD), visual impairment is mainly caused by degeneration of the cone-rich fovea in the central macula. Thus, the patient loses central vision and acuity, but often the peripheral macula is relatively well preserved, albeit limited by the lack of cones, some useful residual vision outside the fovea. (residual vision) Korean Patent Laid-Open No. 10-2018-0012737 discloses a method for improving visual acuity by extending the function of rod cells using a nucleic acid encoding a gene product regulating endogenous photosensitive signal transduction in visual cells.
한편, 망막의 광수용체가 분화하는 과정 및 정상적인 생리활성에는 많은 전사조절인자들이 작용하게 되는데, 그 중 NRL 전사활성인자는 1992년에 Swaroop 등 (Swarrop et al., Proc Natl Acad Sci USA, 89: 266-270(1992))에 의해 처음으로 보고되었다. 현재까지, NRL이 간상세포 및 원추세포의 분화 과정에서 매우 중요한 기능을 한다는 것에 대한 다양한 연구 결과가 존재한다. On the other hand, many transcriptional regulators act in the process of retinal photoreceptor differentiation and normal physiological activity. Among them, NRL transcriptional activator was identified in Swaroop et al. 266-270 (1992)). To date, there are various research results showing that NRL plays a very important function in the differentiation process of rod and cone cells.
다만, 아직까지 NRL 유전자를 조절함으로써 원추세포의 생성을 유도 또는 증진시키거나, 간상세포의 기능을 확장시킬 수 있는 시력 개선용 치료제에 대한 연구 결과는 없는 실정이다.However, there are still no research results on a therapeutic agent for improving eyesight that can induce or enhance the generation of cone cells or expand the function of rod cells by regulating the NRL gene.
상기한 바와 같은 문제를 해결하기 위하여, 본 발명의 발명자들은, NRL 유전자의 발현을 억제하는 비대칭 RNAi 유도용 핵산 분자를 개발하였고, 상기 비대칭 RNAi 유도용 핵산 분자는 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시킬 수 있음을 확인하여, 상기 비대칭 RNAi 유도용 핵산 분자의 시력 개선용 치료제로의 적용 가능성을 확인함으로써, 본 발명을 완성하였다.In order to solve the above problems, the inventors of the present invention developed a nucleic acid molecule for inducing asymmetric RNAi that inhibits the expression of the NRL gene, and the nucleic acid molecule for inducing asymmetric RNAi is a cone cell or cone-like rod cell. By confirming that the nucleic acid molecule for inducing asymmetric RNAi can be applied to a therapeutic agent for improving eyesight by confirming that it can induce or enhance the production of , the present invention has been completed.
본 발명의 일 목적은 NRL 유전자의 발현을 억제하는 비대칭 RNAi 유도용 핵산 분자를 포함하는 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물을 제공하고자 한다.One object of the present invention is to provide a composition for inducing or enhancing the production of cones or cone-like rods, which includes a nucleic acid molecule for inducing asymmetric RNAi that suppresses the expression of NRL gene.
본 발명의 다른 목적은 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 시력 개선용 약학적 조성물, 안구 또는 망막질환 개선 또는 치료용 약학적 조성물, 또는 세포 배양용 조성물을 제공하고자 한다.Another object of the present invention is to provide a nucleic acid molecule for inducing asymmetric RNAi or a pharmaceutical composition for improving eyesight, a pharmaceutical composition for improving or treating ocular or retinal diseases, or a composition for cell culture comprising the composition.
본 발명의 또 다른 목적은 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 이용하는 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 방법 또는 안구 또는 망막질환을 개선 또는 치료하는 방법을 제공하고자 한다.Another object of the present invention is to provide a method for inducing or enhancing the generation of cone cells or cone-like rod cells using the asymmetric RNAi-inducing nucleic acid molecule or the composition, or a method for improving or treating ocular or retinal diseases. do.
본 발명의 또 다른 목적은 상기 비대칭 RNAi 유도용 핵산 분자 또는 이를 포함하는 조성물의, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키기 위한 용도, 시력을 개선시키기 위한 용도, 또는 안구 또는 망막질환을 개선 또는 치료하기 위한 용도를 제공하고자 한다.Another object of the present invention is the use of the nucleic acid molecule for inducing asymmetric RNAi or a composition comprising the same, for inducing or enhancing the production of cone cells or cone-like rods, for improving visual acuity, or for ocular or An object of the present invention is to provide a use for improving or treating retinal diseases.
그러나 본 발명이 이루고자 하는 기술적 과제는 이상에서 언급한 과제에 제한되지 않으며, 언급되지 않은 또 다른 과제들은 아래의 기재로부터 당업계에서 통상의 지식을 가진 자에게 명확하게 이해될 수 있을 것이다. However, the technical problem to be achieved by the present invention is not limited to the above-mentioned problems, and other problems not mentioned will be clearly understood by those of ordinary skill in the art from the following description.
상기 목적을 달성하기 위하여, 본 발명은 NRL 유전자의 발현을 억제하는 비대칭 RNAi 유도용 핵산 분자를 포함하는 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물을 제공한다.In order to achieve the above object, the present invention provides a composition for inducing or enhancing the production of cones or cone-like rods, comprising a nucleic acid molecule for inducing asymmetric RNAi that inhibits the expression of NRL gene.
본 발명은 또한, 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 시력 개선용 약학적 조성물 또는 안구 또는 망막질환 개선 또는 치료용 약학적 조성물을 제공한다.The present invention also provides a nucleic acid molecule for inducing asymmetric RNAi or a pharmaceutical composition for improving eyesight, or a pharmaceutical composition for improving or treating ocular or retinal diseases, comprising the composition.
본 발명은 또한, 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 세포 배양용 조성물을 제공한다.The present invention also provides a composition for cell culture for inducing or enhancing the production of the asymmetric RNAi-inducing nucleic acid molecule or a cone or cone-like rod comprising the composition.
본 발명은 또한, 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 이용하여 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 방법을 제공한다. The present invention also provides a method for inducing or enhancing the production of cone cells or cone-like rods using the nucleic acid molecule for inducing asymmetric RNAi or the composition.
본 발명은 또한, 상기 비대칭 RNAi 유도용 핵산 분자 또는 상기 조성물을 개체에 투여하는 단계를 포함하는 안구 또는 망막질환을 개선 또는 치료하는 방법을 제공한다.The present invention also provides a method for improving or treating an ocular or retinal disease, comprising administering the nucleic acid molecule for inducing asymmetric RNAi or the composition to a subject.
본 발명은 또한, 상기 비대칭 RNAi 유도용 핵산 분자 또는 이를 포함하는 조성물의, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키기 위한 용도, 시력을 개선시키기 위한 용도, 또는 안구 또는 망막질환을 개선 또는 치료하기 위한 용도를 제공한다.The present invention also relates to the use of the nucleic acid molecule for inducing asymmetric RNAi or a composition comprising the same, for inducing or enhancing the production of cone cells or cone-like rods, for improving visual acuity, or for ocular or retinal diseases. Provides a use for improving or treating
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술 분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로 본 명세서에서 사용된 명명법은 본 기술 분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is those well known and commonly used in the art.
본 발명의 상세한 설명 등에서 사용되는 주요 용어의 정의는 다음과 같다.Definitions of key terms used in the detailed description of the present invention are as follows.
"RNAi (RNA interference: RNA 간섭)"란, 목적 유전자의 mRNA와 상동인 서열을 가지는 가닥과 이것과 상보적인 서열을 가지는 가닥으로 구성되는 이중가닥 RNA (dsRNA)를 세포 등에 도입하여 목적 유전자 mRNA의 분해를 유도함으로써 목적 유전자의 발현을 억제하는 메카니즘을 의미한다."RNAi (RNA interference)" refers to double-stranded RNA (dsRNA) composed of a strand having a sequence homologous to the mRNA of a target gene and a strand having a sequence complementary to this by introducing into a cell, etc. the target gene mRNA. It refers to a mechanism for suppressing the expression of a target gene by inducing degradation.
"RNAi 유도용 핵산 분자 (nucleic acid molecules inducing RNAi)"란 서열-특이적 방식으로 상기 RNA 간섭을 매개함으로써 유전자 발현 또는 바이러스 복제를 억제 또는 하향 조절할 수 있는 임의의 핵산 분자를 지칭한다. 상기 용어는 개별 핵산 분자, 복수의 상기 핵산 분자, 또는 상기 핵산 분자의 풀 (pool) 모두를 지칭할 수 있다. 일 구체예에 있어서 상기 RNAi 유도용 핵산 분자는 siRNA일 수 있다."Nucleic acid molecules inducing RNAi" refers to any nucleic acid molecule capable of inhibiting or down-regulating gene expression or viral replication by mediating said RNA interference in a sequence-specific manner. The term may refer to both an individual nucleic acid molecule, a plurality of said nucleic acid molecules, or a pool of said nucleic acid molecules. In one embodiment, the nucleic acid molecule for inducing RNAi may be siRNA.
"siRNA (small interfering RNA: 짧은 간섭 RNA)"란, 서열 특이적으로 효율적인 유전자 발현 억제 (gene silencing)를 매개하는 짧은 이중 가닥의 RNA (dsRNA)를 의미한다.“Small interfering RNA (siRNA)” refers to a short double-stranded RNA (dsRNA) that mediates sequence-specifically efficient gene silencing.
"안티센스 가닥 (antisense strand)"이란 관심 있는 목적 핵산 (target nucleic acid)에 실질적으로 또는 100% 상보적인 폴리뉴클레오티드로서, 예를 들어 mRNA (messenger RNA), mRNA가 아닌 RNA 서열 (e.g.,microRNA, piwiRNA, tRNA, rRNA, 및 hnRNA), 또는 코딩 또는 비코딩 DNA 서열과 전체로서 또는 일부로서 상보적일 수 있다.An "antisense strand" is a polynucleotide substantially or 100% complementary to a target nucleic acid of interest, for example, messenger RNA (mRNA), a non-mRNA RNA sequence (e.g., microRNA, piwiRNA). , tRNA, rRNA, and hnRNA), or coding or non-coding DNA sequences, in whole or in part.
"센스 가닥 (sense strand)"이란 목적 핵산과 동일한 핵산 서열을 갖는 폴리뉴클레오티드로서, mRNA (messenger RNA), mRNA가 아닌 RNA 서열 (e.g., microRNA, piwiRNA, tRNA, rRNA, 및 hnRNA), 또는 코딩 또는 비코딩 DNA 서열과 전체로서 또는 일부로서 동일한 폴리뉴클레오티드를 말한다.The term "sense strand" is a polynucleotide having the same nucleic acid sequence as a target nucleic acid, wherein the mRNA (messenger RNA), non-mRNA RNA sequence (e.g., microRNA, piwiRNA, tRNA, rRNA, and hnRNA), or coding or Refers to a polynucleotide identical in whole or in part to a non-coding DNA sequence.
"유전자"란 최광의의 의미로 간주되어야 하며, 구조 단백질 또는 조절 단백질을 암호화할 수 있다. 이때, 조절단백질은 전사인자, 열 충격단백질, 또는 DNA/RNA 복제, 전사, 및/또는 번역에 관여하는 단백질을 포함한다. 본 발명에 있어서, 발현 억제의 대상이 되는 목적 유전자는 바이러스 게놈에 내재된 것으로, 동물 유전자로 통합되거나 염색체 외 구성요소로서 존재할 수 있다. 예컨대, 목적 유전자는 HIV 게놈상의 유전자일 수 있다. 이 경우, siRNA 분자는 포유동물 세포 내 HIV 유전자의 번역을 불활성화시키는데 유용하다.A “gene” is to be taken in its broadest sense and may encode a structural protein or a regulatory protein. In this case, the regulatory protein includes a transcription factor, a heat shock protein, or a protein involved in DNA/RNA replication, transcription, and/or translation. In the present invention, the target gene to be suppressed is inherent in the viral genome, and may be integrated into an animal gene or exist as an extrachromosomal component. For example, the gene of interest may be a gene on the HIV genome. In this case, the siRNA molecule is useful for inactivating translation of an HIV gene in a mammalian cell.
"NRL (Neural retina leucine zipper)"은 전사인자로서 로돕신 (Rhodosin)과 같은 간상세포의 특징을 갖게 하는 유전자의 프로모터에 붙어 망막 전구체 세포가 간상세포로 분화되도록 한다. NRL은 직접적으로, 또는 downstream에 있는 Nr2e3를 통해 망막 전구체 세포가 원추세포로 분화되는 것을 막는다. 따라서 NRL은 망막 전구체 세포가 간상세포로 분화되게 하는 중요한 인자이다."NRL (Neural retina leucine zipper)" is a transcription factor that attaches to the promoter of a gene that has characteristics of rod cells, such as rhodosin, so that retinal precursor cells are differentiated into rod cells. NRL prevents retinal progenitor cells from differentiating into cones either directly or through downstream Nr2e3. Therefore, NRL is an important factor that allows retinal progenitor cells to differentiate into rod cells.
"원추세포 (cone cell)"는 망막에서도 황반의 중심부에 밀집되어 있는 통통한 모양을 하고 있는 눈의 망막에 있는 시세포를 지칭한다. 상기 원추세포는 약 0.1 Lux 이상의 밝은 빛을 감지할 수 있고, 색깔을 구별할 수 있으며, 추상세포라고도 지칭된다."Cone cell" refers to photoreceptor cells in the retina of the eye, which are densely packed in the center of the macula even in the retina and have a plump shape. The cone cells can sense bright light of about 0.1 Lux or more, can distinguish colors, and are also referred to as cone cells.
"간상세포 (rod cell)"는 망막의 주변부에 많이 분포하며 약한 빛에 민감한 가늘고 긴 모양을 하고 있는 눈의 망막에 있는 시세포를 지칭한다. 상기 간상세포는 명암과 물체의 형태는 감각하지만 색깔은 구별하지 못하고, 약 0.1 Lux 이하의 약한 빛을 감지할 수 있다."Rod cell" refers to a photoreceptor cell in the retina of the eye, which is widely distributed in the periphery of the retina and has an elongated shape sensitive to weak light. The rod cells sense the light and shade and the shape of the object, but cannot distinguish the color, and can sense a weak light of about 0.1 Lux or less.
"원추세포-유사 간상세포 (cone cell-like rod cell)"는 간상세포 본래의 성질이 상실되거나 감소되어 원추세포의 성질을 갖는 간상세포를 의미한다. 상기 원추세포-유사 간상세포는 간상세포의 생성 과정 중에 발생되거나, 완전히 생성된 간상세포가 성질이 변화되어 원추세포-유사 간상세포로 전환됨으로써 발생되는 것일 수 있다. "Cone cell-like rod cell" refers to a rod cell having the properties of a cone cell due to loss or reduction in the original properties of the rod cell. The cone cell-like rod cells may be generated during the production process of rod cells, or may be generated when completely generated rod cells change their properties and are converted into cone cell-like rod cells.
색맹 (color blindness)은 망막의 시세포에 이상이 있어서, 색깔을 제대로 구별하지 못하는 유전 형질을 의미하고, 색각 이상이라고도 한다. 색맹은 많은 경우 선천적인 원인에 기인하나 후천적으로 발생되기도 한다. 대부분의 색맹은 색깔을 구별할 수 있는 원추세포에 이상이 있어서 발생된다. 구체적으로, 완전 색맹 (achromatopsia) 환자에 있어서, 출생시부터 원추세포 기능이 완전히 부재하지만 정상의 간상세포 기능을 갖는 중증 유전성 망막이영양증 (retinal dystrophy)인 경우가 보고되고 있다 (Hess et al, 1986; Nishiguchi et al, 2005). 따라서, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도하거나 증진시키는 것에 의하여 색맹의 증상 또는 질환을 개선 또는 치료할 수 있는 가능성이 있다.Color blindness refers to a genetic trait that does not properly distinguish colors due to abnormalities in the photoreceptor cells of the retina, and is also called color vision abnormality. In many cases, color blindness is due to congenital causes, but it can also be acquired. Most color blindness is caused by abnormalities in the cone cells that can distinguish colors. Specifically, in patients with complete color blindness (achromatopsia), the case of severe hereditary retinal dystrophy (retinal dystrophy) with a complete absence of cone function from birth but normal rod function has been reported (Hess et al, 1986; Nishiguchi et al, 2005). Therefore, there is a possibility to improve or treat the symptoms or diseases of color blindness by inducing or enhancing the production of cone cells or cone cell-like rod cells.
본 발명에 있어서, RNAi 유도용 핵산 분자는 센스 가닥은 15 내지 17nt의 길이를 가지고, 안티센스 가닥은 16nt 이상의 길이를 가지는 것을 특징으로 할 수 있다. 이에 제한되는 것은 아니나, 상기 안티센스 가닥은 16 내지 31nt의 길이를 가지는 것을 특징으로 할 수 있으며, 바람직하게는 19 내지 26nt의 길이를 가지는 것을 특징으로 할 수 있다. 더욱 바람직하게는, 상기 센스 가닥의 길이는 16nt, 이와 상보적인 안티센스 가닥의 길이는 19nt, 24nt, 25nt, 또는 26nt인 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the nucleic acid molecule for inducing RNAi may be characterized in that the sense strand has a length of 15 to 17 nt and the antisense strand has a length of 16 nt or more. Although not limited thereto, the antisense strand may be characterized as having a length of 16 to 31 nt, preferably having a length of 19 to 26 nt. More preferably, the length of the sense strand is 16 nt, and the length of the antisense strand complementary thereto may be characterized in that it is 19 nt, 24 nt, 25 nt, or 26 nt, but is not limited thereto.
상기 센스 가닥의 3' 말단 및 안티센스 가닥의 5' 말단은 블런트 말단 (blunt end)을 형성한다. 안티센스 가닥의 3' 말단은 예를 들어 1 내지 16nt의 오버행 (overhang)을 포함할 수 있다.The 3' end of the sense strand and the 5' end of the antisense strand form a blunt end. The 3' end of the antisense strand may include, for example, an overhang of 1 to 16 nt.
본 발명에 있어서, 상기 RNAi 유도용 핵산 분자의 센스 가닥 또는 안티센스 가닥은 하나 이상의 화학적 변형 (chemical modification)을 포함하는 것을 특징으로 할 수 있다.In the present invention, the sense strand or the antisense strand of the nucleic acid molecule for inducing RNAi may include one or more chemical modifications.
일반적인 siRNA는 포스페이트 백본 구조에 의한 높은 음전하 및 높은 분자량 등의 이유로 세포막을 통과할 수 없고 혈액에서의 빠른 분해 및 제거되어 실제 표적 부위에 RNAi 유도를 위한 충분한 양을 전달하는데 어려움이 있다. 현재 in vitro 전달의 경우 cationic lipids와 cationic polymers들을 이용한 높은 효율의 delivery 방법이 많이 개발되어 있지만, in vivo의 경우에는 in vitro만큼의 높은 효율로 siRNA를 전달하기 어렵고, 생체 내에 존재하는 다양한 단백질들과 상호작용에 의하여 siRNA 전달 효율이 감소하는 문제점이 있다.General siRNA cannot pass through the cell membrane for reasons such as high negative charge and high molecular weight due to the phosphate backbone structure, and it is rapidly degraded and removed from the blood, so it is difficult to deliver a sufficient amount for RNAi induction to the actual target site. Currently, in the case of in vitro delivery, many high-efficiency delivery methods using cationic lipids and cationic polymers have been developed, but in vivo, it is difficult to deliver siRNA as high as in vitro, and it is difficult to There is a problem in that the siRNA delivery efficiency is reduced due to the interaction.
이에, 본 발명자들은 비대칭 siRNA 구조에 화학적 변형을 도입하여 별도의 전달체 없이 효과적이고 세포 내 전달을 할 수 있는 자가 전달능을 가진 asiRNA 구조체 (cp-asiRNA)를 개발하였다.Accordingly, the present inventors introduced a chemical modification to the asymmetric siRNA structure to develop an asiRNA construct (cp-asiRNA) with self-delivery ability that can be effectively and intracellularly delivered without a separate carrier.
본 발명에 있어서, 상기 센스 가닥 또는 안티센스 가닥에서 화학적 변형은 다음으로 구성된 군에서 선택된 하나 이상을 포함할 수 있다:In the present invention, the chemical modification in the sense strand or the antisense strand may include one or more selected from the group consisting of:
뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -CH3 (메틸), -OCH3 (methoxy), -NH2, -F (불소), -O-2-메톡시에틸-O-프로필 (propyl), -O-2-메틸티오에틸 (methylthioethyl), -O-3-아미노프로필, 또는 -O-3-디메틸아미노프로필로 치환; 뉴클레오티드 내 당 (sugar) 구조의 산소가 황으로 치환; 뉴클레오티드 결합이 포스포로티오에이트 (phosphorothioate), 보라노포스페이트 (boranophosphate), 또는 메틸포스포네이트 (methyl phosphonate)로 변형; PNA (peptide nucleic acid), LNA (locked nucleic acid), 또는 UNA (unlocked nucleic acid) 형태로의 변형; 및 인산기 (phosphate group), 친유성 화합물 (lipophilic compound), 또는 세포 침투 펩타이드 결합.-OH group at the 2' carbon position of the sugar structure in the nucleotide is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl-O-propyl ( propyl), -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; replacement of oxygen in the nucleotide structure with sulfur; nucleotide linkages are modified with phosphorothioate, boranophosphate, or methyl phosphonate; transformation into peptide nucleic acid (PNA), locked nucleic acid (LNA), or unlocked nucleic acid (UNA) form; and a phosphate group, a lipophilic compound, or a cell penetrating peptide bond.
본 발명에 있어서, 상기 친유성 화합물 (lipophilic compound)은 콜레스테롤, 토코페롤, 스테아르산 (stearic acid), 레티노산 (retinoic acid), DHA (docosahexaenoic acid), 팔미트산 (palmitic acid), 리놀레산 (linoleic acid), 리놀렌산 (linolenic acid), 및 탄소수 10 개 이상의 장쇄지방산으로 구성된 군에서 선택되는 어느 하나 이상인 것을 특징으로 할 수 있다. 바람직하게는 콜레스테롤인 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the lipophilic compound is cholesterol, tocopherol, stearic acid, retinoic acid, DHA (docosahexaenoic acid), palmitic acid, linoleic acid ), linolenic acid, and any one or more selected from the group consisting of long-chain fatty acids having 10 or more carbon atoms. Preferably, it may be characterized as cholesterol, but is not limited thereto.
일 구체예에 있어서, 상기 센스 가닥은 하기로부터 선택된 하나 이상의 화학적 변형을 포함할 수 있다: 3' 말단으로부터 인접한 2 내지 4 개의 뉴클레오티드 결합이 포스포로티오에이트 (phosphorothioate), 보라노포스페이트 (boranophosphate), 또는 메틸포스포네이트 (methyl phosphonate)로 변형; 2 개 이상의 뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -CH3 (methyl), -OCH3 (methoxy), -NH2, -F (불소), -O-2-메톡시에틸-O-프로필 (propyl), -O-2-메틸티오에틸 (methylthioethyl), -O-3-아미노프로필, 또는 -O-3-디메틸아미노프로필로 치환; 및 3' 말단에 친유성 화합물 (lipophilic compound) 또는 세포 침투 펩타이드의 결합.In one embodiment, the sense strand may comprise one or more chemical modifications selected from: 2 to 4 nucleotide bonds adjacent to the 3' end are phosphorothioate, boranophosphate, or modified with methyl phosphonate; -OH group at the 2' carbon position of the sugar structure within 2 or more nucleotides is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl-O substituted with -propyl, -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; and binding of a lipophilic compound or a cell penetrating peptide to the 3' end.
일 구체예에 있어서, 상기 안티센스 가닥은 하기로부터 선택되는 어느 하나 이상의 화학적 변형을 포함할 수 있다: 3' 말단으로부터 인접한 3 내지 5 개의 뉴클레오티드 결합이 포스포로티오에이트 (phosphorothioate), 보라노포스페이트 (boranophosphate), 또는 메틸포스포네이트 (methyl phosphonate)로 변형; 2 개 이상의 뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -CH3 (methyl), -OCH3 (methoxy), -NH2, -F (불소), -O-2-메톡시에틸 -O-프로필 (propyl), -O-2-메틸티오에틸 (methylthioethyl), -O-3-아미노프로필, 또는 -O-3-디메틸아미노프로필로 치환; 및 5' 말단에 인산기 (phosphate group) 또는 세포 침투 펩타이드의 결합.In one embodiment, the antisense strand may contain any one or more chemical modifications selected from the following: 3 to 5 nucleotide bonds adjacent to the 3' end are phosphorothioate, boranophosphate ), or modified with methyl phosphonate; -OH group at the 2' carbon position of the sugar structure within 2 or more nucleotides is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl -O substituted with -propyl, -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl; and binding of a phosphate group or a cell penetrating peptide to the 5' end.
다른 구체예에 있어서, 상기 RNAi 유도용 핵산 분자는 상기 센스 가닥 또는 안티센스 가닥 중 2 개 이상의 뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -OCH3 (methoxy) 또는 -F (불소)로 치환되는 변형; 상기 센스 가닥 또는 안티센스 가닥에서 10% 이상의 뉴클레오티드 결합이 포스포로티오에이트 (phosphorothioate)로 변형; 상기 센스 가닥의 3' 말단에 콜레스테롤 결합; 및 상기 안티센스 가닥의 5' 말단에 인산기 (phosphate group) 결합으로 구성된 군에서 선택된 하나 이상의 변형을 포함하는 것을 특징으로 할 수 있다.In another embodiment, in the nucleic acid molecule for inducing RNAi, the -OH group at the 2' carbon position of the sugar structure within two or more nucleotides of the sense strand or the antisense strand is replaced with -OCH 3 (methoxy) or -F (fluorine) being transformed; at least 10% of the nucleotide linkages in the sense strand or the antisense strand are modified to phosphorothioate; cholesterol binding to the 3' end of the sense strand; and one or more modifications selected from the group consisting of a phosphate group bond to the 5' end of the antisense strand.
예시적인 RNAi 유도용 핵산 분자는 하기의 센스 가닥을 포함할 수 있다:Exemplary nucleic acid molecules for inducing RNAi may include the following sense strands:
mC(2'-F-G)mG(2'-F-C)mG(2'-F-C)mU(2'-F-G)mG(2'-F-U)mC(2'-F-U)mC(2'-F-G)*mA*(2'-F-U)*-Chol.mC(2'-F-G)mG(2'-F-C)mG(2'-F-C)mU(2'-F-G)mG(2'-F-U)mC(2'-F-U)mC(2'-F-G)* mA*(2'-F-U)*-Chol.
또한, 예시적인 RNAi 유도용 핵산 분자는 하기의 안티센스 가닥을 포함할 수 있다:In addition, exemplary nucleic acid molecules for inducing RNAi may include the following antisense strands:
PmA(2'-F-U)mC(2'-F-G)mA(2'-F-G)mA(2'-F-C)mC(2'-F-A)mG(2'-F-C)mG(2'-F-C)mC(2'-F-G)mC(2'-F-G)mU(2'-F-C)mG(2'-F-G)*mA*(2'-F-A)*mA*(2'-F-A). PmA(2'-F-U)mC(2'-F-G)mA(2'-F-G)mA(2'-F-C)mC(2'-F-A)mG(2'-F-C)mG(2'-F-C)mC (2'-F-G)mC(2'-F-G)mU(2'-F-C)mG(2'-F-G)*mA*(2'-F-A)*mA*(2'-F-A).
상기에서, *는 포스포로티오에이트 결합 (phosphorothioated bond)으로의 변형, m은 2'-O-메틸 (Methyl)로의 치환, 2'-F-는 2'-플루오르 (Fluoro)로의 치환, -chol은 3'-콜레스테롤의 결합, P는 5'-인산기의 결합을 의미한다.In the above, * is a phosphorothioate bond (phosphorothioated bond) modification, m is 2'-O-methyl (Methyl) substitution, 2'-F- is 2'-Fluoro substitution, -chol denotes a bond with 3'-cholesterol, and P denotes a bond with a 5'-phosphate group.
본 발명에 있어서, 상기 안티센스 가닥의 5' 말단에 인산기가 한 개 내지 세 개 결합될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, one to three phosphate groups may be bonded to the 5' end of the antisense strand, but the present invention is not limited thereto.
본 발명에 있어서, RNAi 유도용 핵산 분자는 NRL 유전자의 발현을 억제하는 것을 특징으로 할 수 있다. 상기 RNAi 유도용 핵산 분자는 NRL을 코딩하는 mRNA와 상보적으로 결합하는 것에 의하여, NRL 유전자의 발현을 억제하는 것을 특징으로 할 수 있다. 따라서, 상기 NRL 유전자, 예컨대, NRL을 코딩하는 mRNA는 상기 RNAi 유도용 핵산 분자가 표적하는 표적 서열을 포함할 수 있다. In the present invention, the nucleic acid molecule for inducing RNAi may be characterized in that it inhibits the expression of the NRL gene. The nucleic acid molecule for inducing RNAi may be characterized in that it inhibits the expression of the NRL gene by complementary binding to the mRNA encoding the NRL. Accordingly, the NRL gene, for example, the mRNA encoding the NRL may include a target sequence targeted by the RNAi-inducing nucleic acid molecule.
본 발명은 다른 관점에서, 상기 RNAi 유도용 핵산 분자를 포함하는 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물에 관한 것이다.In another aspect, the present invention relates to a composition for inducing or enhancing the production of cones or cone-like rods comprising the nucleic acid molecule for inducing RNAi.
일 구체예에 있어서, 상기 RNAi 유도용 핵산 분자는 광수용세포 중 간상세포의 수적 감소 및 원추세포의 수적 증가를 유도할 수 있다. 구체적으로, 상기 RNAi 유도용 핵산 분자는 NRL 유전자의 발현을 억제할 수 있고, 이로 인해, 간상세포가 생성되는 과정에서 간상세포 본래의 성질이 상실되거나 감소되어 원추세포의 성질을 갖는 원추세포-유사 간상세포의 생성을 유도할 수 있다. 또한, 상기 RNAi 유도용 핵산 분자는 NRL 유전자의 발현을 억제할 수 있고, 이로 인해, 이미 생성된 간상세포의 성질을 원추세포의 성질로 변화시킴으로써, 간상세포를 원추세포-유사 간상세포로 전환시킬 수 있다. 또한, 상기 RNAi 유도용 핵산 분자는 NRL 유전자의 발현을 억제할 수 있고, 이로 인해, 간상세포의 수적 감소 및 원추세포의 수적 증가를 유도할 수 있다. 상기 간상세포의 수적 감소는 간상세포로의 분화가 감소되거나 간상세포의 사멸이 촉진되는 것에 의한 것일 수 있다. 또한, 상기 원추세포의 수적 증가는 원추세포로의 분화 또는 증식이 촉진되는 것에 의한 것일 수 있다. In one embodiment, the RNAi-inducing nucleic acid molecule can induce a decrease in the number of rod cells and an increase in the number of cone cells among photoreceptor cells. Specifically, the RNAi-inducing nucleic acid molecule can suppress the expression of the NRL gene, and thus, the original properties of rod cells are lost or reduced in the process of rod cell generation, and thus cone cell-like properties with cone-like properties. It can induce the production of rod cells. In addition, the RNAi-inducing nucleic acid molecule can inhibit the expression of the NRL gene, thereby changing the properties of previously generated rod cells to those of cone cells, thereby converting rod cells into cone-like rod cells. can In addition, the RNAi-inducing nucleic acid molecule can inhibit the expression of the NRL gene, thereby inducing a decrease in the number of rod cells and an increase in the number of cone cells. The decrease in the number of rod cells may be due to reduced differentiation into rod cells or promotion of rod cell death. In addition, the increase in the number of the cone cells may be due to promotion of differentiation or proliferation into cone cells.
따라서, 상기 RNAi 유도용 핵산 분자를 포함하는 조성물은 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시킬 수 있다. 구체적으로 상기 조성물은 NRL 유전자의 발현을 억제할 수 있고, 이로 인해, 간상세포가 생성되는 과정에서 간상세포 본래의 성질이 상실되거나 감소되어 원추세포의 성질을 갖는 원추세포-유사 간상세포의 생성을 유도하거나; 이미 생성된 간상세포의 성질을 원추세포의 성질로 변화시킴으로써, 간상세포를 원추세포-유사 간상세포로 전환시키거나; 원추세포 자체의 수적 증가를 유도할 수 있다.Accordingly, the composition comprising the RNAi-inducing nucleic acid molecule can induce or enhance the production of cone cells or cone-like rod cells. Specifically, the composition can inhibit the expression of the NRL gene, whereby the original properties of the rod cells are lost or reduced in the process of generating rod cells, thereby preventing the production of cone-like rod cells having cone-like properties. induce; converting rod cells into cone-like rod cells by changing the properties of previously generated rod cells to those of cone cells; It can induce an increase in the number of cone cells themselves.
따라서, 상기 RNAi 유도용 핵산 분자 또는 이를 포함하는 조성물은 원추세포가 손실되었거나 또는 기능 이상인 상태에서, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시켜, 원추세포의 기능 이상을 보충하는데 기여할 수 있으므로, 시력 개선용 약학적 조성물, 또는 안구 또는 망막질환 개선 또는 치료용 약학적 조성물의 유효 성분으로 활용될 수 있다.Accordingly, the RNAi-inducing nucleic acid molecule or a composition comprising the same induces or enhances the production of cone cells or cone-like rod cells in a state in which cone cells are lost or dysfunctional, thereby supplementing the dysfunction of cone cells. Since it can contribute, it can be utilized as an active ingredient of a pharmaceutical composition for improving eyesight, or a pharmaceutical composition for improving or treating eye or retinal diseases.
따라서, 본 발명은 다른 관점에서, 상기 RNAi 유도용 핵산 분자를 포함하고, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 시력 개선용 약학적 조성물에 관한 것이다.Accordingly, in another aspect, the present invention relates to a pharmaceutical composition for improving eyesight comprising the RNAi-inducing nucleic acid molecule, and inducing or enhancing the production of cone cells or cone-like rod cells.
또한, 본 발명은 다른 관점에서, 상기 RNAi 유도용 핵산 분자를 포함하고, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 안구 또는 망막질환 개선 또는 치료용 약학적 조성물에 관한 것이다.In another aspect, the present invention relates to a pharmaceutical composition for improving or treating ocular or retinal diseases, including the RNAi-inducing nucleic acid molecule, and inducing or enhancing the production of cone cells or cone-like rod cells.
상기 안구 또는 망막질환은 원추세포가 손실되었거나 또는 기능 이상인 상태에서, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시켜, 원추세포의 기능 이상을 보완 또는 보충하는 것에 의하여 증상이 개선되거나 치료될 수 있는 질환을 모두 포함할 수 있다. 바람직하게는, 상기 안구 또는 망막질환은 원추세포가 손실되었거나 또는 기능 이상이면서, 간상세포는 정상적으로 존재하는 상태에서 발생하는 질환일 수 있다. The ocular or retinal disease is a condition in which cone cells are lost or malfunctioning, by inducing or enhancing the production of cone cells or cone cell-like rod cells, thereby compensating or supplementing the functional abnormality of cone cells. It may include any disease that can be treated. Preferably, the ocular or retinal disease may be a disease in which cone cells are lost or malfunctioning, while rod cells are normally present.
가장 바람직하게는, 상기 안구 또는 망막질환은 색맹을 동반하는 질환이거나 색맹일 수 있다. 구체적으로, 상기 색맹은 선척적이거나 후천적으로 발생될 수 있다. 또한, 상기 색맹은 완전 색맹, 부분 색맹, 또는 색약일 수 있다. 상기 완전 색맹은 단색형 색각 이상 (achromatopsia)으로 색상을 구별하는 원추세포 자체가 망막에서 아예 존재하지 않는 경우로, 다른 색맹과는 다르게 색상을 전혀 구별할 수 없는 증상 또는 질환을 의미한다. 상기 부분 색맹은 망막에서 삼원색인 적색, 녹색, 청색을 인식하는 원추세포의 결핍으로 야기되는 것으로, 일부 색상을 구별할 수 없는 증상 또는 질환을 의미한다. 상기 부분 색맹은 제 1 색각 이상, 제 2 색각 이상, 및 제 3 색각 이상으로 구성된 군으로부터 선택된 하나 이상일 수 있다. 상기 제 1 색각 이상은 적색을 인식하는 L원추세포 (ρ세포)가 없는 경우로 적색과 녹색을 구별할 수 없고, 적색을 어두운 색상으로 인식하는 증상 또는 질환을 의미한다. 상기 제 2 색각 이상은 녹색을 인식하는 M원추세포 (Г세포)가 없으며 제 1 색각 이상과 마찬가지로 적색과 녹색을 구별할 수 없으나, 이 경우는 제1 색각이상과는 달리 녹색을 어두운 색상으로 인식하는 증상 또는 질환을 의미한다. 상기 제 3 색각 이상은 청색을 인식하는 S원추세포 (β세포)가 존재하지 않는 경우로 청색과 노란색을 구별할 수 없고, 청색을 짙은 녹색으로 노란색을 옅은 적색으로 인식하는 증상 또는 질환을 의미한다. 상기 색약은 낮은 채도의 색상을 인식하지 못하거나 단시간에 분별할 수 없는 증상 또는 질환을 의미한다. Most preferably, the ocular or retinal disease may be a disease accompanying color blindness or color blindness. Specifically, the color blindness may be congenital or acquired. In addition, the color blindness may be complete color blindness, partial color blindness, or color weakness. The complete color blindness refers to a case in which cone cells that discriminate colors do not exist in the retina at all in achromatopsia, and refers to a symptom or disease that cannot distinguish colors at all, unlike other color blindness. The partial color blindness is caused by a lack of cone cells that recognize the three primary colors of red, green, and blue in the retina, and refers to a symptom or disease in which some colors cannot be distinguished. The partial color blindness may be at least one selected from the group consisting of a first color vision or more, a second color vision or more, and a third color vision or more. The first color vision abnormality refers to a symptom or disease in which there is no L cone cell (ρ cell) for recognizing red, red and green cannot be distinguished, and red is recognized as a dark color. The second color vision abnormality does not have M cone cells (Г cells) that recognize green and cannot distinguish red from green like the first color vision abnormality, but in this case, unlike the first color vision abnormality, green is recognized as a dark color. means a symptom or disease. The third color vision abnormality refers to a symptom or disease in which S cone cells (β cells) that recognize blue do not exist, cannot distinguish between blue and yellow, and recognize blue as dark green and yellow as pale red. . The color weakness refers to a symptom or disease in which a low-saturation color cannot be recognized or distinguished in a short time.
상기 약학적 조성물은 유효성분인 RNAi 유도용 핵산 분자 이외에 추가로 약제학적으로 허용 가능한 담체를 1 종 이상 포함하여 제조할 수 있다. 약제학적으로 허용 가능한 담체는 본 발명의 유효성분과 양립 가능하여야 하며, 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로즈 용액, 말토덱스트린 용액, 글리세롤, 에탄올, 및 이들 성분 중 한 성분 또는 둘 이상의 성분을 혼합하여 사용할 수 있고, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제, 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형으로 제제화 할 수 있다. 특히, 동결건조 (lyophilized)된 형태의 제형으로 제제화하여 제공하는 것이 바람직하다. 동결건조 제형 제조를 위해서 본 발명이 속하는 기술 분야에서 통상적으로 알려져 있는 방법이 사용될 수 있으며, 동결건조를 위한 안정화제가 추가될 수도 있다.The pharmaceutical composition may be prepared by including one or more pharmaceutically acceptable carriers in addition to the nucleic acid molecule for inducing RNAi as an active ingredient. A pharmaceutically acceptable carrier must be compatible with the active ingredient of the present invention, and saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or two or more of these ingredients may be used by mixing, and other conventional additives such as antioxidants, buffers, and bacteriostats may be added as needed. In addition, diluents, dispersants, surfactants, binders, and lubricants may be additionally added to form an injectable formulation such as an aqueous solution, suspension, emulsion, or the like. In particular, it is preferable to provide the formulation in a lyophilized form. For the preparation of the freeze-dried formulation, a method commonly known in the art to which the present invention pertains may be used, and a stabilizer for freeze-drying may be added.
상기 약학적 조성물의 투여방법은 통상의 환자의 증후와 질병의 심각도에 기초하여 본 기술 분야의 통상의 전문가가 결정할 수 있다. 또한, 산제, 정제, 캡슐제, 액제, 주사제, 연고제, 시럽제 등의 다양한 형태로 제제화 할 수 있으며 단위-투여량 또는 다-투여량 용기, 예를 들면 밀봉된 앰플 및 병 등으로 제공될 수도 있다.The method of administering the pharmaceutical composition may be determined by a person skilled in the art based on the symptoms of the patient and the severity of the disease. In addition, it may be formulated in various forms such as powders, tablets, capsules, liquids, injections, ointments, syrups, etc., and may be provided in unit-dose or multi-dose containers, such as sealed ampoules and bottles. .
본 발명의 약학적 조성물은 경구 또는 비경구 투여가 가능하다. 본 발명에 따른 조성물의 투여경로는 이들로 한정되는 것은 아니지만, 예를 들면, 구강, 정맥 내, 근육 내, 동맥 내, 골수 내, 경막 내, 심장 내, 경피, 피하, 복강 내, 장관, 설하, 또는 국소 투여가 가능하다. 본 발명에 따른 조성물의 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 방법, 배설율, 또는 질병의 중증도 등에 따라 그 범위가 다양하며, 본 기술 분야의 통상의 전문가가 용이하게 결정할 수 있다. 또한, 임상 투여를 위해 공지의 기술을 이용하여 본 발명의 조성물을 적합한 제형으로 제제화할 수 있다.The pharmaceutical composition of the present invention can be administered orally or parenterally. The route of administration of the composition according to the present invention is not limited thereto, but for example, oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, subcutaneous, intraperitoneal, intestinal, sublingual , or topical administration is possible. The dosage of the composition according to the present invention varies according to the patient's weight, age, sex, health status, diet, administration time, method, excretion rate, or severity of disease, etc., and a person of ordinary skill in the art can be easily determined. In addition, the composition of the present invention may be formulated into a suitable dosage form for clinical administration using known techniques.
본 발명은 다른 관점에서, 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 세포 배양용 조성물에 관한 것이다.In another aspect, the present invention relates to a cell culture composition for inducing or enhancing the production of the RNAi-inducing nucleic acid molecule or cone cells or cone-like rod cells comprising the composition.
상기 세포 배양용 조성물은 줄기세포, 망막 전구체 세포, 망막유사체 (retinal organoid) 세포, 광수용체 전구체 세포, 원추세포, 또는 간상세포로 이루어지는 군으로부터 선택되는 어느 하나 이상의 세포를 배양하여 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 것으로, 배지 (medium) 조성물 또는 배지 원료 조성물의 형태로 제공될 수 있다. The cell culture composition comprises culturing any one or more cells selected from the group consisting of stem cells, retinal progenitor cells, retinal organoid cells, photoreceptor progenitor cells, cone cells, or rod cells to obtain cone cells or cone cells. - It is for inducing or enhancing the production of rod-like cells, and may be provided in the form of a medium composition or a medium raw material composition.
상기 줄기세포는 유도만능줄기세포 (induced pluripotent stem cells: iPS cells)일 수 있다. 상기 망막 전구체 세포는 줄기세포가 분화되어 망막 세포가 되기 직전의 세포를 의미하는 것일 수 있다. 상기 망막유사체 (retinal organoid) 세포는 줄기세포를 시험관에서 키워 사람의 망막 구조와 같은 조직을 구현한 세포를 의미하는 것일 수 있다. 상기 광수용체 전구체 세포는 줄기세포가 분화되어 광수용체 세포 즉, 원추세포 또는 간상세포가 되기 직전의 세포를 의미하는 것일 수 있다.The stem cells may be induced pluripotent stem cells (iPS cells). The retinal precursor cells may refer to cells just before stem cells are differentiated into retinal cells. The retinal organoid cells may refer to cells in which stem cells are grown in vitro to implement a tissue such as a human retinal structure. The photoreceptor progenitor cells may refer to cells just before stem cells are differentiated to become photoreceptor cells, that is, cone cells or rod cells.
상기 세포 배양용 조성물은 세포를 분화 또는 증식시킬 수 있는 성분들을 포함하는 배지로서, 이에 한정되는 것은 아니나, DMEM (Dulbecco's Modified Eagle's Medium), 80% knockout DMEM, MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, F-10, F-12, B27, DMEM-F12, α-MEM (α-Minimal Essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Iscove's Modified Dulbecco's Medium), MEM Non-Essential Amino Acids Solution, MacCoy's 5A 배지, AmnioMax, AminoMaxⅡ complete Medium, Chang's Medium MesemCult-XF Medium, 신경 유도 배지 (Neural induction medium), 및 신경 망막 배지 (Neural Retinal Differentiation Medium)로 구성된 군으로부터 선택되는 배지의 구성 원료 또는 성분을 포함할 수 있다. 또한 상기 세포 배양용 조성물은 20% knockout serum replacer (Gibco), 글루타민, 머캅토에탄올, 비필수 아미노산, 베이직 섬유아세포 증식인자 (basic fibroblast growth factor: bFGF), 비트로넥틴 재조합 인간 단백질 (Vitronectin Recombinant Human Protein), N2 Supplement, 헤파린 (Heparin), 소태아혈청 (Fetal bovine serum), 타우린 (Taurine), 글루타맥스, 블레비스타틴 (Blebbistatin) 등을 추가적으로 포함할 수 있다.The composition for cell culture is a medium containing components capable of differentiating or proliferating cells, but is not limited thereto, but is not limited thereto, but is not limited thereto, but is not limited thereto, but is DMEM (Dulbecco's Modified Eagle's Medium), 80% knockout DMEM, MEM (Minimal Essential Medium), BME (Basal). Medium Eagle), RPMI 1640, F-10, F-12, B27, DMEM-F12, α-MEM (α-Minimal Essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Iscove's Modified Dulbecco's Medium), MEM Non-Essential Amino Acids Solution, MacCoy's 5A Medium, AmnioMax, AminoMaxII complete Medium, Chang's Medium MesemCult-XF Medium, Neural induction medium, and Neural Retinal Differentiation Medium It may contain ingredients or components of the medium. In addition, the composition for cell culture is 20% knockout serum replacer (Gibco), glutamine, mercaptoethanol, non-essential amino acids, basic fibroblast growth factor (bFGF), vitronectin recombinant human protein (Vitronectin Recombinant Human Protein) ), N2 Supplement, heparin, fetal bovine serum, taurine, glutamax, blebbistatin, and the like may be additionally included.
상기 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 세포 배양용 조성물은, 상기 RNAi 유도용 핵산 분자를 약 0.01 내지 100 μM, 약 0.01 내지 50 μM, 약 0.01 내지 10 μM, 약 0.01 내지 5 μM, 약 0.01 내지 1 μM, 약 0.01 내지 0.5 μM, 약 0.05 내지 0.5 μM, 또는 약 0.1 내지 0.2 μM의 농도로 포함하는 것일 수 있다. 일 구체예에 따르면, 상기 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 세포 배양용 조성물이 상기 RNAi 유도용 핵산 분자를 상기 수치 범위를 벗어나는 농도로 포함하는 경우, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진 효과가 감소할 수 있다.The composition for cell culture for inducing or enhancing the production of the cone cells or cone cell-like rod cells contains about 0.01 to 100 μM, about 0.01 to 50 μM, about 0.01 to 10 μM, about the RNAi-inducing nucleic acid molecule. It may be included in a concentration of 0.01 to 5 μM, about 0.01 to 1 μM, about 0.01 to 0.5 μM, about 0.05 to 0.5 μM, or about 0.1 to 0.2 μM. According to one embodiment, when the composition for cell culture for inducing or promoting the production of the cone cells or cone cell-like rod cells contains the RNAi-inducing nucleic acid molecule at a concentration outside the numerical range, cone cells or The effect of inducing or enhancing the production of cone-like rods may be reduced.
본 발명은 또 다른 관점에서, 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 이용하여 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 방법에 관한 것이다. 상기 방법은 체외에서 수행되는 것일 수 있다. In another aspect, the present invention relates to a method for inducing or enhancing the production of cone cells or cone-like rod cells using the RNAi-inducing nucleic acid molecule or the composition. The method may be performed in vitro.
상기 방법은 체외에서 줄기세포, 망막 전구체 세포, 망막유사체 세포, 광수용체 전구체 세포, 원추세포, 또는 간상세포로 이루어지는 군으로부터 선택되는 어느 하나 이상의 세포 배양액에 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 첨가하는 단계; 또는 체외에서 줄기세포, 망막 전구체 세포, 망막유사체 세포, 광수용체 전구체 세포, 원추세포, 또는 간상세포로 이루어지는 군으로부터 선택되는 어느 하나 이상의 세포를 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 배지에서 배양하는 단계를 포함할 수 있다.In the method, the RNAi-inducing nucleic acid molecule or the composition is added to any one or more cell culture medium selected from the group consisting of stem cells, retinal progenitor cells, retinal analogue cells, photoreceptor progenitor cells, cone cells, and rod cells in vitro. to do; Or in vitro, any one or more cells selected from the group consisting of stem cells, retinal progenitor cells, retinal analog cells, photoreceptor progenitor cells, cone cells, or rod cells in a medium containing the RNAi-inducing nucleic acid molecule or the composition It may include the step of culturing.
상기 방법에 있어서, 상기 세포 배양액에 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 첨가하는 단계에서, 상기 RNAi 유도용 핵산 분자는, 상기 세포 배양액에 약 0.01 내지 100 μM, 약 0.01 내지 50 μM, 약 0.01 내지 10 μM, 약 0.01 내지 5 μM, 약 0.01 내지 1 μM, 약 0.01 내지 0.5 μM, 약 0.05 내지 0.5 μM, 또는 약 0.1 내지 0.2 μM의 농도로 첨가되는 것일 수 있다. 또는, 상기 방법에 있어서, 상기 세포를 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 포함하는 배지에서 배양하는 단계에서 상기 RNAi 유도용 핵산 분자는, 상기 세포 배양액에 약 0.01 내지 100 μM, 약 0.01 내지 50 μM, 약 0.01 내지 10 μM, 약 0.01 내지 5 μM, 약 0.01 내지 1 μM, 약 0.01 내지 0.5 μM, 약 0.05 내지 0.5 μM, 또는 약 0.1 내지 0.2 μM의 농도로 포함되는 것일 수 있다. 일 구체예에 따르면, 상기 방법에 있어서, 상기 RNAi 유도용 핵산 분자가 상기 수치 범위를 벗어나는 농도로 상기 세포 배양액에 첨가되거나, 포함되는 경우, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진 효과가 감소할 수 있다. In the method, in the step of adding the RNAi-inducing nucleic acid molecule or the composition to the cell culture medium, the RNAi-inducing nucleic acid molecule is added to the cell culture medium at about 0.01 to 100 μM, about 0.01 to 50 μM, about 0.01 It may be added at a concentration of 10 μM to 10 μM, about 0.01 to 5 μM, about 0.01 to 1 μM, about 0.01 to 0.5 μM, about 0.05 to 0.5 μM, or about 0.1 to 0.2 μM. Alternatively, in the method, in the step of culturing the cell in a medium containing the RNAi-inducing nucleic acid molecule or the composition, the RNAi-inducing nucleic acid molecule is added to the cell culture medium at about 0.01 to 100 μM, about 0.01 to 50 It may be included in a concentration of μM, about 0.01 to 10 μM, about 0.01 to 5 μM, about 0.01 to 1 μM, about 0.01 to 0.5 μM, about 0.05 to 0.5 μM, or about 0.1 to 0.2 μM. According to one embodiment, in the method, when the RNAi-inducing nucleic acid molecule is added to or included in the cell culture medium at a concentration outside the numerical range, the effect of inducing or enhancing the production of cone cells or cone-like rods may decrease.
본 발명은 또 다른 관점에서, 상기 RNAi 유도용 핵산 분자 또는 상기 조성물을 개체에 투여하는 단계를 포함하는 안구 또는 망막질환을 개선 또는 치료하는 방법에 관한 것이다.In another aspect, the present invention relates to a method of improving or treating an ocular or retinal disease, comprising administering the RNAi-inducing nucleic acid molecule or the composition to a subject.
본 발명에 따른 상기 방법에 포함되는 구성은 앞서 설명한 발명에 포함되는 구성과 동일하므로, 위 설명은 본 발명에 따른 상기 방법에도 동일하게 적용될 수 있다.Since the configuration included in the method according to the present invention is the same as the configuration included in the above-described invention, the above description is equally applicable to the method according to the present invention.
본 발명은 또 다른 관점에서, 상기 RNAi 유도용 핵산 분자 또는 이를 포함하는 조성물의, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키기 위한 용도, 시력을 개선시키기 위한 용도, 또는 안구 또는 망막질환을 개선 또는 치료하기 위한 용도에 관한 것이다.In another aspect, the present invention provides a nucleic acid molecule for inducing RNAi or a composition comprising the same for inducing or enhancing the production of cone cells or cone-like rod cells, use for improving visual acuity, or ocular or It relates to use for improving or treating retinal diseases.
본 발명에 따른 상기 용도에 포함되는 구성은 앞서 설명한 발명에 포함되는 구성과 동일하므로, 위 설명은 본 발명에 따른 상기 용도에도 동일하게 적용될 수 있다.Since the configuration included in the use according to the present invention is the same as the configuration included in the invention described above, the above description can be equally applied to the use according to the present invention.
본 발명에 따르면, 간상세포 및 원추세포의 분화 과정에서 매우 중요한 기능을 하는 NRL 유전자의 발현을 효율적으로 억제시킬 수 있는 비대칭 RNAi 유도용 핵산 분자를 이용하여 원추세포 또는 원추세포-유사 간상세포의 생성을 유도하거나 증진시킬 수 있다. 이로 인해, 상기 비대칭 RNAi 유도용 핵산 분자는 원추세포의 손실 또는 기능 이상을 보충하여, 시력을 향상시키거나 안구 또는 망막 질환을 개선 또는 치료할 수 있는 치료제로서 유용하게 활용될 수 있다.According to the present invention, a cone cell or cone cell-like rod cell is generated using a nucleic acid molecule for inducing asymmetric RNAi that can efficiently suppress the expression of the NRL gene, which plays a very important function in the differentiation process of rod cells and cone cells. can induce or promote For this reason, the nucleic acid molecule for inducing asymmetric RNAi may be usefully utilized as a therapeutic agent capable of improving vision or improving or treating ocular or retinal diseases by supplementing the loss or dysfunction of cone cells.
도 1은 cp-asiNRL을 처리한 망막유사체 (retinal organoid)와 cp-asiNRL을 처리하지 않은 대조군 망막유사체의 조직절편에 대하여 로돕신 (Rhodosin) 및 L/M-옵신 (L/M-opsin) 항체를 사용하여 면역형광염색한 결과를 나타내는 도면이다 (Scale bar = 100 μm). 도 1에서 상대적으로 낮은 명도로 표현된 부분은 Hoechst 33342에 의해 염색된 세포핵을 나타내고, 상대적으로 높은 명도로 표현된 부분은 로돕신 항체 또는 L/M-옵신 항체에 의해 형광염색된 세포를 나타낸다.1 shows rhodosin (Rhodosin) and L/M-opsin (L/M-opsin) antibodies to tissue sections of retinal organoids treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. It is a diagram showing the results of immunofluorescence staining using (Scale bar = 100 μm). In FIG. 1 , a portion expressed in a relatively low brightness indicates a cell nucleus stained by Hoechst 33342, and a portion expressed in a relatively high brightness indicates a cell fluorescently stained by a rhodopsin antibody or an L/M-opsin antibody.
도 2는 cp-asiNRL을 처리한 망막유사체와 cp-asiNRL을 처리하지 않은 대조군 망막유사체에 대하여 면역형광염색한 이미지로부터 로돕신 항체 및 L/M-옵신 항체에 형광염색된 각각의 광수용세포를 정략적으로 분석한 결과를 나타내는 그래프이다. 2 is a schematic diagram of each photoreceptor cell fluorescently stained with rhodopsin antibody and L/M-opsin antibody from immunofluorescent staining images for retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. It is a graph showing the analysis result.
도 3은 cp-asiNRL을 처리한 망막유사체 및 cp-asiNRL을 처리하지 않은 대조군 망막유사체로부터 추출된 단백질인 로돕신, L/M-옵신, 및 S-옵신 (S-opsin)에 대하여 웨스턴 블랏 분석한 결과를 나타내는 도면이다.Figure 3 shows rhodopsin, L/M-opsin, and S-opsin, which are proteins extracted from the retinal analogs treated with cp-asiNRL and the control retinal analogs not treated with cp-asiNRL, Western blot analysis. It is a drawing showing the result.
이하 본 발명을 실시예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail through examples. However, these examples are for illustrative purposes only, and the scope of the present invention is not limited to these examples.
실시예 1. NRL을 표적하는 세포 관통능 (cell-penetrating ability)이 있는 비대칭 RNAi 유도용 핵산 분자의 제조Example 1. Preparation of a nucleic acid molecule for inducing asymmetric RNAi having cell-penetrating ability to target NRL
비대칭 RNAi 유도용 핵산 분자인, NRL을 표적하는 세포 관통능이 있는 cp-asiNRL의 서열 및 화학적 변형은 이미 WO2020/179702에 개시되어 있는 것으로, 이의 제조도 동 특허문헌에 개시된 바에 따라 제조되었다.The sequence and chemical modification of cp-asiNRL, which is a nucleic acid molecule for inducing asymmetric RNAi, which has cell-penetrating ability to target NRL, has already been disclosed in WO2020/179702, and its preparation was also prepared as disclosed in the same patent document.
구체적으로, 표 1에 NRL을 표적으로 하는 asiNRL의 염기 서열을 나타내었다.Specifically, Table 1 shows the base sequence of asiNRL targeting NRL.
No.No. Sequence (5'->3')Sequence (5'->3') 서열번호SEQ ID NO:
#1#One S (16mer)S (16mer) CGGCGCUGGUCUCGAUCGGCGCUGGUCUCGAU 1One
AS (26mer)AS (26mer) AUCGAGACCAGCGCCGCGUCGGAAAAAUCGAGACCAGCGCCGCGUCGGAAAA 22
상기 표 1의 asiNRL을 세포 관통능을 가지고 핵산가수분해 효소에 저항성을 가지는 cp-asiNRL로 만들기 위해서 modification을 진행하였다. 표 2에 세포 관통능을 가지는 cp-asiNRL의 modification을 나타내었다.Modification was performed to make asiNRL of Table 1 into cp-asiNRL having cell penetrating ability and resistance to nucleolytic enzymes. Table 2 shows the modifications of cp-asiNRL with cell penetrating ability.
cp-asiRNAcp-asiRNA NoNo Sequence (5'→3')Sequence (5'→3')
cp-asiNRLcp-asiNRL #1#One sensesense mC(2'-F-G)mG(2'-F-C)mG(2'-F-C)mU(2'-F-G)mG(2'-F-U)mC(2'-F-U)mC
(2'-F-G)*mA*(2'-F-U)*-Chol
mC(2'-FG)mG(2'-FC)mG(2'-FC)mU(2'-FG)mG(2'-FU)mC(2'-FU)mC
(2'-FG)*mA*(2'-FU)*-Chol
antisenseantisense PmA(2'-F-U)mC(2'-F-G)mA(2'-F-G)mA(2'-F-C)mC(2'-F-A)mG(2'-F-C)
mG(2'-F-C)mC(2'-F-G)mC(2'-F-G)mU(2'-F-C)mG(2'-F-G)*mA*(2'-F-A)
*mA*(2'-F-A)
PmA(2'-FU)mC(2'-FG)mA(2'-FG)mA(2'-FC)mC(2'-FA)mG(2'-FC)
mG(2'-FC)mC(2'-FG)mC(2'-FG)mU(2'-FC)mG(2'-FG)*mA*(2'-FA)
*mA*(2'-FA)
한편, 상기 표 2에 "*", "m", "2'-F-", "-chol", "P"로 표기된 화학적 변형은 표 3에 나타낸 바와 같다. On the other hand, the chemical modifications indicated by "*", "m", "2'-F-", "-chol", and "P" in Table 2 are as shown in Table 3.
표기법notation 도입된 화학적 변형introduced chemical modifications
** 포스포로티오에이트 결합 (phosphorothioated bond)phosphorothioate bond
mm 2'-O-메틸 (Methyl)2'-O-methyl (Methyl)
2'-F-2'-F- 2'-플루오르 (Fluoro)2'-Fluoro
-chol-chol 3'-콜레스테롤 3'-cholesterol
PP 5'-인산기 5'-phosphate group
구체적으로, 상기 표 3에서, "*"은 기존의 포스포다이에스터 결합이 포스포로티오에이트 결합 (phosphorothioated bond)으로 치환된 형태를 의미하는 것이며, "m"은 기존의 2'-OH가 2'-O-메틸 (Methyl)로 치환된 형태를 의미하는 것이다. 또한, "2'-F-"는 예를 들어, 2'-F-G의 경우, 기존의 G (구아닌)의 2'-OH가 플루오르 (Fluoro)로 치환된 형태를 의미하는 것이며, "-Chol"은 3'-말단에 콜레스테롤이 첨가된 형태를 의미하는 것이고, "P"는 5'-말단이 인산화된 것 (예컨대, 상기 안티 센스 가닥의 5' 말단에 phosphate modification 또는 phosphorothioate modification의 추가)을 의미하는 것이다.Specifically, in Table 3, "*" means a form in which the existing phosphodiester bond is substituted with a phosphorothioate bond, and "m" indicates that the existing 2'-OH is 2 It means a form substituted with '-O-methyl (Methyl). In addition, "2'-F-" means, for example, in the case of 2'-F-G, the 2'-OH of the existing G (guanine) is substituted with fluorine (Fluoro), and "-Chol" denotes a form in which cholesterol is added to the 3'-terminus, and "P" denotes a form in which the 5'-terminus is phosphorylated (eg, phosphate modification or phosphorothioate modification is added to the 5' end of the antisense strand) will do
센스 가닥의 3' 말단에 붙은 콜레스테롤은 세포막을 투과할 수 있게 해준다. 또한 센스 가닥과 안티센스 가닥의 3' 말단과 가까운 phosphate backbone을 포스포로티오에이트로 치환한 것은 핵산외부가수분해효소에 대한 저항성을 증가시키며, cellular uptake와 in vivo에서 생물학적인 이용이 가능하게 한다. 또한 일부 당의 2'을 O-메틸로 modification하면 핵산가수분해효소에 대한 저항성이 높아지며, siRNA immunogenicity와 off-target 효과가 감소한다. 마지막으로, 일부 당의 2'을 플루오르로 modification하면 double strand RNA duplex를 안정화시키고 seurm에서의 안정성을 높이며 in vitro, in vivo에서 효율적인 silencing을 가능하게 한다.Cholesterol attached to the 3' end of the sense strand allows it to penetrate the cell membrane. In addition, the substitution of phosphorothioate for the phosphate backbone close to the 3' end of the sense and antisense strands increases resistance to exohydrolase, cellular uptake and in vivo bioavailability. In addition, if 2' of some sugars are modified with O-methyl, resistance to nucleases is increased, and siRNA immunogenicity and off-target effect are reduced. Finally, the 2' modification of some sugars with fluorine stabilizes the double-stranded RNA duplex, increases stability in seurm, and enables efficient silencing in vitro and in vivo.
또한, 안티센스 가닥의 5'-인산화 (phosphorylation)는 유전자 침묵과정에서 필수적인 RNA-induced silencing complex (RISC)에 loading을 위해 siRNA duplex 중 표적 mRNA와 상보적인 strand에 요구된다. 따라서 대부분의 경우에 합성 siRNA는 세포 내로 들어가면, 인산화가 일어난다. 그러나 더 확실한 RISC-loading을 위하여 안티 센스 가닥의 5' 말단에 phosphate modification을 진행하였다.In addition, 5'-phosphorylation of the antisense strand is required for the strand complementary to the target mRNA in the siRNA duplex for loading into the RNA-induced silencing complex (RISC), which is essential for gene silencing. Therefore, in most cases, when synthetic siRNA enters the cell, phosphorylation occurs. However, for more reliable RISC-loading, phosphate modification was performed at the 5' end of the antisense strand.
상기 표 2에 나타낸 바와 같이, 본 실시예를 통해 NRL을 표적하는 세포 관통능이 있는 비대칭 RNAi 유도용 핵산 분자인, cp-asiNRL을 수득하였다. As shown in Table 2 above, cp-asiNRL, a nucleic acid molecule for inducing asymmetric RNAi having cell-penetrating ability to target NRL, was obtained in this example.
참고예 1. cp-asiNRL에 의한 광수용세포 생성 분석을 위한 실험 재료 및 실험 방법 Reference Example 1. Experimental materials and methods for the analysis of photoreceptor cell generation by cp-asiNRL
1-1. 정상인 유래 유도만능줄기세포 (iPS cells) 제작 1-1. Production of normal human-derived induced pluripotent stem cells (iPS cells)
유도만능줄기세포 (iPS cells)는 43세 또는 62세 정상인의 혈액에서 분리한 단핵세포 (mononuclear cells)로 부터 제작되었다. 약 0.5 x 106 개의 단핵세포를 SFEM Ⅱ Supplement (Stemcell Technologies)가 포함된 StemSpan™ SFEM Ⅱ (Stemcell Technologies) 배지에서 약 7 일 동안 배양 후, 리프로그래밍 유전자들이 삽입된 플라스미드 pCXLE-hOCToct3/4-shp53-F, pCXLEe-hSK, 및 pCXLEe-hUL, 및 유전자 발현을 촉진하는 pCXLEe-hOEBNA1을 전기천공법 (Electroporation)을 사용하여 세포에 주입하였다. 플라스미드가 주입된 세포를 i-MATIRX 511 (Takara)로 코팅한 배양접시에서 리프로그래밍 배지 (StemFit® Basic02, AJINOMOTO)를 사용하여 약 8 내지 10 일 동안 성장시키고, 유도만능줄기세포 군집 (colony) 형성이 시작되는 시점에서 줄기세포 배양 배지 (Essential 8™ Medium, ThermoFisher scientific)로 교체하여 약 10 내지 12 일 동안 배양한 후 세포 군집을 채집하였다. 채집된 유도만능줄기세포는 비트로넥틴 재조합 인간 단백질 (Vitronectin Recombinant Human Protein, ThermoFisher scientific)이 코팅된 배양접시에서 약 20 내지 24 회 계대배양한 후 오가노이드 제작에 사용하였다. Induced pluripotent stem cells (iPS cells) were prepared from mononuclear cells isolated from the blood of a 43- or 62-year-old normal human. After culturing about 0.5 x 10 6 mononuclear cells in StemSpan™ SFEM II (Stemcell Technologies) medium containing SFEM II Supplement (Stemcell Technologies) for about 7 days, plasmid pCXLE-hOCToct3/4-shp53 with reprogramming genes inserted -F, pCXLEe-hSK, and pCXLEe-hUL, and pCXLEe-hOEBNA1, which promote gene expression, were injected into cells using electroporation. Cells injected with the plasmid were grown for about 8 to 10 days using a reprogramming medium (StemFit ® Basic02, AJINOMOTO) in a culture dish coated with i-MATIRX 511 (Takara), and induced pluripotent stem cell colony was formed. At this starting point, the stem cell culture medium (Essential 8™ Medium, ThermoFisher scientific) was replaced and cultured for about 10 to 12 days, and then the cell population was collected. The collected induced pluripotent stem cells were passaged about 20 to 24 times in a culture dish coated with Vitronectin Recombinant Human Protein (ThermoFisher scientific), and then used to prepare organoids.
1-2. 망막유사체 (Retinal organoid) 제작 및 cp-asiNRL의 처리1-2. Preparation of retinal organoids and processing of cp-asiNRL
약 60 cm2 배양접시에 약 50 내지 70 개의 군집을 형성한 유도만능줄기세포를 TrypLE™ Select Enzyme (ThermoFisher scientific)을 사용하여 단일 세포로 분리한 후 Ultra-low attachment 6 well plate에 옮겨 약 10 μM (-)-블레비스타틴 ((-)-Blebbistatin)이 포함된 줄기세포 배양 배지가 포함된 Ultra-low attachment dish (Corning, NY, USA)에서 하루 동안 배양하였다. 그 후, 신경 유도 배지 (Neural induction medium; DMEM/F12 (1:1), MEM Non-Essential Amino Acids Solution, N2 Supplement, 약 0.2% Heparin)로 점차적으로 교체하여 약 6 일 동안 배양하여 배상체 (Embryoid body)를 제작하였다. 약 20 개/cm2의 배상체를 Matrigel™ GFR Membrane Matrix (Corning)로 코팅한 배양접시에서 신경 유도 배지와 함께 약 15 일 동안 배양하였다. 신경 구조 (neural structure)가 형성되는 배양 후 약 13 내지 15 일에 신경 망막 배지 (Neural Retinal Differentiation Medium; DMEM/F12 (3:1), B27, MEM Non-Essential Amino Acids Solution)로 교체하여 배양하였고, 배양 후 약 25 내지 30 일에 신경 망막 구조를 보이는 세포체를 채집하였다. 채집된 세포체를 신경 망막 배지가 담긴 Ultra-low attachment dish에서 약 5 일 동안 배양한 후, 배지를 약 10% 소태아혈청 (Fetal bovine serum), 약 100 μM 타우린 (Taurine), 및 1X 글루타맥스 (Glutamax)가 추가된 신경 망막 배지로 교체한 후 약 160 일 동안 배양하여 망막유사체를 수득하였다. About 50 to 70 colonies of induced pluripotent stem cells in a 60 cm 2 culture dish were separated into single cells using TrypLE™ Select Enzyme (ThermoFisher scientific), and then transferred to an ultra-low attachment 6 well plate, about 10 μM (-)-Blebbistatin ((-)-Blebbistatin) was cultured for one day in an ultra-low attachment dish (Corning, NY, USA) containing stem cell culture medium. After that, the embryoid body (Neural induction medium; DMEM/F12 (1:1), MEM Non-Essential Amino Acids Solution, N2 Supplement, about 0.2% Heparin) was gradually replaced and cultured for about 6 days ( Embryoid body) was fabricated. The embryoid bodies of about 20 pieces/cm 2 were cultured in a culture dish coated with Matrigel™ GFR Membrane Matrix (Corning) together with a nerve induction medium for about 15 days. About 13 to 15 days after the culture in which the neural structure is formed, it was replaced with a neural retinal differentiation medium (DMEM/F12 (3:1), B27, MEM Non-Essential Amino Acids Solution) and cultured. , Cell bodies showing neural retinal structures were collected about 25 to 30 days after culture. After culturing the collected cell bodies in an ultra-low attachment dish containing neural retina medium for about 5 days, the medium was cultured with about 10% Fetal bovine serum, about 100 μM Taurine, and 1X Glutamax. (Glutamax) was replaced with the added neural retinal medium and then cultured for about 160 days to obtain a retinal analogue.
cp-asiNRL의 효과를 평가하기 위해 상기 신경 망막 배지 교체 배양 후 약 142 일 시점, 즉 간상세포 및 원추세포의 형성이 이미 이루어진 시점에, 망막유사체를 포함하는 배양액에 cp-asiNRL을 약 48 시간 간격으로 약 3 회에 걸쳐 처리하였다. 이때, 망막유사체를 포함하는 배양액에 포함된 cp-asiNRL의 최종 농도가 약 0.1 또는 0.2 μM이 되도록 처리하였다. cp-asiNRL의 유효성 평가에 약 30 개의 망막유사체를 사용하였다. In order to evaluate the effect of cp-asiNRL, at about 142 days after the neural retinal medium replacement culture, that is, at a time point where rods and cones have already been formed, cp-asiNRL was added to a culture medium containing retinal analogues at about 48 hour intervals. was treated about 3 times. At this time, it was treated so that the final concentration of cp-asiNRL contained in the culture medium containing the retinal analogue was about 0.1 or 0.2 μM. About 30 retinal analogues were used to evaluate the effectiveness of cp-asiNRL.
1-3. 망막유사체의 면역형광염색 분석 1-3. Immunofluorescence staining analysis of retinal analogues
망막유사체에 cp-asiNRL을 처음 처리한 시점으로부터 약 18 일 후, cp-asiNRL을 처리한 망막유사체와 cp-asiNRL을 처리하지 않은 대조군 망막유사체를 약 4% 파라포름알데하이드 (paraformaldehyde: PFA)를 사용하여 약 4℃에서 약 12 시간 동안 고정하였다. 고정된 망막유사체에 냉동손상을 억제하기 위해 약 30% 수크로스 (sucrose)를 침투시키고, 옵티멀 커팅 템퍼러쳐 컴파운드 (optimal cutting temperature compound) (ThermoFisher scientific)를 사용하여 액화 질소에서 망막유사체를 포매 (embedding)하였다. 동결조직은 약 15 μm 두께로 절편하여 글래스 슬라이드 (glass slide)에 마운트 (mount)하였다. 면역화학염색을 위해 동결조직 절편들은 약 15 분 동안 약 4% PFA에서 고정되었고, 약 0.5% 트리톤 X-100 (Triton X-100)에서 조직연화된 후, PBS로 세척 후 약 2% 소혈청알부민 (bovine serum albumin) (Gibco, Dublin, Ireland)과 약 5% 정상염소혈청 (normal goat serum) (Jackson Immunoresearch, PA, USA)으로 약 4℃에서 약 12 시간 동안 블로킹되었다. 그 후, 조직절편을 래빗 로돕신 (Rhodopsin) (Millipore) 및 마우스 L/M 옵신 (L/M opsin) (Millipore) 항체와 함께 약 12 시간 동안 약 4℃에서 반응시킨 다음 PBS로 약 3 회 세척하였다. 세척된 조직절편을 Alexa FluorTM 488 항체 또는 Cy3가 결합되어 있는 2차 항체와 함께 약 1 시간 동안 상온 (약 15 내지 25℃)에서 반응시킨 후, Hoechst 33342 염료를 이용하여 세포핵을 염색하였다. 염색된 조직절편을 AxioCam 카메라 (Zeiss, Jena, Germany)가 장착된 형광현미경으로 촬영하여 형광 이미지를 수득하였다. About 18 days after the first treatment with cp-asiNRL on the retinal analog, about 4% paraformaldehyde (PFA) was used for the retinal analog treated with cp-asiNRL and the control retinal analog not treated with cp-asiNRL. and fixed at about 4°C for about 12 hours. In order to suppress cryo damage to the fixed retinal analogs, about 30% sucrose was infiltrated, and the retinal analogs were embedded in liquid nitrogen using an optimal cutting temperature compound (ThermoFisher scientific). ) was done. The frozen tissue was sectioned to a thickness of about 15 μm and mounted on a glass slide. For immunochemical staining, frozen tissue sections were fixed in about 4% PFA for about 15 minutes, softened in about 0.5% Triton X-100, washed with PBS, and then washed with about 2% bovine serum albumin. (bovine serum albumin) (Gibco, Dublin, Ireland) and about 5% normal goat serum (Jackson Immunoresearch, PA, USA) were blocked at about 4°C for about 12 hours. Thereafter, the tissue sections were reacted with rabbit rhodopsin (Millipore) and mouse L/M opsin (Millipore) antibodies at about 4° C. for about 12 hours, and then washed with PBS about 3 times. . After the washed tissue sections were reacted with Alexa Fluor TM 488 antibody or a secondary antibody to which Cy3 is bound at room temperature (about 15 to 25° C.) for about 1 hour, cell nuclei were stained using Hoechst 33342 dye. The stained tissue sections were photographed with a fluorescence microscope equipped with an AxioCam camera (Zeiss, Jena, Germany) to obtain fluorescence images.
1-4. 망막유사체의 웨스턴 블랏 분석 1-4. Western blot analysis of retinal analogues
망막유사체에 cp-asiNRL을 처음 처리한 시점으로부터 약 18 일 후, 망막유사체로부터 RIPA 버퍼를 사용하여 단백질을 추출하였다. 동량의 단백질 (약 20 μg)을 SDS-PAGE (Sodium Dodecyl Sulfate-Poly Acrylamide Gel Electrophoresis)에 의해 분리하고, 플루오르화 폴리비닐리덴 (PVDF) 막 (Millipore, Billerica, MA)으로 블랏팅하였다. 상기 막을 상온 (약 15 내지 25℃)에서 약 2 시간 동안 약 5% 소혈청알부민 (bovine serum albumin) 및 약 0.1% 트윈 20 (Tween 20)이 포함된 TBS에서 반응시킨 후, 래빗 로돕신 (Millipore), 마우스 L/M 옵신 (Millipore), 및 래빗 S-opsin (S-옵신) (Millipore) 항체와 함께 약 4℃에서 오버-나잇 배양하였다. 그 후, 막을 TBST로 세척한 후, HRP (Horse Radish Peroxidase)가 결합된 2차 항체 (ThermoFisher scientific)와 함께 약 1 시간 동안 배양하였다. 배양 후, 막의 면역 활성 단백질을 루미네선스 (luminescence) 기질 (ClarityTM Western ECL Substrate; Bio-Rad Laboratories Inc, Hercules, CA)을 사용하여 화학반응시켰고, 그 결과를 디지털 이미징 시스템 (ChemiDoc Touch Imager; Bio-Rad)을 이용하여 가시화하였다. After about 18 days from the time when the retinal analog was first treated with cp-asiNRL, the protein was extracted from the retinal analog using RIPA buffer. Equal amounts of protein (about 20 μg) were separated by SDS-PAGE (Sodium Dodecyl Sulfate-Poly Acrylamide Gel Electrophoresis) and blotted with polyvinylidene fluoride (PVDF) membrane (Millipore, Billerica, MA). After the membrane was reacted in TBS containing about 5% bovine serum albumin and about 0.1% Tween 20 at room temperature (about 15 to 25° C.) for about 2 hours, rabbit rhodopsin (Millipore) , mouse L/M opsin (Millipore), and rabbit S-opsin (S-opsin) (Millipore) antibodies were incubated over-night at about 4°C. Thereafter, the membrane was washed with TBST, and then incubated for about 1 hour with a secondary antibody (ThermoFisher scientific) bound to Horse Radish Peroxidase (HRP). After incubation, the membrane immunoactive protein was chemically reacted using a luminescence substrate (Clarity™ Western ECL Substrate; Bio-Rad Laboratories Inc, Hercules, CA), and the results were recorded with a digital imaging system (ChemiDoc Touch Imager; Bio -Rad) was used for visualization.
실험예 1. cp-asiNRL에 의한 광수용세포 생성 분석 Experimental Example 1. Analysis of photoreceptor cell generation by cp-asiNRL
1-1. cp-asiNRL에 의한 광수용세포 생성의 면역형광염색 분석1-1. Immunofluorescence staining analysis of photoreceptor cell generation by cp-asiNRL
본 실험예에서는, 광수용세포의 생성과정에서 cp-asiNRL에 의하여 NRL 유전자의 발현이 억제되는 경우, 광수용세포 중 간상세포 (rod cell)의 감소 및 원추세포 (cone cell)의 증가를 유도할 수 있는지를 확인하기 위하여 면역형광염색 분석을 수행하였다. In this experimental example, when the expression of the NRL gene is suppressed by cp-asiNRL during the generation of photoreceptors, it is possible to induce a decrease in rod cells and an increase in cone cells among the photoreceptors. Immunofluorescence staining analysis was performed to confirm that it can.
구체적으로, 유도만능줄기세포로부터 제작된 망막유사체에 cp-asiNRL을 약 0.1 μM 농도로 처리하였다. 상기 cp-asiNRL의 처리는 간상세포와 원추세포의 형성이 이미 이루어진 시점인, 망막유사체 배양 후 약 142 일에 수행되었다 (상기 참고예 1-2 참고). cp-asiNRL 투여 후 약 18 일 (망막유사체 배양 후 약 160 일)에 면역형광염색법을 이용하여 간상세포와 원추세포의 생성 양상을 분석하였다. 망막유사체에서 로돕신 항체에 형광염색된 세포는 간상세포를, L/M-옵신 항체에 형광염색된 세포는 원추세포를 나타낸다. 대조군으로서 cp-asiNRL을 처리하지 않은 망막유사체를 사용하였다. 구체적인 실험 재료 및 실험 방법은 상기 참고예에 상세히 기재하였다. Specifically, the retinal analogues prepared from induced pluripotent stem cells were treated with cp-asiNRL at a concentration of about 0.1 μM. The cp-asiNRL treatment was performed at about 142 days after culturing the retinal analog, which is a time point at which rod cells and cone cells were already formed (refer to Reference Example 1-2 above). About 18 days after cp-asiNRL administration (about 160 days after culturing retinal analogues), the production patterns of rods and cones were analyzed using immunofluorescence staining. Cells fluorescently stained with rhodopsin antibody in retinal analogues represent rod cells, and cells fluorescently stained with L/M-opsin antibody represent cone cells. As a control, retinal analogues not treated with cp-asiNRL were used. Specific test materials and test methods were described in detail in the reference example above.
도 1은 cp-asiNRL을 처리한 망막유사체와 cp-asiNRL을 처리하지 않은 대조군 망막유사체의 조직절편에 대하여 로돕신 및 L/M-옵신 항체를 사용하여 면역형광염색한 결과를 나타내는 도면이다 (Scale bar = 100 μm). 도 1에서 상대적으로 낮은 명도로 표현된 부분은 Hoechst 33342에 의해 염색된 세포핵을 나타내고, 상대적으로 높은 명도로 표현된 부분은 로돕신 항체 또는 L/M-옵신 항체에 의해 형광염색된 세포를 나타낸다.1 is a diagram showing the results of immunofluorescence staining using rhodopsin and L/M-opsin antibody for tissue sections of retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL (Scale bar) = 100 μm). In FIG. 1 , a portion expressed in a relatively low brightness indicates a cell nucleus stained by Hoechst 33342, and a portion expressed in a relatively high brightness indicates a cell fluorescently stained by a rhodopsin antibody or an L/M-opsin antibody.
그 결과, 도 1에 나타낸 바와 같이, 망막유사체에 cp-asiNRL을 약 0.1 μM 농도로 처리한 경우, 대조군과 비교하여 로돕신 항체에 형광염색된 간상세포의 수는 감소한 반면, L/M-옵신 항체에 형광염색된 원추세포의 수는 증가하였음을 확인하였다.As a result, as shown in FIG. 1, when the retinal analogue was treated with cp-asiNRL at a concentration of about 0.1 μM, the number of rods fluorescently stained with the rhodopsin antibody was reduced compared to the control, whereas the L/M-opsin antibody It was confirmed that the number of fluorescently stained cone cells increased.
또한, 약 0.1 μM 농도로 cp-asiNRL을 처리한 4 개의 망막유사체와 cp-asiNRL을 처리하지 않은 5 개의 대조군 망막유사체에 대하여 면역형광염색 시행 후 무작위로 얻은 이미지로부터 정량적 분석을 수행하였다.In addition, quantitative analysis was performed from images randomly obtained after immunofluorescence staining for 4 retinal analogs treated with cp-asiNRL at a concentration of about 0.1 μM and 5 control retinal analogs not treated with cp-asiNRL.
도 2는 cp-asiNRL을 처리한 망막유사체와 cp-asiNRL을 처리하지 않은 대조군 망막유사체에 대하여 면역형광염색한 이미지로부터 로돕신 항체 및 L/M-옵신 항체에 형광염색된 각각의 광수용세포를 정략적으로 분석한 결과를 나타내는 그래프이다. 2 is a schematic diagram of each photoreceptor cell fluorescently stained with rhodopsin antibody and L/M-opsin antibody from immunofluorescent staining images for retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. It is a graph showing the analysis result.
그 결과, cp-asiNRL을 처리한 망막유사체에서 로돕신 항체에 의해 염색된 간상세포의 수는, 대조군과 비교하여 0.2 mm2 당 19 ± 1.85 개의 수적 감소를 나타낸 반면, L/M-옵신 항체에 의해 염색된 원추세포 수는, 대조군과 비교하여 0.2 mm2 당 19 ± 1.85 개의 수적 증가를 나타냄을 확인하였다.As a result, the number of rod cells stained by the rhodopsin antibody in the retinal analogs treated with cp-asiNRL showed a decrease in the number of 19 ± 1.85 per 0.2 mm 2 compared to the control, whereas by the L/M-opsin antibody It was confirmed that the number of stained cone cells showed an increase in the number of 19 ± 1.85 per 0.2 mm 2 compared to the control.
1-2. cp-asiNRL에 의한 광수용세포의 마커 단백질 발현 수준 분석 1-2. Analysis of marker protein expression level in photoreceptor cells by cp-asiNRL
본 실험예에서는, 광수용세포의 생성과정에서 cp-asiNRL에 의하여 NRL 유전자의 발현이 억제되는 경우의 광수용세포 중 간상세포 및 원추세포의 생성 양상을 분석하기 위하여, 간상세포 및 원추세포 각각의 마커인 로돕신 및 L/M-옵신의 발현 수준을 분석하였다.In this experimental example, in order to analyze the production patterns of rods and cones among photoreceptors when the expression of the NRL gene is suppressed by cp-asiNRL during the generation of photoreceptors, each of rods and cones The expression levels of the markers rhodopsin and L/M-opsin were analyzed.
구체적으로, 망막유사체에 cp-asiNRL을 약 0.1 μM 또는 약 0.2 μM 농도로 처리한 후 약 18 일 시점 (망막유사체 배양 후 160 일)에, 망막유사체로부터 단백질을 추출하였다. 즉, cp-asiNRL이 약 0.1 μM 농도로 처리된 7 개의 망막유사체, cp-asiNRL이 약 0.2 μM 농도로 처리된 7 개의 망막유사체, 및 cp-asiNRL이 처리되지 않은 7 개의 대조군 망막유사체로부터 단백질을 추출하였다. 추출된 단백질에 대하여, 로돕신, L/M-옵신, 및 S-옵신에 대한 항체를 이용하여 웨스턴 블랏 분석을 수행하였다. 구체적인 실험 재료 및 실험 방법은 상기 참고예에 상세히 기재하였다.Specifically, after treatment with cp-asiNRL at a concentration of about 0.1 μM or about 0.2 μM to the retinal analog, the protein was extracted from the retinal analog at about 18 days (160 days after retinal analog culture). That is, proteins from 7 retinal analogues treated with cp-asiNRL at a concentration of about 0.1 μM, 7 retinal analogues treated with cp-asiNRL at a concentration of about 0.2 μM, and 7 control retinal analogues not treated with cp-asiNRL were isolated. extracted. Western blot analysis was performed on the extracted proteins using antibodies against rhodopsin, L/M-opsin, and S-opsin. Specific test materials and test methods were described in detail in the reference example above.
도 3은 cp-asiNRL을 처리한 망막유사체 및 cp-asiNRL을 처리하지 않은 대조군 망막유사체로부터 추출된 단백질인 로돕신, L/M-옵신, 및 S-옵신에 대하여 웨스턴 블랏 분석한 결과를 나타내는 도면이다. 3 is a view showing the results of western blot analysis for rhodopsin, L/M-opsin, and S-opsin, which are proteins extracted from retinal analogs treated with cp-asiNRL and control retinal analogs not treated with cp-asiNRL. .
그 결과, 도 3에 나타낸 바와 같이, 망막유사체에 처리한 cp-asiNRL의 농도가 증가함에 따라, 로돕신 단백질의 발현은 감소한 반면, L/M-옵신 및 S-옵신 단백질의 발현은 증가함을 확인하였다.As a result, as shown in Figure 3, as the concentration of cp-asiNRL treated on the retinal analog increased, the expression of rhodopsin protein decreased, while the expression of L/M-opsin and S-opsin protein increased. did.
본 실험예를 통해, 광수용세포의 생성과정에서 cp-asiNRL에 의하여 NRL 유전자의 발현을 억제하는 경우, 광수용세포 중 간상세포의 수적 감소 및 원추세포의 수적 증가를 유도할 수 있음을 확인하였다.Through this experimental example, it was confirmed that when the expression of the NRL gene was suppressed by cp-asiNRL during the generation of photoreceptors, it was possible to induce a decrease in the number of rod cells and an increase in the number of cone cells among the photoreceptors. .
이러한 실험 결과는, cp-asiNRL의 처리 시점이 광수용세포 중 간상세포 및 원추세포의 세포 운명이 결정되어 각 세포의 형성이 이미 이루어진 시점 (예컨대, 간상세포 또는 원추세포로의 분화가 시작된 이후의 시점이거나, 간상세포 또는 원추세포로의 분화가 완료된 시점)이라는 점에 비추어, cp-asiNRL은, 간상세포가 생성되는 과정에서 간상세포 본래의 성질이 아닌 원추세포의 성질을 갖는 원추세포-유사 간상세포의 생성을 유도하거나, 이미 생성된 건강한 간상세포의 성질을 원추세포의 성질로 변화시킴으로써, 간상세포를 원추세포-유사 간상세포로 전환시킬 수 있거나, 이미 생성된 건강한 원추세포의 증식을 증가시킬 수 있고, 이로 인해 간상세포의 수적 감소 및 원추세포의 수적 증가를 유도할 수 있음을 시사한다. 즉, cp-asiNRL은 원추세포의 생성을 증진시키거나, 원추세포-유사 간상세포의 생성을 유도 또는 증진시킬 수 있음을 시사한다.These experimental results show that the time point of cp-asiNRL treatment is the time point at which the cell fates of rods and cones among photoreceptor cells are determined and the formation of each cell has already taken place (eg, after differentiation into rod or cone cells has begun). In view of the time point or time point when differentiation into rod or cone cells is completed), cp-asiNRL is a cone-like rod having cone-like properties rather than the original properties of rod cells in the process of rod cell generation. By inducing the production of cells or changing the properties of already generated healthy rods to those of cones, rods can be converted into cone-like rods, or the proliferation of healthy cones that have already been generated can be increased. This suggests that it can induce a decrease in the number of rod cells and an increase in the number of cone cells. That is, cp-asiNRL suggests that it can enhance the production of cone cells or induce or enhance the production of cone-like rod cells.
따라서, cp-asiNRL은 원추세포가 손실되었거나 또는 기능 이상인 상태에서, 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시켜, 원추세포의 기능 이상을 보충하는데 기여할 수 있으므로, cp-asiNRL은 시력 향상을 위한 안구 또는 망막질환 개선 또는 치료용 약학적 조성물의 유효 성분으로 활용될 수 있다.Therefore, cp-asiNRL can contribute to replenishing the dysfunction of cone cells by inducing or enhancing the production of cone cells or cone-like rods in a state in which cone cells are lost or dysfunctional. It can be used as an active ingredient of a pharmaceutical composition for improving or treating eye or retinal diseases for improving eyesight.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다고 할 것이다.As the specific parts of the present invention have been described in detail above, for those of ordinary skill in the art, these specific descriptions are only preferred embodiments, and it is clear that the scope of the present invention is not limited thereto. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (10)

  1. 서열번호 1의 염기 서열을 포함하는 센스 가닥 및 서열번호 2의 염기 서열을 포함하는 안티센스 가닥을 포함하고, 상기 안티센스 가닥의 5' 말단 및 센스 가닥의 3' 말단은 블런트 말단 (blunt end)을 형성하는 것인 RNAi 유도용 핵산 분자를 포함하는, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.A sense strand comprising the nucleotide sequence of SEQ ID NO: 1 and an antisense strand comprising the nucleotide sequence of SEQ ID NO: 2, wherein the 5' end of the antisense strand and the 3' end of the sense strand form a blunt end A composition for inducing or enhancing the production of cones or cone cells-like rods, comprising a nucleic acid molecule for inducing RNAi to that.
  2. 청구항 1에 있어서, 상기 센스 가닥 또는 안티센스 가닥은 하나 이상의 화학적 변형 (chemical modification)을 포함하는 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.The method according to claim 1, wherein the sense strand or the antisense strand comprises at least one chemical modification, the composition for inducing or enhancing the production of cone cells or cone-like rod cells.
  3. 청구항 2에 있어서, 상기 화학적 변형은 다음으로 구성된 군에서 선택된 하나 이상을 포함하는 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물:The method according to claim 2, wherein the chemical modification comprises one or more selected from the group consisting of, cone cells or cone cell-like composition for inducing or enhancing the production of rods:
    뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -CH3 (methyl), -OCH3 (methoxy), -NH2, -F (불소), -O-2-메톡시에틸-O-프로필 (propyl), -O-2-메틸티오에틸 (methylthioethyl), -O-3-아미노프로필, 또는 -O-3-디메틸아미노프로필로 치환;-OH group at the 2' carbon position of the sugar structure in the nucleotide is -CH 3 (methyl), -OCH 3 (methoxy), -NH 2 , -F (fluorine), -O-2-methoxyethyl-O-propyl ( propyl), -O-2-methylthioethyl, -O-3-aminopropyl, or -O-3-dimethylaminopropyl;
    뉴클레오티드 내 당 (sugar) 구조의 산소가 황으로 치환;replacement of oxygen in the nucleotide structure with sulfur;
    뉴클레오티드 결합이 포스포로티오에이트 (phosphorothioate), 보라노포스페이트 (boranophosphate), 또는 메틸포스포네이트 (methyl phosphonate)로 변형;nucleotide linkages are modified with phosphorothioate, boranophosphate, or methyl phosphonate;
    PNA (peptide nucleic acid), LNA (locked nucleic acid), 또는 UNA (unlocked nucleic acid) 형태로의 변형; 및transformation into peptide nucleic acid (PNA), locked nucleic acid (LNA), or unlocked nucleic acid (UNA) form; and
    인산기 (phosphate group), 친유성 화합물 (lipophilic compound), 또는 세포 침투 펩타이드 결합.A phosphate group, a lipophilic compound, or a cell penetrating peptide bond.
  4. 청구항 3에 있어서, 상기 친유성 화합물은 콜레스테롤, 토코페롤, 스테아르산 (stearic acid), 레티노산 (retinoic acid), DHA (docosahexaenoic acid), 팔미트산 (palmitic acid), 리놀레산 (linoleic acid), 리놀렌산 (linolenic acid), 및 탄소수 10 개 이상의 장쇄지방산으로 이루어지는 군으로부터 선택되는 어느 하나 이상인 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.The method according to claim 3, wherein the lipophilic compound is cholesterol, tocopherol, stearic acid, retinoic acid, docosahexaenoic acid (DHA), palmitic acid, linoleic acid, linolenic acid ( linolenic acid), and any one or more selected from the group consisting of a long-chain fatty acid having 10 or more carbon atoms, a composition for inducing or enhancing the production of cones or cone cells-like rods.
  5. 청구항 2에 있어서, 상기 RNAi 유도용 핵산 분자는 The method according to claim 2, wherein the nucleic acid molecule for inducing RNAi is
    상기 센스 가닥 또는 안티센스 가닥 중 2 개 이상의 뉴클레오티드 내 당 구조의 2' 탄소 위치에서 -OH기가 -OCH3 또는 -F로 치환되는 변형;a modification in which the -OH group is substituted with -OCH 3 or -F at the 2' carbon position of the sugar structure within two or more nucleotides of the sense strand or the antisense strand;
    상기 센스 가닥 또는 안티센스 가닥에서 10% 이상의 뉴클레오티드 결합이 포스포로티오에이트로 변형;at least 10% of the nucleotide linkages in the sense strand or antisense strand are modified to phosphorothioate;
    상기 센스 가닥의 3' 말단에 콜레스테롤 결합; 및cholesterol binding to the 3' end of the sense strand; and
    상기 안티센스 가닥의 5' 말단에 인산기 결합으로 이루어지는 군으로부터 선택되는 하나 이상의 변형을 포함하는 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.A composition for inducing or enhancing the production of cones or cone cells-like rods, comprising one or more modifications selected from the group consisting of a phosphate group at the 5' end of the antisense strand.
  6. 청구항 2에 있어서, 상기 RNAi 유도용 핵산 분자는 하기의 센스 가닥을 포함하며,The method according to claim 2, wherein the nucleic acid molecule for inducing RNAi comprises the following sense strand,
    mC(2'-F-G)mG(2'-F-C)mG(2'-F-C)mU(2'-F-G)mG(2'-F-U)mC(2'-F-U)mC(2'-F-G)*mA*(2'-F-U)*-Chol,mC(2'-F-G)mG(2'-F-C)mG(2'-F-C)mU(2'-F-G)mG(2'-F-U)mC(2'-F-U)mC(2'-F-G)* mA*(2'-F-U)*-Chol,
    상기에서, *는 포스포로티오에이트 결합 (phosphorothioated bond)으로의 변형, m은 2'-O-메틸 (Methyl)로의 치환, 2'-F-는 2'-플루오르 (Fluoro)로의 치환, -chol은 3'-콜레스테롤의 결합을 의미하는 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.In the above, * is a phosphorothioate bond (phosphorothioated bond) modification, m is 2'-O-methyl (Methyl) substitution, 2'-F- is 2'-Fluoro substitution, -chol is 3'- that means the binding of cholesterol, cone cells or cone cells-like rod cell production induction or enhancement composition.
  7. 청구항 2에 있어서, 상기 RNAi 유도용 핵산 분자는 하기의 안티센스 가닥을 포함하며,The method according to claim 2, wherein the nucleic acid molecule for inducing RNAi comprises the following antisense strand,
    PmA(2'-F-U)mC(2'-F-G)mA(2'-F-G)mA(2'-F-C)mC(2'-F-A)mG(2'-F-C)mG(2'-F-C)mC(2'-F-G)mC(2'-F-G)mU(2'-F-C)mG(2'-F-G)*mA*(2'-F-A)*mA*(2'-F-A),PmA(2'-F-U)mC(2'-F-G)mA(2'-F-G)mA(2'-F-C)mC(2'-F-A)mG(2'-F-C)mG(2'-F-C)mC (2'-F-G)mC(2'-F-G)mU(2'-F-C)mG(2'-F-G)*mA*(2'-F-A)*mA*(2'-F-A),
    상기에서, *는 포스포로티오에이트 결합으로의 변형, m은 2'-O-메틸로의 치환, 2'-F-는 2'-플루오르로의 치환, P는 5'-인산기의 결합을 의미하는 것인, 원추세포 또는 원추세포-유사 간상세포 생성 유도 또는 증진용 조성물.In the above, * denotes a phosphorothioate bond, m denotes a substitution with 2'-O-methyl, 2'-F- denotes a substitution with 2'-fluoro, and P denotes a bond with a 5'-phosphate group. The composition for inducing or enhancing the production of cone cells or cone cells-like rod cells.
  8. 청구항 1 내지 7 중 어느 한 항의 조성물을 포함하는 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진하기 위한 세포 배양용 조성물.A composition for cell culture for inducing or promoting the production of cone cells or cone cells-like rod cells comprising the composition of any one of claims 1 to 7.
  9. 청구항 1 내지 7 중 어느 한 항의 조성물을 이용하여 원추세포 또는 원추세포-유사 간상세포의 생성을 유도 또는 증진시키는 방법.A method for inducing or enhancing the production of cones or cone-like rods using the composition of any one of claims 1 to 7.
  10. 청구항 9에 있어서, 상기 방법은 10. The method of claim 9, wherein the method
    체외에서 줄기세포, 망막 전구체 세포, 망막유사체 (retinal organoid) 세포, 광수용체 전구체 세포, 원추세포, 또는 간상세포로 이루어지는 군으로부터 선택되는 어느 하나 이상의 세포 배양액에 청구항 1 내지 7 중 어느 한 항의 조성물을 첨가하는 단계; 또는 In vitro, the composition of any one of claims 1 to 7 in any one or more cell culture medium selected from the group consisting of stem cells, retinal progenitor cells, retinal organoid cells, photoreceptor progenitor cells, cone cells, or rod cells. adding; or
    체외에서 줄기세포, 망막 전구체 세포, 망막유사체 세포, 광수용체 전구체 세포, 원추세포, 또는 간상세포로 이루어지는 군으로부터 선택되는 어느 하나 이상의 세포를 청구항 1 내지 7 중 어느 한 항의 조성물을 포함하는 배지에서 배양하는 단계를 포함하는 것인, 방법.Any one or more cells selected from the group consisting of stem cells, retinal progenitor cells, retinal analog cells, photoreceptor progenitor cells, cone cells, or rod cells in vitro are cultured in a medium containing the composition of any one of claims 1 to 7 A method comprising the step of:
PCT/KR2021/018460 2020-12-07 2021-12-07 Composition for inducing or enhancing generation of cones or cone-like rods WO2022124760A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020200169854A KR20220080597A (en) 2020-12-07 2020-12-07 Composition for inducing or enhancing the production of cone cells or cone cell-like rod cells
KR10-2020-0169854 2020-12-07

Publications (1)

Publication Number Publication Date
WO2022124760A1 true WO2022124760A1 (en) 2022-06-16

Family

ID=81973411

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/018460 WO2022124760A1 (en) 2020-12-07 2021-12-07 Composition for inducing or enhancing generation of cones or cone-like rods

Country Status (2)

Country Link
KR (1) KR20220080597A (en)
WO (1) WO2022124760A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140046339A (en) * 2012-10-10 2014-04-18 서울대학교산학협력단 Method for differentiation into retinal cells from stem cells using inhibition of mirna-203
KR20200090132A (en) * 2019-01-18 2020-07-28 올릭스 주식회사 Asymmetric siRNA Inhibiting Expression of NRL
WO2020154693A1 (en) * 2019-01-25 2020-07-30 Nayan Therapeutics, Inc. Nrl expression reducing oligonucleotides, compositions containing the same, and methods of their use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140046339A (en) * 2012-10-10 2014-04-18 서울대학교산학협력단 Method for differentiation into retinal cells from stem cells using inhibition of mirna-203
KR20200090132A (en) * 2019-01-18 2020-07-28 올릭스 주식회사 Asymmetric siRNA Inhibiting Expression of NRL
WO2020154693A1 (en) * 2019-01-25 2020-07-30 Nayan Therapeutics, Inc. Nrl expression reducing oligonucleotides, compositions containing the same, and methods of their use

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MOORE SPENCER M., SKOWRONSKA-KRAWCZYK DOROTA, CHAO DANIEL L.: "Targeting of the NRL Pathway as a Therapeutic Strategy to Treat Retinitis Pigmentosa", JOURNAL OF CLINICAL MEDICINE, vol. 9, no. 7, 1 January 2020 (2020-01-01), pages 1 - 18, XP055941958, DOI: 10.3390/jcm9072224 *
WENHAN YU, SUDDHASIL MOOKHERJEE, VIJENDER CHAITANKAR, SUJA HIRIYANNA, JUNG-WOONG KIM, MATTHEW BROOKS, YASAMAN ATAEIJANNATI, XUN SU: "Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice", NATURE COMMUNICATIONS, vol. 8, 1 January 2017 (2017-01-01), pages 1 - 15, XP055479667, DOI: 10.1038/ncomms14716 *

Also Published As

Publication number Publication date
KR20220080597A (en) 2022-06-14

Similar Documents

Publication Publication Date Title
WO2019050071A1 (en) Composition for preventing or treating liver fibrosis, containing exosome or exosome-derived ribonucleic acid
WO2020149702A1 (en) Asymmetric sirna for inhibiting expression of neural retina leucine zipper (nrl)
WO2014181954A1 (en) Culture medium composition for improving regenerative capacity of stem cells, and stem cell culturing method using same
WO2017135795A1 (en) Mesenchymal stem cell expressing hepatocyte growth factor, and use thereof
WO2023011248A1 (en) Gene therapy for wet age-related macular degeneration using ipsc-derived cells as vectors
WO2018030630A1 (en) Nonviral minicircle vector carrying sox gene and construction method therefor
TWI285219B (en) Method and composition for inducing neural differentiation
US20070054399A1 (en) Method for differentiating mesenchymal stem cell into neural cell and pharmaceutical composition containing the neural cell for neurodegenerative disease
WO2022124760A1 (en) Composition for inducing or enhancing generation of cones or cone-like rods
WO2022035243A1 (en) Oral composition comprising milk exosomes encapsulating therapeutic agent, and method for producing same
Tao et al. Noggin induces human bone marrow-derived mesenchymal stem cells to differentiate into neural and photoreceptor cells
AU2009280769A1 (en) Agent for promoting neuronal differentiation and method therefor
WO2019066519A1 (en) Pharmaceutical composition for prevention or treatment of age-related macular degeneration containing rna complex targeting connective tissue growth factor
JP2022502047A (en) Methods of isolation and culture of human retinal progenitor cells
WO2012133992A1 (en) Method for differentiating adipose-derived mesenchymal stem cells into chondrocytes
CN112094808B (en) MiR-204-containing exosome and preparation method and application thereof
WO2021125762A1 (en) Composition for preventing or treating dementia, containing peptide nucleic acid complex with blood-brain barrier penetration ability as active ingredient
WO2008149356A1 (en) Methods of generating dopaminergic cells and uses thereof
WO2022124739A1 (en) Nucleic acid molecule for induction of asymmetric rnai for inhibiting expression of ror-beta
WO2016178519A1 (en) Composition for inhibiting muscle aging or treating muscle aging related diseases
KR20140046339A (en) Method for differentiation into retinal cells from stem cells using inhibition of mirna-203
US20220162606A1 (en) Nrl expression reducing oligonucleotides, compositions containing the same, and methods of their use
KR20180028094A (en) Composition for Improving or Treating Spinal Cord Injury Comprising Induced Neural Stem Cells
CN102618490B (en) Method for inducing retinal stem cells to differentiate into photosensory cells
WO2015056970A1 (en) Composition for preventing or treating osteoporosis, containing progranulin inhibitor as active ingredient

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21903806

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21903806

Country of ref document: EP

Kind code of ref document: A1