[go: up one dir, main page]

WO2025225866A1 - Hypoimmunogenic universal induced pluripotent stem cells - Google Patents

Hypoimmunogenic universal induced pluripotent stem cells

Info

Publication number
WO2025225866A1
WO2025225866A1 PCT/KR2025/003036 KR2025003036W WO2025225866A1 WO 2025225866 A1 WO2025225866 A1 WO 2025225866A1 KR 2025003036 W KR2025003036 W KR 2025003036W WO 2025225866 A1 WO2025225866 A1 WO 2025225866A1
Authority
WO
WIPO (PCT)
Prior art keywords
hla
gene encoding
exon
region
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/KR2025/003036
Other languages
French (fr)
Korean (ko)
Inventor
김주련
주지현
남유준
전도영
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Yipscell Inc
Original Assignee
Yipscell Inc
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 Yipscell Inc filed Critical Yipscell Inc
Publication of WO2025225866A1 publication Critical patent/WO2025225866A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • 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
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPR]
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to a universal induced pluripotent stem cell with low immunogenicity.
  • immunogenicity is a critical factor in determining the success of allogeneic transplantation.
  • stem cells as cell therapy has been conducted using adult stem cells, MSCs, ESCs, and iPSCs.
  • iPSCs are somatic cells that have been engineered with Yamanaka factors to achieve pluripotency and infinite proliferation, enabling differentiation into target cells such as heart, nerve, cartilage, and bone.
  • MHC The major histocompatibility complex
  • HLA Human Leukocyte Antigen
  • HLA-A, B, C HLA class I
  • DR, DQ, DP class II molecules
  • Class I MHC, HLA-A, B, C has the role of presenting foreign proteins to cytotoxic T cells and is composed of a polymorphic ⁇ chain and a non-polymorphic Beta 2M.
  • Class II MHC, HLA-DR, DQ, DP has a polymorphic beta 1 and a non-polymorphic alpha chain and Beta 2, and has the role of presenting foreign proteins to helper T cells.
  • HLA Human leukocyte antigens
  • A, B, C HLA class I
  • DR, DQ, DP class II
  • HLA protein polymorphism is essential for immune protection, but it often leads to failure due to immune responses when transplanting organs from different genetic backgrounds. This same principle applies to cell therapy.
  • donor cells are classified as non-self due to mismatched HLA types, they can be attacked by CD4 T cells, CD8 T cells, and NK cells, which in turn triggers an immune response.
  • One aspect is to provide hypoimmunogenic stem cells genetically engineered to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and HLA class II compared to parent cells.
  • HLA human leukocyte antigen
  • Another aspect provides a method for producing hypoimmunogenic stem cells, comprising the step of genetically engineering stem cells to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II compared to parent cells.
  • HLA human leukocyte antigen
  • One aspect provides hypoimmunogenic stem cells genetically engineered to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II compared to parent cells.
  • HLA human leukocyte antigen
  • the above "parent cell” is a cell that has not been artificially manipulated to reduce the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II according to one aspect, and may be a cell that has been induced to have pluripotent differentiation ability through an artificial dedifferentiation process from a cell freshly isolated from a human body, a cell cultured therefrom, or a differentiated cell.
  • HLA Human Leukocyte Antigen
  • MHC Major Histocompatibility Antigen Complex
  • HLA classes are used to test the tissue compatibility between the donor and the recipient during cell, tissue, and organ transplantation. Transplantation is possible if the HLA antigens of the donor and recipient are all the same, but since there are more than 9719 types of alleles of HLA classes I and II, and each person has numerous genetic polymorphisms in each HLA gene, it is not easy to find a donor with a completely matching HLA type.
  • transplanting cells, tissues, or organs if the HLA-A genes of the donor and the HLA-A genes of the recipient do not match, the immune cells in the recipient's body recognize the difference and attack the donor cells, ultimately leading to transplant failure due to immune rejection.
  • one aspect provides a method for manufacturing transplant-compatible cells, specifically cells that are homozygous or null for immunocompatibility antigens through genetic manipulation.
  • Transplantability refers to the property of not causing an immune rejection response upon cell transplantation.
  • Cells, tissues, or organs can be made transplantable by reducing the expression of genes that cause immune rejection.
  • cells, tissues, or organs can be made transplantable by reducing the expression level of the HLA genes.
  • Transplantable cells may include cells that are homozygous for or deficient in immunocompatibility antigens.
  • HLA human leukocyte antigens
  • HLA-A, HLA-B, and HLA-C belong to HLA class I
  • HLA-DR, HLA-DP, and HLA-DQ belong to HLA class II.
  • Normal somatic cells express only three pairs of HLA class I, HLA-A, HLA-B, and HLA-C, while immune cells express a total of six pairs of HLA class I and HLA class II.
  • the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A, HLA-B, and HLA-C
  • the human leukocyte antigen class II may be at least one selected from the group consisting of HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1, and HLA-DPB.
  • the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A and HLA-B
  • the human leukocyte antigen class II may be at least one selected from the group consisting of HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, and HLA-DRB5
  • the human leukocyte antigen class I may be HLA-A and HLA-B
  • the human leukocyte antigen class II may be HLA-DRA.
  • the stem cells may be genetically engineered to have reduced expression of one or more proteins selected from the group consisting of HLA-A, HLA-B and HLA-DRA, specifically, the stem cells may be genetically engineered to have reduced expression of all of the HLA-A, HLA-B and HLA-DRA proteins.
  • HLA-A, HLA-B and HLA-DRA By manipulating the expression of HLA-A, HLA-B and HLA-DRA to be reduced, immune evasion can be obtained, while at the same time maintaining the expression of HLA-C, HLA-E, HLA-F and HLA-G, there is an effect of being able to defend against attacks by NK cells.
  • the stem cell may be genetically engineered with at least one region selected from the group consisting of exon 1 to exon 3 of a gene encoding HLA-A, exon 1 to exon 3 of a gene encoding HLA-B, and exon 1 to exon 3 of a gene encoding HLA-DRA, and specifically, the stem cell may be genetically engineered with at least one region selected from the group consisting of exon 2 of a gene encoding HLA-A, exon 3 of a gene encoding HLA-A, exon 2 of a gene encoding HLA-B, exon 3 of a gene encoding HLA-B, exon 2 of a gene encoding HLA-DRA, and exon 3 of a gene encoding HLA-DRA.
  • one or more regions selected from the group consisting of the exon 2 region of the gene encoding HLA-A of the stem cell, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be genetically engineered.
  • the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA of the stem cells may all be genetically engineered.
  • genetic engineering or “genetically engineered” refers to the act of introducing one or more genetic modifications into a cell or a cell produced thereby.
  • the genetic manipulation may be induced by a physical method to modify the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • the physical method may be, for example, X-ray irradiation, gamma ray irradiation, or the like.
  • the genetic manipulation may be induced by a chemical method to modify the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, or to change the expression of the gene.
  • the chemical method may be, for example, treatment with ethyl methanesulfonate, treatment with dimethyl sulfate, etc.
  • the genetic manipulation may be induced by a modification in the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II by a gene editing system.
  • the gene editing system may be, for example, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, etc.
  • the genetic manipulation may be induced by a change in gene expression by binding to mRNA transcribed from a gene encoding one or more proteins selected from the group consisting of HLA class I and HLA class II by an RNA interference (RNAi) system.
  • RNAi RNA interference
  • the stem cell may be genetically engineered through an RNA interference (RNAi) system, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, X-ray irradiation, gamma ray irradiation, ethyl methanesulfonate treatment, or dimethyl sulfate treatment, and specifically, may be engineered through a CRISPR/Cas system, and the genetic engineering may be induced by a modification in the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • RNAi RNA interference
  • a meganuclease system a zinc finger nuclease system
  • TALEN Transcription Activator-Like Effector Nuclease
  • Genetically engineered to have "reduced expression” or “inactivated” one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II means that one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II or the genes encoding the same exhibit expression or activity at a level lower than the level of expression or activity of one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II or the genes encoding the same measured in comparable stem cells of the same species or parent cells thereof, or that there is no expression or activity.
  • the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same may be reduced by about 20% or more, about 30% or more, about 40% or more, about 50% or more, about 55% or more, about 60% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more compared to the expression or activity of the original unmanipulated stem cells, or a 100% reduction, that is, one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same may not be expressed at all.
  • Genetically engineered stem cells in which the expression or activity of one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II is reduced can be identified using any method known in the art.
  • activation may mean that a gene is not expressed at all, or that a protein is produced that is not active even if it is expressed.
  • depression may mean that a gene encoding a specific protein is expressed at a lower level than in non-engineered cells, or that the protein expressed by the specific gene is expressed but its activity is low.
  • the decrease in expression or activity of one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II or the genes encoding them may be caused by mutation, replacement, deletion of part or all of the genes encoding one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II or by insertion of one or more bases into the genes, and may be caused by a genetic manipulation means encoding one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II.
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • a tracr trans-activating CRISPR sequence
  • tracrRNA or active portion tracrRNA e.g., tracrRNA or active portion tracrRNA
  • a tracr-mate sequence including a "direct repeat” and a tracrRNA-processing portion direct repeat in the context of an endogenous CRISPR system
  • a guide sequence also referred to as a "spacer” in the context of an endogenous CRISPR system
  • Cas CRISPR-associated gene or induce its activity.
  • one or more elements of the CRISPR system are derived from a type I, type II or type III CRISPR system.
  • one or more elements of a CRISPR system are derived from a particular organism comprising an endogenous CRISPR system, such as Streptococcus pyogenes .
  • a CRISPR system is characterized by an element (also referred to as a protospacer in the context of an endogenous CRISPR system) that promotes the formation of a CRISPR complex at the site of the target sequence.
  • a target sequence or “target gene” refers to a sequence designed to have complementarity with a guide sequence, wherein hybridization between the target sequence and the guide sequence promotes the formation of a CRISPR complex. While perfect complementarity is not required, sufficient complementarity exists to cause hybridization and promote the formation of a CRISPR complex.
  • the target sequence can comprise any polynucleotide, such as a DNA or RNA polynucleotide. In one embodiment, the target sequence is located within the nucleus or cytoplasm of a cell.
  • the above Cas protein forms an active endonuclease, or nickase, when it forms a complex with two RNAs called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA).
  • CRISPR RNA CRISPR RNA
  • tracrRNA trans-activating crRNA
  • Non-limiting examples of the above Cas proteins include Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, C
  • Modified versions may be included. These enzymes are known; for example, the amino acid sequence of the Streptococcus pyogenes Cas9 protein is available from the SwissProt database under accession number Q99ZW2. In one embodiment, the unmodified CRISPR enzyme, e.g., Cas9, has DNA cleavage activity.
  • the CRISPR enzyme may be a Cas9 protein
  • the Cas9 protein may be one or more Cas9 proteins selected from the group consisting of a Cas9 protein derived from Streptococcus pyogenes , a Cas9 protein derived from Campylobacter jejuni , a Cas9 protein derived from Streptococcus thermophiles, a Cas9 protein derived from Streptococcus aureus , and a Cas9 protein derived from Neisseria meningitidis , and specifically, a Cas9 protein derived from Streptococcus pyogenes .
  • the Cas9 protein is codon-optimized for expression in a eukaryotic cell, and when using the Streptococcus pyogenes derived Cas9 protein, the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be maximally reduced.
  • nuclear localization sequence or signal refers to an amino acid sequence that functions to transport a specific substance (e.g., a protein) into the cell nucleus, and generally functions to transport it into the cell nucleus through a nuclear pore.
  • a specific substance e.g., a protein
  • nuclear localization sequence is not required for the activity of the CRISPR complex in eukaryotes, it is believed that including such a sequence enhances the activity of the system, particularly targeting nucleic acid molecules in the nucleus.
  • RNA-guided CRISPR clustered regularly interspaced short palindrome repeats
  • CRISPR clustered regularly interspaced short palindrome repeats
  • Cas9 offers a breakthrough technology for knockout, transcriptional activation, and repression of target genes using a single guide RNA (sgRNA) (i.e., crRNA-tracrRNA fusion transcript), and this technology is known to target numerous genetic loci.
  • sgRNA single guide RNA
  • the Cas9 (or Cpf1) protein refers to an essential protein component in the CRISPR/Cas9 system, and information on the Cas9 (or Cpf1) gene and protein can be obtained from GenBank of the National Center for Biotechnology Information (NCBI).
  • CRISPR-associated genes encoding Cas (or Cpf1) proteins are known to exist in more than 40 different Cas (or Cpf1) protein families, and eight CRISPR subtypes (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, and Mtube) can be defined according to specific combinations of Cas genes and repeat structures. Therefore, each of the above CRISPR subtypes can form a repeat unit to form a polyribonucleotide-protein complex.
  • the above gene knockout may refer to the regulation of the activity of a gene, e.g., inactivation, by deletion, substitution, and/or insertion of one or more nucleotides in whole or in part of the gene.
  • the gene inactivation may refer to the suppression or downregulation of the expression of the gene, or the modification so that the gene encodes a protein that has lost its original function.
  • the gene regulation may refer to a change in the function of the gene due to the structural modification of the protein obtained by the deletion of the exon region by simultaneously targeting both intron regions surrounding one or more exons of the target gene, or the expression of the protein in a dominant negative form.
  • the target sequence used to knock out a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II in the stem cell may include, for example, a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, specifically, at least one selected from the group consisting of a gene encoding HLA-A, a gene encoding HLA-B, and a gene encoding HLA-DRA, and more specifically, at least one selected from the group consisting of an exon 2 region of a gene encoding HLA-A, an exon 3 region of a gene encoding HLA-B, an exon 3 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-DRA, and an exon 3 region of a gene encoding HLA-DRA.
  • it may include all of the exon 2 region of the gene encoding HLA-A, the exon 3 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA.
  • ICE analysis was performed to confirm the effectiveness of the designed gRNA sequences, and as a result, it was confirmed that G0002-HLA-A-g1 had an efficiency of 91%, G0002-HLA-B-g1 had an efficiency of 78%, and G0002-HLA-DRA-g2 had an efficiency of 86% (see Example 1).
  • the genetic manipulation that is artificially performed to reduce the expression or activity of one or more selected from the group consisting of the human leukocyte antigen class I and human leukocyte antigen class II or the gene encoding the same may be such that one or more selected from the group consisting of the human leukocyte antigen class I and human leukocyte antigen class II is not expressed in the form of a protein having its original function.
  • the manipulation of the gene may be induced by one or more of the following:
  • nucleotides e.g., 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide (each independently selected from A, T, C, and G) into any position of the target gene, and
  • the modified portion ('target region') of the gene encoding the HLA class I and/or HLA class II is at least 1 bp, at least 3 bp, at least 5 bp, at least 7 bp, at least 10 bp, at least 12 bp, at least 15 bp, at least 17 bp, at least 20 bp of the gene, for example, 1 bp to 30 bp, 3 bp to 30 bp, 5 bp to 30 bp, 7 bp to 30 bp, 10 bp to 30 bp, 12 bp to 30 bp, 15 bp to 30 bp, 17 bp to 30 bp, 20 bp to 30 bp, 1 bp to 27 bp, 3 bp to 27 bp, 5 bp to 27 bp, 7 bp to 27 bp, 10bp to 27bp, 12bp to 27bp, 15bp to 27bp, 17bp
  • one million induced pluripotent stem cells were mixed with the CRISPR/CAS9 system and electroporated to induce intracellular transfection. After approximately 28 days of culture, PCR and Sanger sequencing were performed to screen the collected clones, and genotyping was performed to ultimately select cells in which the HLA-A, HLA-B, and HLA-DRA genes were manipulated. Sequencing of the cells revealed that a 1 bp insertion occurred in the HLA-A region, a 28 bp deletion occurred in the HLA-B region, and a 1 bp deletion occurred in the HLA-DRA region (see Example 2).
  • chimeric RNA refers to a polynucleotide sequence comprising a guide sequence, a tracr sequence and/or a tracr mate sequence.
  • guide sequence refers to a sequence of about 20 bp within a guide RNA that directs a target site and may be used interchangeably with the term “guide” or “spacer”.
  • tracr mate sequence may also be used interchangeably with the term “direct repeat(s)”.
  • the guide RNA may be composed of two RNAs, namely a CRISPR RNA (crRNA) and a transactivating crRNA (tracrRNA), or may be a single-chain RNA (sgRNA) comprising portions of crRNA and tracrRNA and hybridizing to the target DNA.
  • crRNA CRISPR RNA
  • tracrRNA transactivating crRNA
  • sgRNA single-chain RNA
  • a guide sequence is any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and induce sequence-specific binding of a CRISPR complex to the target sequence.
  • any base sequence that can be used for genetic manipulation to reduce the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be used as a guide RNA without limitation.
  • the base sequence may be a sequence that can hybridize with a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • a portion of the guide RNA base sequence can be modified to modify/enhance the function of the guide RNA.
  • the degree of complementarity between the guide sequence and its corresponding target sequence is about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99% or more when optimally aligned using an appropriate alignment algorithm.
  • An optimal alignment can be determined by the use of any suitable algorithm for aligning sequences, non-limiting examples of which include the Smith-Waterman algorithm, the Needleman-Wunsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g., the Burrows Wheeler Aligner), ClustalW, Clustal X, BLAT, Novoalign (Novocraft Technologies), ELAND (Illumina, San Diego, CA), SOAP (available at soap.genomics.org.cn), and Maq (available at maq.sourceforge.net).
  • any suitable algorithm for aligning sequences include the Smith-Waterman algorithm, the Needleman-Wunsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g., the Burrows Wheeler Aligner), ClustalW, Clustal X, BLAT, Novoalign (Novocraft Technologies), ELAND (Illumina, San Diego, CA), SOAP (available at soap.genomics.org.cn), and Ma
  • the guide sequence comprises, for example, about 5, 10, 11, 12, 13, 14, 15,
  • the guide sequence is at least about 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75 nucleotides in length.
  • the guide sequence is no more than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12 nucleotides in length.
  • components of a CRISPR system sufficient to form a CRISPR complex comprising the tested guide sequence can be assessed by, for example, assessing preferential cleavage within the target sequence by, for example, a Surveyor assay as described herein, after transfection with a vector encoding components of the CRISPR sequence.
  • a host cell containing the corresponding target sequence can be provided.
  • cleavage of the target polynucleotide sequence can be assessed in vitro by providing components of a CRISPR complex comprising the target sequence, the test guide sequence, and a control guide sequence different from the test guide sequence, and comparing the binding or cleavage rates between the test and control guide sequence reactions at the target sequence.
  • Other assays are possible and will be readily available to those skilled in the art.
  • the guide sequence can be selected to target any target sequence.
  • the target sequence is a sequence within the genome of the cell.
  • Exemplary target sequences can include those that are unique within the target genome.
  • a unique target sequence within the genome can include a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNNNXGG, wherein NNNNNNNNNNXGG (N is A, G, T, or C; X can be anything) has a single occurrence within the genome.
  • a unique target sequence within the genome can include a Streptococcus pyogenes Cas9 target site of the form MMMMMMMNNNNNNNNNNNNNXGG, wherein NNNNNNNNNXGG (N is A, G, T, or C; X can be anything) has a single occurrence within the genome.
  • the unique target sequence within the genome can comprise a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNNNNNXXAGAAW, wherein NNNNNNNNNNXXAGAAW (N is A, G, T, or C; X can be anything; and W is A or T) has a single occurrence within the genome.
  • the unique target sequence within the genome can comprise a Streptococcus thermophilus CRISPR1 Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNNNXXAGAAW, wherein NNNNNNNNNXXAGAAW (N is A, G, T, or C; X can be anything; and W is A or T) has a single occurrence within the genome.
  • the unique target sequence within the genome can comprise a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNNNNNXGGXG, wherein NNNNNNNNNNNNXGGXG (N is A, G, T, or C; and X can be anything) has a single occurrence within the genome.
  • the unique target sequence within the genome can comprise a Streptococcus pyogenes Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNNNXGGXG, wherein NNNNNNNNNXGGXG (N is A, G, T, or C; and X can be anything) has a single occurrence within the genome.
  • N is A, G, T, or C; and X can be anything
  • M can be A, G, T, or C.
  • the CRISPR/Cas system may include one or more polynucleotides selected from the group consisting of a polynucleotide comprising a base sequence of SEQ ID NO: 1, a polynucleotide comprising a base sequence of SEQ ID NO: 2, and a polynucleotide comprising a base sequence of SEQ ID NO: 3.
  • the CRISPR/Cas system may include a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3.
  • the polynucleotide can complementarily bind to a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, and specifically, can bind to at least one site selected from a site consisting of exon 1 to exon 3 of the gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 1, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 1.
  • the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 2, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 2.
  • the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 3, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 3.
  • a base sequence comprising a polynucleotide consisting of the base sequence of SEQ ID NOs: 1 to 3 can serve as a gRNA of the CRISPR/CAS system and can target a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • one or more regions selected from regions consisting of exons 1 to 3 of the gene encoding HLA-A, exons 1 to 3 of the gene encoding HLA-B, and exons 1 to 3 of the gene encoding HLA-DRA may be targeted, and more specifically, one or more regions selected from regions consisting of exon 2 of the gene encoding HLA-A, exon 3 of the gene encoding HLA-A, exon 2 of the gene encoding HLA-B, exon 3 of the gene encoding HLA-B, exon 2 of the gene encoding HLA-DRA, and exon 3 of the gene encoding HLA-DRA may be targeted.
  • all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be targeted.
  • the exon 2 region of the gene encoding HLA-A may include a polynucleotide consisting of a base sequence of SEQ ID NO: 4, and the exon 2 region and exon 3 region of the gene encoding HLA-A may include a polynucleotide consisting of a base sequence of SEQ ID NO: 5.
  • the polynucleotide consisting of the base sequence of SEQ ID NO: 4 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 4, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 4.
  • the polynucleotide consisting of the base sequence of SEQ ID NO: 5 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 5, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 5.
  • the exon 2 region of the gene encoding HLA-B may include a polynucleotide consisting of a base sequence of SEQ ID NO: 6, and the exon 2 region and exon 3 region of the gene encoding HLA-B may include a polynucleotide consisting of a base sequence of SEQ ID NO: 7.
  • the polynucleotide consisting of the base sequence of SEQ ID NO: 6 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 6, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 6.
  • the polynucleotide comprising the base sequence of SEQ ID NO: 7 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 7, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 7.
  • the exon 2 region of the gene encoding the HLA-DRA may include a polynucleotide consisting of the base sequence of SEQ ID NO: 8
  • the exon 2 region and the exon 3 region of the gene encoding the HLA-DRA may include a polynucleotide consisting of the base sequence of SEQ ID NO: 9.
  • the polynucleotide comprising the base sequence of SEQ ID NO: 8 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 8, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 8.
  • the polynucleotide comprising the base sequence of SEQ ID NO: 9 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 9, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 9.
  • the CRISPR/Cas system may include a Cas9 (CRISPR associated protein 9) protein or a gene encoding a Cas9 protein and a NLS (Nuclear Localization Signal) protein or a gene encoding an NLS protein.
  • Cas9 CRISPR associated protein 9
  • NLS Nuclear Localization Signal
  • the "system” may include a complex of a nucleic acid and a Cas protein, wherein the nucleic acid includes at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3, and specifically, a complex of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a Cas protein. More specifically, the Cas protein may be a Cas9 protein.
  • preformed RNPs can be introduced into cells to initiate target genomic DNA editing.
  • This RNP-based delivery has the advantage of reducing off-target effects compared to vector delivery and enabling direct delivery of the Cas protein and gRNA to the nucleus of the cell.
  • RNPs ribonucleoproteins
  • Cas proteins and gRNAs can be synthesized and purified individually to produce RNPs.
  • Cas proteins and cleavage factor RNAs can be directly expressed via plasmids, and then the self-assembled RNPs can be purified.
  • the "system” may be a vector including a polynucleotide encoding at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein; and a vector including a polynucleotide encoding a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein.
  • the Cas protein may be a Cas9 protein.
  • a polynucleotide encoding at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein may be contained in the same vector, or may be contained in different vectors.
  • vector refers to a means for expressing a target gene in a host cell.
  • it may include a plasmid vector, a cosmid vector, a bacteriophage vector, an adenovirus vector, a retrovirus vector, and a viral vector such as an adeno-associated virus vector.
  • a vector that can be used as the above recombinant vector can be produced by manipulating a plasmid (e.g., V1k_GE, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX series, pET series, and pUC19, etc.), a phage, or a virus (e.g., SV40, etc.) that is frequently used in the art.
  • a plasmid e.g., V1k_GE, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX
  • a gene encoding a guide RNA that binds to a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, a gene encoding the Cas protein, and a gene encoding the NLS may be operably linked to a promoter.
  • operably linked refers to a functional linkage between a nucleotide expression regulatory sequence (e.g., a promoter sequence) and another nucleotide sequence.
  • the regulatory sequence can regulate transcription and/or translation of another nucleotide sequence by being “operably linked.”
  • the CRISPR/Cas system may be delivered to cells by electroporation.
  • the term "hypoimmunogenicity" above means that the cells, when transplanted into another organism, exhibit a reduced or eliminated immune rejection response compared to the wild type.
  • the immune rejection response may be reduced by at least 20%, at least about 30%, at least about 40%, at least about 50%, at least about 55%, at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%.
  • the immunogenicity of genetically engineered stem cells was analyzed, and it was confirmed that the expression levels of HLA-A, HLA-B, and HLA-DRA were significantly reduced compared to the parent cells both when IFN ⁇ stimulation was given and when it was not given, and it was also confirmed that central memory T cells (TCM) and effector memory T cells (TEM) did not proliferate, indicating that the genetically engineered stem cells do not exhibit immunogenicity (see Example 4).
  • TCM central memory T cells
  • TEM effector memory T cells
  • stem cell refers to an undifferentiated cell that has the ability to self-replicate and differentiate into two or more different types of cells.
  • the stem cell may be an autologous or allogeneic stem cell.
  • the stem cell may be an induced pluripotent stem cell, an embryonic stem cell, a somatic cell nuclear transfer embryonic stem cell, or an adult stem cell, and specifically, may be an induced pluripotent stem cell.
  • induced pluripotent stem cells refer to cells that are induced to have pluripotent differentiation capacity through an artificial dedifferentiation process from differentiated cells, and are also called induced stem cells.
  • the artificial dedifferentiation process may be performed by using viral or non-viral vectors such as retrovirus, lentivirus, and Sendai virus, or by introducing non-viral-mediated dedifferentiation factors such as proteins and cell extracts, or may include a dedifferentiation process using stem cell extracts, compounds, etc.
  • Induced pluripotent stem cells have almost the same characteristics as embryonic stem cells, specifically, they show a similar cell shape, have similar gene and protein expression patterns, have pluripotency in vitro and in vivo , form teratomas, form chimera mice when inserted into a mouse blastocyst, and are capable of germline transmission of genes.
  • Induced pluripotent stem cells include induced pluripotent stem cells derived from any species, including humans, monkeys, pigs, horses, cows, sheep, dogs, cats, mice, rats, or rabbits, but may specifically be induced pluripotent stem cells derived from humans.
  • the somatic cells before the above-mentioned induced pluripotent stem cells are dedifferentiated may be somatic cells derived from umbilical cord, umbilical cord blood, blood, bone marrow, fat, muscle, nerve, skin, amniotic membrane, amniotic fluid, or placenta.
  • the somatic cells may include peripheral blood mononuclear cells, fibroblasts, hepatocytes, adipocytes, epithelial cells, epidermal cells, chondrocytes, muscle cells, cardiac muscle cells, melanocytes, neural cells, glial cells, astroglial cells, monocytes, macrophages, and the like, and more specifically, may be induced pluripotent stem cells derived from peripheral blood mononuclear cells.
  • the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A and HLA-B, and may include both HLA-A and HLA-B.
  • the human leukocyte antigen class II may be HLA-DRA.
  • the stem cell can express at least one protein selected from the group consisting of OCT4, SSEA4, NANOG, SOX2, ESRRB, TRA-1-60, SOX17, BRACHYURY and PAX 6 at the same level as the parent cell, and specifically, can express at least one protein selected from the group consisting of OCT4, SSEA4, NANOG, TRA-1-60, SOX17, BRACHYURY and PAX 6 at the same level as the parent cell.
  • the degree of expression of pluripotency markers of genetically engineered stem cells was analyzed, and it was confirmed that the pluripotency markers OCT4, SSEA4, NANOG, and TRA-1-60 were expressed at a level similar to that of the parent cells that had not been genetically engineered.
  • the differentiation potential of genetically engineered stem cells into mesodermal lineage endothelial cells was confirmed, and it was confirmed that they differentiated into endothelial cells through mesoderm, similar to the parent cells, and expressed both endothelial cell markers CD31 and VE-Cadherin (Example 5).
  • Another aspect provides a method for producing hypoimmunogenic stem cells, comprising the step of genetically engineering stem cells to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II.
  • HLA human leukocyte antigen
  • human leukocyte antigen “stem cell” and “low immunogenicity” may be within the ranges described above.
  • the genetically manipulating step may be genetically manipulating one or more regions selected from the group consisting of exon 1 to exon 3 of a gene encoding HLA-A, exon 1 to exon 3 of a gene encoding HLA-B, and exon 1 to exon 3 of a gene encoding HLA-Dra of the stem cell, and specifically, may be genetically manipulating one or more regions selected from the group consisting of exon 2 of a gene encoding HLA-A, exon 3 of a gene encoding HLA-A, exon 2 of a gene encoding HLA-B, exon 3 of a gene encoding HLA-B, exon 2 of a gene encoding HLA-DRA, and exon 3 of a gene encoding HLA-DRA of the stem cell.
  • it may be a method of genetically manipulating one or more regions selected from the group consisting of the exon 2 region of the gene encoding HLA-A of the stem cell, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA.
  • the genetically manipulating step may be genetically manipulating all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA of the stem cells.
  • genetic engineering or “genetically engineered” refers to the act of introducing one or more genetic modifications into a cell or a cell produced thereby.
  • the genetic manipulation may be induced by a gene editing system to modify the base sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.
  • the gene editing system may be, for example, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, or the like.
  • the above Cas protein can form an active endonuclease or nickase when it forms a complex with two RNAs called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA).
  • CRISPR RNA CRISPR RNA
  • tracrRNA trans-activating crRNA
  • Non-limiting examples of the above Cas proteins include Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, Cs
  • the unmodified CRISPR enzyme e.g., Cas9
  • has DNA cleavage activity e.g., Cas9
  • the CRISPR enzyme may be a Cas9 protein
  • the Cas9 protein may be one or more Cas9 proteins selected from the group consisting of a Cas9 protein derived from Streptococcus pyogenes , a Cas9 protein derived from Campylobacter jejuni , a Cas9 protein derived from Streptococcus thermophiles, a Cas9 protein derived from Streptococcus aureus , and a Cas9 protein derived from Neisseria meningitidis , and specifically, a Cas9 protein derived from Streptococcus pyogenes .
  • the Cas9 protein is codon-optimized for expression in a eukaryotic cell, and when using the Streptococcus pyogenes derived Cas9 protein, the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be maximally reduced.
  • nuclear localization sequence or signal refers to an amino acid sequence that functions to transport a specific substance (e.g., a protein) into the cell nucleus, and generally functions to transport it into the cell nucleus through a nuclear pore.
  • a specific substance e.g., a protein
  • nuclear localization sequence is not required for the activity of the CRISPR complex in eukaryotes, it is believed that including such a sequence enhances the activity of the system, particularly targeting nucleic acid molecules in the nucleus.
  • RNA-guided CRISPR clustered regularly interspaced short palindrome repeats
  • CRISPR clustered regularly interspaced short palindrome repeats
  • Cas9 offers a breakthrough technology for knockout, transcriptional activation, and repression of target genes using a single guide RNA (sgRNA) (i.e., crRNA-tracrRNA fusion transcript), and this technology is known to target numerous genetic loci.
  • sgRNA single guide RNA
  • the Cas9 (or Cpf1) protein refers to an essential protein component in the CRISPR/Cas9 system, and information on the Cas9 (or Cpf1) gene and protein can be obtained from GenBank of the National Center for Biotechnology Information (NCBI).
  • CRISPR-associated genes encoding Cas (or Cpf1) proteins are known to exist in more than 40 different Cas (or Cpf1) protein families, and eight CRISPR subtypes (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, and Mtube) can be defined according to specific combinations of Cas genes and repeat structures. Therefore, each of the above CRISPR subtypes can form a repeat unit to form a polyribonucleotide-protein complex.
  • the genetic manipulation that is artificially performed so that the expression or activity of at least one gene encoding at least one selected from the group consisting of human leukocyte antibody class I and HLA human leukocyte antibody class II is reduced may be such that at least one selected from the group consisting of human leukocyte antibody class I and human leukocyte antibody class II is not expressed in a protein form having its original function.
  • the manipulation of the gene may be induced by at least one of the following:
  • nucleotides e.g., 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide (each independently selected from A, T, C, and G) into any position of the target gene, and
  • the gene editing system can be introduced into cells by electroporation, a gene gun, sonoporation, magnetofection, transient cell compression, or cell squeezing.
  • introduction may refer to inserting a nucleic acid and protein complex into a cell, or may refer to inserting a vector comprising a polynucleotide encoding a nucleic acid and a protein into a cell.
  • the genetically manipulating step may be performed by introducing into the stem cell a CRISPR/Cas system comprising at least one selected from the group consisting of a polynucleotide comprising a base sequence of SEQ ID NO: 1, a polynucleotide comprising a base sequence of SEQ ID NO: 2, and a polynucleotide comprising a base sequence of SEQ ID NO: 3.
  • a base sequence comprising a polynucleotide consisting of the base sequences of SEQ ID NOS: 1 to 3 can serve as a gRNA of the CRISPR/CAS system and can target a gene encoding one or more selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II.
  • HLA human leukocyte antigen
  • one or more regions selected from regions consisting of exons 1 to 3 of the gene encoding HLA-A, exons 1 to 3 of the gene encoding HLA-B, and exons 1 to 3 of the gene encoding HLA-DRA may be targeted, and more specifically, one or more regions selected from regions consisting of exon 2 of the gene encoding HLA-A, exon 3 of the gene encoding HLA-A, exon 2 of the gene encoding HLA-B, exon 3 of the gene encoding HLA-B, exon 2 of the gene encoding HLA-DRA, and exon 3 of the gene encoding HLA-DRA may be targeted.
  • all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be targeted.
  • low immunogenicity, universal induced pluripotent stem cells can be useful in developing cell therapy products using them as a source because they have low immunogenicity and do not cause immune rejection in the recipient's body when used for allogeneic transplantation for the purpose of treating diseases.
  • Figure 1 is a schematic diagram showing the overall method for producing iPSCs with triple HLA gene knockout and the results thereof.
  • Figure 2 is a diagram showing the results of evaluating the effectiveness of gRNA used for knockout and confirming that the gene was knocked out, and whether the HLA gene and protein were expressed when IFN ⁇ stimulation was applied to the manufactured iPSC.
  • Figure 3 is a diagram showing the results of an experiment that can confirm the morphology and karyotype of the manufactured iPSC and its pluripotency.
  • Figure 4 is a diagram showing the results of an experiment confirming the in vitro immunogenicity evaluation using PBMC or NK cells when IFN ⁇ stimulation was applied to manufactured iPSCs.
  • Figure 5 is a diagram showing the results of an experiment confirming the differentiation ability of manufactured iPSCs into endothelial cells and the expression of HLA proteins.
  • iPSCs induced pluripotent stem cells
  • PBMC Peripheral Blood Mononuclear Cell
  • HLA-A had HLA-A 11:01:01:01 and HLA-A 29:01:01:01
  • HLA-B had HLA-B 13:02:01:01 and HLA-B 58:01:01:01
  • HLA-C had HLA-C 03:02:02 and HLA-C 02:02:02
  • HLA-DRA had HLA-DRA 01:02:01 and HLA-DRA 01:01:02.
  • HLA-A and HLA-B which exhibit polymorphism in HLA class 1
  • CRISPR/CAS9 as the knockout region
  • HLA-DRA to eliminate HLA-DR
  • HLA-C which is known to be minor in polymorphism
  • CRISPR/CAS9 was used as a gene knockout system, and the PAM site (NGG) was designed to exist in the part excluding the heterogeneous region in the HLA-A gene and to allow 20 base pairs to function as biallelic genes.
  • NGG PAM site
  • HLA-A the g RNA, G0002-HLA-A-g1 (+, ACAGCGACGCCGCGAGCCAG, PAM: AGG), located within codons 37 to 43 in exon 2
  • HLA-B the g RNA, G0002-HLA-B-g1 (-, GCTGTCGAACCTCACGAACT, PAM: GGG), located within codons 31 to 38 in exon 2
  • HLA-DRA the g RNA, G0002-HLA-DRA-g2 (+, TGGCAAAGAAGGAGACGGTC, PAM: TGG), located within codons 36 to 42 in exon 2, were designed (
  • G0002-HLA-A-g1 showed an efficiency of 91%
  • G0002-HLA-B-g 1 which is a g RNA of HLA-B
  • G0002-HLA-DRA-g2 which is a g RNA of HLA-DRA
  • Figs. 2a to 2c showed an efficiency of 86%
  • iPSCs On Day 0, one million iPSCs were mixed with CRISPR/CAS9 Reagent (RNP complex) and electroporated. To determine the RNP complex conditions, two conditions of 40 ⁇ g each gRNA and 80 ⁇ g each gRNA were performed, and the degree of KO of each gene was analyzed using ICE analysis.
  • RNP complex CRISPR/CAS9 Reagent
  • transfected cells were collected and single cell cloning was performed through fax analysis. Approximately 10,000 cells were collected and EP (Electrostatic Potential) pool analysis was performed. From Day 5 to Day 11-13, the genotype of the transfected EP pool was analyzed through Next Generation Sequencing (NGS). Then, 48 single clones were collected by seeding in 96-well plates. From Days 11-13 to Days 25-28, each clone was screened and genotyped through PCR and Sanger sequencing. After this, clones with six triple HLA genes knocked out were collected, and cells with the final HLA-A, HLA-B, and HLA-DRA genes manipulated were finally selected through NGS analysis.
  • NGS Next Generation Sequencing
  • HLA-A, HLA-B, and HLA-DR were not expressed at the protein level, and HLA-C, which was not genetically modified, was not expressed either.
  • IFN ⁇ was stimulated in iPSCs for two days to activate HLA proteins, it was confirmed that the protein expression of HLA-A, HLA-B, and HLA-C increased in YiP3 before genetic modification, and HLA-A increased by 99.03%, HLA-B by 91.61%, HLA-C by 88.35%, and HLA-DR by 0.04%.
  • HLA-A was significantly reduced by 0.07%, HLA-B by 0.15%, and HLA-DR by 0.02%.
  • HLA-C which was not a genetically modified region, protein expression was confirmed to be not reduced by 97.34% (Fig. 2h). This indicates that HLA-A, HLA-B, and HLA-DRA were selectively knocked out as a result of genetic manipulation, and this can be confirmed at the protein level.
  • iPSCs Alkaline Phosphatase (AP) staining was performed to confirm morphology and differentiation potential.
  • iPSCs were seeded at low density on VTN-N-coated 6-well culture dishes and cultured for 5 days to form colonies.
  • the dishes were washed with 1 mL of 0.05% PBST and fixed with 1 mL of 4% PFA for 10 min at RT.
  • the cells were washed once with 1 mL of 0.05% PBST, and 1 mL of a staining solution containing Fast Red Violet, Naphthol AS-BI phosphate solution, and TDW (Sigma, SCR004) was added and incubated in the dark at room temperature for 30 min. Afterwards, the cells were washed once with 0.05% PBST and twice with 1 ⁇ DPBS.
  • RNA isolation was performed using TRIZOL reagent (Invitrogen, 15596026).
  • cDNA was synthesized from the isolated RNA using the RevertAid First Strand cDNA synthesis kit (ThermoFisher, K1622).
  • qRT-PCR QuantStudio 3 instrument (Applied Biosystems) and Power SYBR Green PCR Master Mix (Applied Biosystems, 4367659) .
  • the pluripotency markers OCT4, SOX2, KLF4, LIN28, and NANOG were expressed, while the endoderm differentiation marker SOX17, the mesoderm differentiation marker BRACHYUARY, and the ectoderm differentiation marker PAX6 were not expressed (Fig. 3c).
  • iPSCs differentiation was induced using the STEMdiff Trilineage Differentiation Kit (STEMCELL technology, 05230).
  • Cells were cultured at a density of 2.0 x 105 cells/well (ectoderm, endoderm) or 1.0 x 105 cells/well (mesoderm) in 24-well plates coated with 1 ug/mL iMatrix-511MG (Nippi, MX892012), and 10 ⁇ m Y-27632 was also added.
  • iMatrix-511MG Neppi, MX892012
  • 10 ⁇ m Y-27632 10 ⁇ m Y-27632 was also added.
  • For differentiation into ectoderm the medium was changed daily for 1 week, and for differentiation into mesoderm or endoderm, the medium was changed daily for 5 days.
  • immunostaining cells were fixed with 4% paraformaldehyde and immunofluorescence staining was performed.
  • T cells from PBMCs with different YiP3 and HLA types were isolated and co-cultured to determine the degree of proliferation of effector memory T cells (TEM) and central memory T cells (TCM).
  • TEM effector memory T cells
  • TCM central memory T cells
  • PBMCs from donors with different YiP3 and HLA types for each allele were selected, and PBMCs from donors with allele 1 of HLA-A 02:01, HLA-B 15:01, HLA-C 01:02, and HLA-DRB1 11:01 and allele 2 of HLA-A 02:07, HLA-B 46:01, HLA-C 04:01, and HLA-DRB1 15:02 were selected (Fig. 4a). Both YiP3 before genetic manipulation and iPSCs after manipulation were cultured after IFN ⁇ stimulation for two days before co-culture with PBMCs.
  • T cells activated by antigens presented by antigen-presenting cells in PBMCs were analyzed.
  • proliferation of CD4 + TCM (CD3 + CD4 + CD45RO + CD62L + ) and CD4 + TEM (CD3 + CD4 + CD45RO + CD62L - ) cells was evaluated by flow cytometry analysis.
  • An immunogenicity assay was performed to evaluate CD4 + T cell responses.
  • PBMCs were cultured in AIM-V medium (Gibco, 12055-083) for 1 day in a 5% CO2 incubator at 37 °C.
  • a CD4 + T cell isolation kit (Miltenyi, 130-096-533) was used to sort CD4 + T cells.
  • 0.5 ⁇ m CFSE (Invitrogen, C34554) was added to the collected CD4 + T cells, and they were incubated for 10 minutes in a darkened 37 °C incubator, and washed once with 1 X DPBS.
  • CD3 microbeads (Miltenyi, 130-050-101) were used to obtain CD3 - /CD4 - cells from CD4 - PBMCs.
  • the obtained CD3 - /CD4 - cells were added with 10 ⁇ g/mL Mitomycin C and incubated for 20 minutes, then washed with AIM - V medium.
  • CD3 - /CD4 - PBMCs (3.0 x 10 5 cells) and CFSE-labeled CD4 + T cells (4 x 10 5 cells) were co-cultured in 24-well plates.
  • 3 x 10 3 iPSCs of each type were added to each well and cultured in an incubator at 37 °C for 1 week after treatment with 1 mg/mL IL-2.
  • the proliferation of CFSE-labeled CD4 + T cells was analyzed by flow cytometry.
  • co-culture was performed with CD3 - /CD4 - PBMCs (3.0 x 10 5 cells) and iPSCs (3.0 x 10 3 cells) for 7 days, and analyzed weekly for a total of 3 weeks to assess long-term immunogenicity.
  • CD107a a NK cell activation marker
  • NK cells were isolated from peripheral blood mononuclear cells (Fig. 4e) of different donors with different HLA types (HLA-A 30:04, HLA-A 02:03, HLA-B 38:02, HLA-B 14:01, HLA-DRB1 15:02, HLA-DRB1 07:01), stimulated with IL-2, and co-cultured with genetically engineered iPSCs.
  • HLA-A 30:04, HLA-A 02:03, HLA-B 38:02, HLA-B 14:01, HLA-DRB1 15:02, HLA-DRB1 07:01 HLA-DRB1
  • IL-2 co-cultured with genetically engineered iPSCs.
  • CD107a an activation marker of NK cells
  • NK cells were significantly reduced in genetically modified iPSCs compared to non-genetically modified stem cells (p ⁇ 0.01, compared to non-genetically modified stem cells) (Fig. 4g).
  • 1 X protease cocktail (Invitrogen, 78430) was added to RIPA lysis buffer (Invitrogen, 89901) at 5.0 x 10 6 cells per mL and incubated for 10 minutes in a refrigerated state. After centrifugation at 13,200 x g for 20 minutes, the lysate was collected and the protein concentration was measured using a BCA assay kit (Invitrogen, 23227).
  • HLA-A Primary antibodies HLA-A (Abcam, ab52922, 1:5,000), HLA-B (Abcam, ab193415, 1:1,000), HLA-C (Abcam, ab126722, 1:1,000), HLA-DR (Abcam, ab92511, 1:1,000), HLA-DRA (Proteintech, PTG-17221-1-AP, 1:1:000), ⁇ -Actin (SantaCruz, sc-47778 HRP, 1:1,000) were diluted in 5% skim milk, and the membranes were incubated overnight at 4°C. The membrane was washed three times with 1X PBS-T buffer on an orbital shaker and then incubated with secondary antibody diluted 1:10,000 for 1 hour. The membrane was then washed three times and detected using Advansta ECL reagent (K-12045) on an iBright 1500 instrument (Invitrogen).
  • HLA-DRA protein could not be confirmed in differentiated EC cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Reproductive Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Transplantation (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention relates to hypoimmunogenic universal induced pluripotent stem cells and a method for producing same The induced pluripotent stem cells according to one aspect have reduced immunogenicity in allogeneic transplantation for the purpose of disease treatment and do not cause immune rejection in the recipient's body, and thus may be utilized as a source for developing cell therapeutic agents.

Description

저면역원성의 범용성 유도만능줄기세포Low-immunogenicity, universal induced pluripotent stem cells

본 발명은 저면역원성의 범용성 유도만능줄기세포에 관한 것이다.The present invention relates to a universal induced pluripotent stem cell with low immunogenicity.

세포치료제에서 면역원성은 동종 이식 시 이식의 성공 여부를 결정짓는 중요한 요소라 볼 수 있다. 세포치료제로서 줄기세포는 성체 줄기세포, MSC, ESC, iPSC를 이용한 연구가 진행되어 오고 있다. 이중 iPSC는 체세포에 야마나카 factor를 도입하여 다형성(pluripotent)을 가지고 있으며 무한 증식이 가능한 세포로서 심장, 신경, 연골, 뼈와 같은 타겟 세포로 분화가 가능한 세포이다. 이를 이용하여 세포 치료제로 이용하고자 연구가 진행되어 오고 있으나, 동종 요법에는 면역원성이라는 한계를 지니고 있다. 본 발명에서는 세포치료제의 원천 세포로서 iPSC를 응용하기 위해 유전자 조작을 통해 면역원성을 극복한 저 면역원성 범용 유도만능줄기세포(hypoimmunogenic Universal iPSC)를 만들고자 하였다.In cell therapy, immunogenicity is a critical factor in determining the success of allogeneic transplantation. Research on stem cells as cell therapy has been conducted using adult stem cells, MSCs, ESCs, and iPSCs. Among these, iPSCs are somatic cells that have been engineered with Yamanaka factors to achieve pluripotency and infinite proliferation, enabling differentiation into target cells such as heart, nerve, cartilage, and bone. Research has been conducted to utilize iPSCs in cell therapy, but their use in allogeneic therapy is limited by immunogenicity. In the present invention, we aimed to create hypoimmunogenic universal iPSCs (iPSCs) by genetically modifying iPSCs to overcome immunogenicity and utilize them as a source cell for cell therapy.

주조직적합복합체(major histocompatibility complex, MHC) 즉 HLA(Human Leukocyte Antigen)이라고 알려진 유전자는 면역계에서 작용하는 'MHC 분자(MHC molecule)'를 암호화하는 유전자 자리이며 염색체 6번에 위치하고 있다. HLA class I (A, B, and C)과 the class II (DR, DQ, DP) 분자로 이루어져 있고 인간 유전자 중에 가장 다형성을 많이 가지고 있는 부분이다. Class I MHC인 HLA-A, B, C의 경우 자기와 다른 단백질이 존재하면 세포 독성 T 세포에 제시하는 역할을 하고 다형성을 가지는 α chain 과 다형성을 갖지 않는 Beta 2M으로 구성되어 있다. Class Ⅱ MHC인 HLA-DR, DQ, DP의 경우 다형성을 가지는 beta 1 과 다형성을 갖지 않는 alpha chain, Beta2을 가지고 있고 자기와 다른 단백질이 존재하면 보조 T 세포에 제시하는 역할을 한다.The major histocompatibility complex (MHC), also known as HLA (Human Leukocyte Antigen), encodes the 'MHC molecule' that functions in the immune system and is located on chromosome 6. It is composed of HLA class I (A, B, and C) and the class II (DR, DQ, DP) molecules and is the part with the most polymorphism among human genes. Class I MHC, HLA-A, B, C, has the role of presenting foreign proteins to cytotoxic T cells and is composed of a polymorphic α chain and a non-polymorphic Beta 2M. Class II MHC, HLA-DR, DQ, DP, has a polymorphic beta 1 and a non-polymorphic alpha chain and Beta 2, and has the role of presenting foreign proteins to helper T cells.

인간 백혈구 항원(HLA)은 이식을 고려할 때 가장 중요한 유전자이다. 해당 유전자는 HLA 클래스 I (A, B, C)와 클래스 II (DR, DQ, DP)로 구성되며, 인간 유전자 중에서 가장 다형성이 높은 부분이다. HLA 단백질의 다형성은 면역 보호에 필수적이나, 이로 말미암아 유전적 배경이 다른 장기를 이식할 시 면역반응에 의해 실패하는 경우가 종종 발생한다. 이는 세포치료제를 고려할 때도 똑같이 적용이 된다. 기증자로부터 받은 세포가 HLA 타입이 틀려 비자기로 구분될 때 CD4 T cell과 CD8 T cell, NK cell의 공격을 받을 수 있고 이는 곧 면역반응으로 이어지게 되어 있다.Human leukocyte antigens (HLA) are the most important genes when considering transplantation. These genes consist of HLA class I (A, B, C) and class II (DR, DQ, DP), and are the most polymorphic part of the human genome. HLA protein polymorphism is essential for immune protection, but it often leads to failure due to immune responses when transplanting organs from different genetic backgrounds. This same principle applies to cell therapy. When donor cells are classified as non-self due to mismatched HLA types, they can be attacked by CD4 T cells, CD8 T cells, and NK cells, which in turn triggers an immune response.

일 양상은 모세포에 비하여 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 HLA 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 유전적으로 조작된 저면역원성 줄기세포를 제공하는 것이다.One aspect is to provide hypoimmunogenic stem cells genetically engineered to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and HLA class II compared to parent cells.

다른 양상은 모세포에 비하여 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 줄기세포를 유전적으로 조작하는 단계를 포함하는 저면역원성 줄기세포의 제조 방법을 제공하는 것이다.Another aspect provides a method for producing hypoimmunogenic stem cells, comprising the step of genetically engineering stem cells to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II compared to parent cells.

일 양상은 모세포에 비하여 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 유전적으로 조작된 저면역원성 줄기세포를 제공한다.One aspect provides hypoimmunogenic stem cells genetically engineered to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II compared to parent cells.

상기 "모세포"는 일 양상에 따른 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II으로 이루어진 군에서 선택되는 하나 이상의 발현 또는 활성이 감소되도록 인위적인 조작을 수행하지 않은 세포로서, 인체에서 갓 분리된 세포, 이를 배양한 세포 또는 분화된 세포들로부터 인위적인 역분화 과정을 통해 다능성 분화능을 가지도록 유도된 세포일 수 있다.The above "parent cell" is a cell that has not been artificially manipulated to reduce the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II according to one aspect, and may be a cell that has been induced to have pluripotent differentiation ability through an artificial dedifferentiation process from a cell freshly isolated from a human body, a cell cultured therefrom, or a differentiated cell.

상기 "인간 백혈구 항원(Human Leukocyte Antigen, HLA)"은 조직적합항원(histocompatibility antigen)과 동의어이며, 주요면역적합항원(Major Histocompatibility Antigen Complex, MHC)라고도 불리는 세포 표면 단백질이다. 사람에서는 부계 유전자에서 유래한 6 종과 모계 유전자에서 유래한 6 종, 즉 총 6 쌍이 발현되고 있는 것으로 알려져 있다. HLA의 역할은 세포 내에 존재하는 단백질들의 조각들을 세포의 표면에 전시(display)하여, 생체 내에서 발생했을 지 모를 감염이나 돌연변이가 면역세포에 의해 감지되도록 하는 것이며, 이런 이유로 항원제공 단백질(antigen presenting protein: APP) 이라고도 한다. 이들 항원제공단백질들은 자가세포(autologous cell)가 아닌 경우 이식 수여자 몸 안에 존재하는 면역세포들의 주요 공격대상이 된다. 일반적으로 세포, 조직, 기관 이식 시 공여자와 수여자 간의 조직적합성 검사에 HLA 클래스를 이용하게 된다. 공여자와 수여자의 HLA 항원이 전부 일치하면 이식이 가능하나, HLA 클래스 I 및 II의 대립유전자는 총 9719가지 이상으로, 사람들마다 각 HLA 유전자들 상에 수많은 유전적 다형성 (polymorphism)을 가지고 있기 때문에 HLA 타입이 완전히 일치하는 공여자를 찾는 것이 쉽지 않다. 세포, 조직, 기관 이식 시 공여자의 HLA-A 유전자와 수여자의 HLA-A 유전자가 일치하지 않는 경우 수여자의 체내 면역세포들이 그 차이를 인식하여 공여자 세포를 공격하게 되고, 결국 면역거부 반응에 의한 이식의 실패로 이어지게 된다. 이와 관련하여 일 양상은 이식적합성 세포, 구체적으로는 유전자 조작을 통하여 면역적합항원이 동형접합성(homozygous)이거나 또는 결손(null)된 세포를 제조할 수 있는 방법을 제공한다.The above "Human Leukocyte Antigen (HLA)" is synonymous with histocompatibility antigen and is a cell surface protein also called Major Histocompatibility Antigen Complex (MHC). In humans, it is known that six types are expressed, six types derived from paternal genes and six types derived from maternal genes, or a total of six pairs. The role of HLA is to display fragments of proteins existing within the cell on the surface of the cell so that infections or mutations that may have occurred in the body can be detected by immune cells. For this reason, it is also called antigen presenting protein (APP). These antigen presenting proteins become the main target of attack by immune cells existing in the body of the transplant recipient if they are not autologous cells. Generally, HLA classes are used to test the tissue compatibility between the donor and the recipient during cell, tissue, and organ transplantation. Transplantation is possible if the HLA antigens of the donor and recipient are all the same, but since there are more than 9719 types of alleles of HLA classes I and II, and each person has numerous genetic polymorphisms in each HLA gene, it is not easy to find a donor with a completely matching HLA type. When transplanting cells, tissues, or organs, if the HLA-A genes of the donor and the HLA-A genes of the recipient do not match, the immune cells in the recipient's body recognize the difference and attack the donor cells, ultimately leading to transplant failure due to immune rejection. In this regard, one aspect provides a method for manufacturing transplant-compatible cells, specifically cells that are homozygous or null for immunocompatibility antigens through genetic manipulation.

상기 용어 "이식적합성"은 세포 이식 시 면역거부 반응을 일으키지 않는 성질을 의미한다. 면역거부반응의 원인인 유전자의 발현을 감소시킴으로써 세포, 조직, 또는 기관이 이식적합성을 갖도록 할 수 있으며, 이에 대한 비제한적 예시로, 상기 HLA 유전자의 발현 수준을 감소시킴으로써 세포, 조직 또는 기관이 이식적합성을 갖도록 할 수 있다. 이식적합성 세포는 면역적합항원이 동형접합성(homozygous)이거나, 결손(null)된 세포를 포함할 수 있다.The term "transplantability" above refers to the property of not causing an immune rejection response upon cell transplantation. Cells, tissues, or organs can be made transplantable by reducing the expression of genes that cause immune rejection. As a non-limiting example, cells, tissues, or organs can be made transplantable by reducing the expression level of the HLA genes. Transplantable cells may include cells that are homozygous for or deficient in immunocompatibility antigens.

상기 인간 백혈구 항원(Humen Leukocyte Antigen, HLA)은 "HLA 클래스 I" 및 "HLA 클래스 II"로 분류할 수 있다. HLA-A, HLA-B, HLA-C가 HLA 클래스 I에 속하며, HLA-DR, HLA-DP, HLA-DQ는 HLA 클래스 II에 속한다. 일반 체세포는 HLA 클래스 I에 속하는 HLA-A, HLA-B, HLA-C, 총 3 쌍 만을 발현하며, 면역세포들은 HLA 클래스 I과 HLA 클래스 II, 모두 합하여 총 6 쌍을 발현한다.The above human leukocyte antigens (HLA) can be classified into "HLA class I" and "HLA class II". HLA-A, HLA-B, and HLA-C belong to HLA class I, and HLA-DR, HLA-DP, and HLA-DQ belong to HLA class II. Normal somatic cells express only three pairs of HLA class I, HLA-A, HLA-B, and HLA-C, while immune cells express a total of six pairs of HLA class I and HLA class II.

일 구체예예서, 상기 인간 백혈구 항원 클래스 I은 HLA-A, HLA-B 및 HLA-C로 이루어진 군에서 선택되는 하나 이상일 수 있고, 상기 인간 백혈구 항원 클래스 II는 HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1 및 HLA-DPB으로 이루어진 군에서 선택되는 하나 이상일 수 있다. 구체적으로는, 상기 인간 백혈구 항원 클래스 I은 HLA-A 및 HLA-B로 이루어진 군에서 선택되는 하나 이상일 수 있고, 인간 백혈구 항원 클래스 II는 HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4 및 HLA-DRB5로 이루어진 군에서 선택되는 하나 이상일 수 있고, 보다 구체적으로는, 인간 백혈구 항원 클래스 I은 HLA-A 및 HLA-B 일 수 있고, 인간 백혈구 항원 클래스 II는 HLA-DRA 일 수 있다.In one specific example, the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A, HLA-B, and HLA-C, and the human leukocyte antigen class II may be at least one selected from the group consisting of HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, HLA-DRB5, HLA-DQA1, HLA-DQB1, HLA-DPA1, and HLA-DPB. Specifically, the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A and HLA-B, and the human leukocyte antigen class II may be at least one selected from the group consisting of HLA-DRA, HLA-DRB1, HLA-DRB3, HLA-DRB4, and HLA-DRB5, and more specifically, the human leukocyte antigen class I may be HLA-A and HLA-B, and the human leukocyte antigen class II may be HLA-DRA.

일 구체예에서, 상기 줄기세포는 상기 HLA-A, HLA-B 및 HLA-DRA로 이루어진 군에서 선택되는 하나 이상의 단백질의 발현이 감소되도록 유전적으로 조작된 것 일 수 있고, 구체적으로는 상기 HLA-A, HLA-B 및 HLA-DRA 단백질이 모두 발현이 감소되도록 유전적으로 조작된 것일 수 있다.In one specific example, the stem cells may be genetically engineered to have reduced expression of one or more proteins selected from the group consisting of HLA-A, HLA-B and HLA-DRA, specifically, the stem cells may be genetically engineered to have reduced expression of all of the HLA-A, HLA-B and HLA-DRA proteins.

상기 HLA-A, HLA-B 및 HLA-DRA의 발현이 감소되도록 조작하여 면역 회피성을 얻을 수 있으며, 동시에 HLA-C, HLA-E, HLA-F 및 HLA-G의 발현은 유지하여 NK 세포의 공격을 방어할 수 있는 효과가 있다.By manipulating the expression of HLA-A, HLA-B and HLA-DRA to be reduced, immune evasion can be obtained, while at the same time maintaining the expression of HLA-C, HLA-E, HLA-F and HLA-G, there is an effect of being able to defend against attacks by NK cells.

일 구체예에서, 상기 줄기세포는 HLA-A를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위, HLA-B를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위가 유전적으로 조작된 것일 수 있고, 구체적으로는, 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-A를 코딩하는 유전자의 엑손 3 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 3 부위, HLA-DRA를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위가 유전적으로 조작된 것일 수 있다. 보다 구체적으로는, 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 2 부위로 이루어진 군에서 선택되는 하나 이상의 부위가 유전적으로 조작된 것일 수 있다.In one specific example, the stem cell may be genetically engineered with at least one region selected from the group consisting of exon 1 to exon 3 of a gene encoding HLA-A, exon 1 to exon 3 of a gene encoding HLA-B, and exon 1 to exon 3 of a gene encoding HLA-DRA, and specifically, the stem cell may be genetically engineered with at least one region selected from the group consisting of exon 2 of a gene encoding HLA-A, exon 3 of a gene encoding HLA-A, exon 2 of a gene encoding HLA-B, exon 3 of a gene encoding HLA-B, exon 2 of a gene encoding HLA-DRA, and exon 3 of a gene encoding HLA-DRA. More specifically, one or more regions selected from the group consisting of the exon 2 region of the gene encoding HLA-A of the stem cell, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be genetically engineered.

일 구체예에서, 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 2 부위가 모두 유전적으로 조작된 것일 수 있다.In one specific example, the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA of the stem cells may all be genetically engineered.

상기 용어 "유전적 조작(genetic engineering)" 또는 "유전적으로 조작된(genetically engineered)"은 세포에 대하여 하나 이상의 유전적 변형(genetic modification)을 도입하는 행위 또는 그에 의하여 만들어진 세포를 나타낸다.The above term "genetic engineering" or "genetically engineered" refers to the act of introducing one or more genetic modifications into a cell or a cell produced thereby.

상기 유전적 조작은 물리적인 방법에 의해 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 핵산 서열 내 변형으로 유도되는 것일 수 있다. 상기 물리적인 방법은 예를 들어, X-선 조사, 감마선 조사 등일 수 있다.The genetic manipulation may be induced by a physical method to modify the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II. The physical method may be, for example, X-ray irradiation, gamma ray irradiation, or the like.

또한, 상기 유전적 조작은 화학적인 방법에 의해 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 핵산서열 내 변형 또는 유전자의 발현의 변화로 유도되는 것일 수 있다. 상기 화학적인 방법은 예를 들어, 에틸 메탄설포네이트(ethyl methanesulfonate) 처리, 다이메틸 설페이트(dimethyl sulfate) 처리 등일 수 있다.Additionally, the genetic manipulation may be induced by a chemical method to modify the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, or to change the expression of the gene. The chemical method may be, for example, treatment with ethyl methanesulfonate, treatment with dimethyl sulfate, etc.

또한, 상기 유전적 조작은 유전자 편집 시스템에 의해 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 핵산서열 내 변형으로 유도되는 것일 수 있다. 상기 유전자 편집 시스템은 예를 들어, 메가뉴클레아제(Meganuclease) 시스템, 징크핑거 뉴클레아제(Zinc finger nuclease) 시스템, 탈렌(TALEN; Transcription Activator-Like Effector Nuclease) 시스템, CRISPR/Cas 시스템 등일 수 있다.Additionally, the genetic manipulation may be induced by a modification in the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II by a gene editing system. The gene editing system may be, for example, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, etc.

또한, 상기 유전적 조작은 RNA 간섭(RNAi; RNA interference) 시스템에 의해 HLA 클래스 I 및 HLA 클래스 II로 이루어진 군에서 선택되는 하나 이상의 단백질을 암호화하는 유전자로부터 전사된 mRNA에 결합하여 유전자 발현의 변화로 유도되는 것일 수 있다.Additionally, the genetic manipulation may be induced by a change in gene expression by binding to mRNA transcribed from a gene encoding one or more proteins selected from the group consisting of HLA class I and HLA class II by an RNA interference (RNAi) system.

일 구체예에서, 상기 줄기세포는 RNA 간섭(RNAi; RNA interference) 시스템, 메가뉴클레아제(Meganuclease) 시스템, 징크핑거 뉴클레아제(Zinc finger nuclease) 시스템, 탈렌(TALEN; Transcription Activator-Like Effector Nuclease) 시스템, CRISPR/Cas 시스템, X-선 조사, 감마선 조사, 에틸 메탄설포네이트(ethyl methanesulfonate) 처리 또는 다이메틸 설페이트(dimethyl sulfate) 처리를 통해 유전적으로 조작된 것일 수 있고, 구체적으로는 CRISPR/Cas 시스템에 통해 조작된 것일 수 있고, 상기 유전적으로 조작은 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 핵산서열 내 변형으로 유도되는 것일 수 있다.In one specific example, the stem cell may be genetically engineered through an RNA interference (RNAi) system, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, X-ray irradiation, gamma ray irradiation, ethyl methanesulfonate treatment, or dimethyl sulfate treatment, and specifically, may be engineered through a CRISPR/Cas system, and the genetic engineering may be induced by a modification in the nucleic acid sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.

상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 "발현이 감소" 또는 이를 암호화하는 유전자가 "불활성화" 되도록 유전적으로 조작된 것은, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자가 비교 가능한 동일 종의 줄기세포 또는 그의 모세포에서 측정된 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성 수준보다 낮은 정도의 발현 또는 활성을 나타내거나 발현 또는 활성이 없는 것을 의미한다. 즉 상기 줄기세포에 있어서 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 본래 조작되지 않은 줄기세포의 발현 또는 활성보다 약 20% 이상, 약 30% 이상, 약 40% 이상, 약 50% 이상, 약 55% 이상, 약 60% 이상, 약 70% 이상, 약 75% 이상, 약 80% 이상, 약 85% 이상, 약 90% 이상, 91% 이상, 92% 이상, 93% 이상, 94% 이상, 95% 이상, 96% 이상, 97% 이상, 98% 이상 또는 99% 이상 감소된 것일 수 있고, 또는 100% 감소 즉, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 또는 이를 암호화하는 유전자가 전혀 발현되지 않는 것일 수 있다.Genetically engineered to have "reduced expression" or "inactivated" one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II means that one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II or the genes encoding the same exhibit expression or activity at a level lower than the level of expression or activity of one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II or the genes encoding the same measured in comparable stem cells of the same species or parent cells thereof, or that there is no expression or activity. That is, in the stem cells, the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same may be reduced by about 20% or more, about 30% or more, about 40% or more, about 50% or more, about 55% or more, about 60% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, about 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more compared to the expression or activity of the original unmanipulated stem cells, or a 100% reduction, that is, one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same may not be expressed at all.

상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 감소된 유전적으로 조작된 줄기세포는 당업계에 공지된 임의의 방법을 사용하여 확인될 수 있다. 상기 용어 "불활성화 (inactivation)"는 전혀 발현이 되지 않는 유전자 또는 발현이 되더라도 그 활성이 없는 단백질이 생성되는 것을 의미할 수 있다. 상기 용어 "감소 (depression)"는 특정 단백질을 암호화하는 유전자가 조작되지 않은 세포에 비하여 낮은 수준으로 발현되거나, 또는 특정 유전자가 발현하는 단백질이 발현이 되더라도 그 활성이 낮은 것을 의미할 수 있다. Genetically engineered stem cells in which the expression or activity of one or more genes encoding the human leukocyte antigen class I and human leukocyte antigen class II is reduced can be identified using any method known in the art. The term "inactivation" may mean that a gene is not expressed at all, or that a protein is produced that is not active even if it is expressed. The term "depression" may mean that a gene encoding a specific protein is expressed at a lower level than in non-engineered cells, or that the protein expressed by the specific gene is expressed but its activity is low.

상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 감소되는 것은 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 일부 또는 전부가 변이, 치환, 삭제되거나 상기 유전자에 하나 이상의 염기가 삽입되는 것일 수 있으며, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자 조작 수단에 의할 수 있다.The decrease in expression or activity of one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II or the genes encoding them may be caused by mutation, replacement, deletion of part or all of the genes encoding one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II or by insertion of one or more bases into the genes, and may be caused by a genetic manipulation means encoding one or more genes selected from the group consisting of the human leukocyte antigen class I and the human leukocyte antigen class II.

일반적으로, 널리 알려진 유전자 조작 수단인 "CRISPR(Clustered Regularly Interspaced Short Palindromic Repeats) 시스템"은 집합적으로 Cas 유전자를 코딩하는 서열, tracr(트랜스-활성화 CRISPR) 서열(예를 들어, tracrRNA 또는 활성 부분 tracrRNA), tracr-메이트 서열(내인성 CRISPR 시스템의 맥락에서 "직접 반복부" 및 tracrRNA-가공 부분 직접 반복부 포함), 가이드 서열(내인성 CRISPR 시스템의 맥락에서 "스페이서"로도 지칭), 가이드 RNA 또는 CRISPR 유전자좌로부터의 기타 서열 및 전사물을 포함하는 CRISPR-관련(CRISPR-associated; 이하 Cas) 유전자의 발현에 수반되거나, 그의 활성을 유도하는 전사물 및 다른 요소를 지칭한다. 일 구체예에서, CRISPR 시스템의 하나 이상의 요소는 I형, II형 또는 III형 CRISPR 시스템으로부터 유래된다. 일 구체예에서, CRISPR 시스템의 하나 이상의 요소는 내인성 CRISPR 시스템을 포함하는 특정 유기체, 예를 들어, 스트렙토코커스 피요게네스(Streptococcus pyogenes)로부터 유래된다. 일반적으로, CRISPR 시스템은 표적 서열의 부위에서 CRISPR 복합체의 형성을 증진시키는 요소(내인성 CRISPR 시스템의 맥락에서 프로토스페이서로도 지칭)를 특징으로 한다. CRISPR 복합체의 형성의 맥락에서, "표적 서열" 또는 "표적 유전자"는 가이드 서열이 상보성을 갖도록 설계된 서열을 지칭하며, 여기서, 표적 서열과 가이드 서열 간의 혼성화는 CRISPR 복합체의 형성을 증진시킨다. 본질적으로 완전한 상보성이 필요하지 않지만, 혼성화를 야기하고, CRISPR 복합체의 형성을 증진시키는 충분한 상보성이 존재한다. 표적 서열은 임의의 폴리뉴클레오티드, 예를 들어, DNA 또는 RNA 폴리뉴클레오티드를 포함할 수 있다. 일 구체예에서, 표적 서열은 세포의 핵 또는 세포질 내에 위치한다. In general, the "CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) system", a widely known genetic engineering tool, collectively refers to a sequence encoding a Cas gene, a tracr (trans-activating CRISPR) sequence (e.g., tracrRNA or active portion tracrRNA), a tracr-mate sequence (including a "direct repeat" and a tracrRNA-processing portion direct repeat in the context of an endogenous CRISPR system), a guide sequence (also referred to as a "spacer" in the context of an endogenous CRISPR system), a guide RNA or other sequences and transcripts from a CRISPR locus, and other elements that accompany the expression of a CRISPR-associated (hereinafter referred to as Cas) gene or induce its activity. In one embodiment, one or more elements of the CRISPR system are derived from a type I, type II or type III CRISPR system. In one embodiment, one or more elements of a CRISPR system are derived from a particular organism comprising an endogenous CRISPR system, such as Streptococcus pyogenes . Typically, a CRISPR system is characterized by an element (also referred to as a protospacer in the context of an endogenous CRISPR system) that promotes the formation of a CRISPR complex at the site of the target sequence. In the context of the formation of a CRISPR complex, a "target sequence" or "target gene" refers to a sequence designed to have complementarity with a guide sequence, wherein hybridization between the target sequence and the guide sequence promotes the formation of a CRISPR complex. While perfect complementarity is not required, sufficient complementarity exists to cause hybridization and promote the formation of a CRISPR complex. The target sequence can comprise any polynucleotide, such as a DNA or RNA polynucleotide. In one embodiment, the target sequence is located within the nucleus or cytoplasm of a cell.

상기 Cas 단백질은 CRISPR RNA(crRNA) 및 트랜스-활성화 crRNA(trans-activating crRNA, tracrRNA)로 불리는 두 RNA와 복합체를 형성할 때, 활성 엔도뉴클레아제 또는 니카아제(nickase)를 형성한다. 상기 Cas 단백질의 비제한적인 예는 Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9(Csn1 및 Csx12로도 알려짐), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, 그의 상동체 또는 그의 변형된 버전을 포함할 수 있다. 이들 효소가 알려져 있으며; 예를 들어, 스트렙토코커스 피오게네스 Cas9 단백질의 아미노산 서열은 수탁 번호 Q99ZW2 하에 스위스프로트(SwissProt) 데이터베이스에서 얻을 수 있다. 일 구체예에서, 비변형 CRISPR 효소, 예를 들어, Cas9는 DNA 절단 활성을 갖는다.The above Cas protein forms an active endonuclease, or nickase, when it forms a complex with two RNAs called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA). Non-limiting examples of the above Cas proteins include Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, a homolog thereof or a homolog thereof. Modified versions may be included. These enzymes are known; for example, the amino acid sequence of the Streptococcus pyogenes Cas9 protein is available from the SwissProt database under accession number Q99ZW2. In one embodiment, the unmodified CRISPR enzyme, e.g., Cas9, has DNA cleavage activity.

일 구체예에서, CRISPR 효소는 Cas9 단백질일 수 있고, 상기 Cas9 단백질은 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질, 캄필로박터 제주니(Campylobacter jejuni) 유래 Cas9 단백질, 스트렙토코커스 써모필러스(Streptococcus thermophiles) 유래 Cas9 단백질, 스트렙토코커스 아우레우스(Streptocuccus aureus) 유래 Cas9 단백질 및 네이세리아 메닝기디티스(Neisseria meningitidis) 유래 Cas9 단백질로 이루어진 군에서 선택되는 하나 이상의 Cas9 단백질일 수 있고, 구체적으로 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질일 수 있다. 일 구체예에서, Cas9 단백질은 진핵 세포에서의 발현을 위해 코돈-최적화되며, 상기 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질을 사용하는 경우, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 최대로 감소될 수 있다.In one specific example, the CRISPR enzyme may be a Cas9 protein, and the Cas9 protein may be one or more Cas9 proteins selected from the group consisting of a Cas9 protein derived from Streptococcus pyogenes , a Cas9 protein derived from Campylobacter jejuni , a Cas9 protein derived from Streptococcus thermophiles, a Cas9 protein derived from Streptococcus aureus , and a Cas9 protein derived from Neisseria meningitidis , and specifically, a Cas9 protein derived from Streptococcus pyogenes . In one embodiment, the Cas9 protein is codon-optimized for expression in a eukaryotic cell, and when using the Streptococcus pyogenes derived Cas9 protein, the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be maximally reduced.

상기 용어 “핵 위치화 서열 또는 신호(Nuclear localization sequence or signal, NLS)"는 특정물질(예컨대, 단백질)을 세포 핵 내로 운반하는 역할을 하는 아미노산 서열을 의미하며, 대체적으로 핵공(Nuclear Pore)을 통하여 세포 핵 내로 운반하는 작용을 한다. 상기 핵 위치화 서열은 진핵 생물에서 CRISPR 복합체 활성에 필요하지 않지만, 이러한 서열을 포함하여, 시스템의 활성을 증진시켜, 특히 핵 내의 핵산 분자를 표적화하는 것으로 여겨진다.The term “nuclear localization sequence or signal (NLS)” refers to an amino acid sequence that functions to transport a specific substance (e.g., a protein) into the cell nucleus, and generally functions to transport it into the cell nucleus through a nuclear pore. Although the nuclear localization sequence is not required for the activity of the CRISPR complex in eukaryotes, it is believed that including such a sequence enhances the activity of the system, particularly targeting nucleic acid molecules in the nucleus.

또한 RNA 유전자 가위(RNA-guided CRISPR)(clustered regularly interspaced short palindrome repeats)-연관된 뉴클레아제 Cas9는 표적 유전자의 넉아웃, 전사 활성화 및 single guide RNA(sgRNA)(즉, crRNA-tracrRNA 융합 전사체)를 이용한 억제에 대한 획기적인 기술을 제공하며, 이 기술은 수많은 유전자 위치를 타겟팅하는 것으로 알려져 있다.Additionally, RNA-guided CRISPR (clustered regularly interspaced short palindrome repeats)-associated nuclease Cas9 offers a breakthrough technology for knockout, transcriptional activation, and repression of target genes using a single guide RNA (sgRNA) (i.e., crRNA-tracrRNA fusion transcript), and this technology is known to target numerous genetic loci.

Cas9 (또는 Cpf1) 단백질은 CRISPR/Cas9 시스템에서 필수적인 단백질 요소를 의미하고, 상기 Cas9 (또는 Cpf1) 유전자 및 단백질의 정보는 국립생명공학정보센터(national center for biotechnology information, NCBI)의 GenBank에서 구할 수 있다. Cas (또는 Cpf1)단백질을 암호화하는 CRISPR - 연관 유전자는 약 40 개 이상의 서로 다른 Cas (또는 Cpf1) 단백질 패밀리가 존재하는 것으로 알려져 있으며, Cas 유전자 및 반복 구조(repeat structure)의 특정 조합에 따라 8개의 CRISPR 하위 유형 (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, 및 Mtube)을 정의할 수 있다. 따라서 상기 각 CRISPR 하위 유형이 반복단위를 이루어 폴리리보뉴클레오티드-단백질 복합체를 형성할 수 있다.The Cas9 (or Cpf1) protein refers to an essential protein component in the CRISPR/Cas9 system, and information on the Cas9 (or Cpf1) gene and protein can be obtained from GenBank of the National Center for Biotechnology Information (NCBI). CRISPR-associated genes encoding Cas (or Cpf1) proteins are known to exist in more than 40 different Cas (or Cpf1) protein families, and eight CRISPR subtypes (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, and Mtube) can be defined according to specific combinations of Cas genes and repeat structures. Therefore, each of the above CRISPR subtypes can form a repeat unit to form a polyribonucleotide-protein complex.

상기 유전자 넉아웃은 유전자의 전부 또는 일부 (예컨대, 하나 이상의 뉴클레오티드)의 결실, 치환, 및/또는 하나 이상의 뉴클레오티드의 삽입에 의한 유전자의 활성 조절, 예컨대, 불활성화를 의미하는 것일 수 있다. 상기 유전자 불활성화는 유전자의 발현 억제 또는 발현 감소 (downregulation) 또는 본래의 기능을 상실한 단백질을 코딩하도록 변형된 것을 의미한다. 또한 유전자 조절은 타겟 유전자의 하나 이상의 엑손을 둘러싸고 있는 양쪽 인트론 부위를 동시에 표적함으로 인한 엑손 부위의 결실로 인해 얻어지는 단백질의 구조 변형, Dominant negative 형태의 단백질 발현 등의 결과에 의한 유전자의 기능 변화를 의미하는 것일 수 있다.The above gene knockout may refer to the regulation of the activity of a gene, e.g., inactivation, by deletion, substitution, and/or insertion of one or more nucleotides in whole or in part of the gene. The gene inactivation may refer to the suppression or downregulation of the expression of the gene, or the modification so that the gene encodes a protein that has lost its original function. In addition, the gene regulation may refer to a change in the function of the gene due to the structural modification of the protein obtained by the deletion of the exon region by simultaneously targeting both intron regions surrounding one or more exons of the target gene, or the expression of the protein in a dominant negative form.

일 구체예에서 상기 줄기세포에서 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자를 넉아웃 시키기 위하여 사용하는 표적 서열은 예컨대 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자, 구체적으로는 HLA-A를 암호화하는 유전자, HLA-B를 암호화하는 유전자 및 HLA-DRA를 암호화하는 유전자로 이루어진 군에서 선택되는 하나 이상 보다 구체적으로는 HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-A를 암호화하는 유전자의 엑손 3 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위, HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 포함할 수 있다.In one specific example, the target sequence used to knock out a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II in the stem cell may include, for example, a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, specifically, at least one selected from the group consisting of a gene encoding HLA-A, a gene encoding HLA-B, and a gene encoding HLA-DRA, and more specifically, at least one selected from the group consisting of an exon 2 region of a gene encoding HLA-A, an exon 3 region of a gene encoding HLA-B, an exon 3 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-DRA, and an exon 3 region of a gene encoding HLA-DRA.

일 구체예에서는, HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 2 부위를 모두 포함할 수 있다.In one specific example, it may include all of the exon 2 region of the gene encoding HLA-A, the exon 3 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA.

일 제조예에서는, HLA-A의 경우 엑손 2 내 코돈 37 - 43 내 위치하는 ACAGCGACGCCGCGAGCCAG 염기서열을 표적 서열로, AGG 서열을 PAM 서열로 하는 G0002-HLA-A-g1(+, ACAGCGACGCCGCGAGCCAG, PAM: AGG) gRNA를 제조하였으며, HLA-B의 경우 엑손 2 내 코돈 31 - 38 내 위치하는 GCTGTCGAACCTCACGAACT 염기서열을 표적 서열로, GGG 서열을 PAM 서열로 하는 G0002 - HLA-B - g1(-, GCTGTCGAACCTCACGAACT, PAM: GGG) gRNA를 제조하였으며, HLA-DRA의 경우는 엑손 2 내 코돈 36 - 42 내 위치하는 TGGCAAAGAAGGAGACGGTC 염기서열을 표적 서열로, TGG 서열을 PAM 서열로 하는 G0002-HLA-DRA-g2(+, TGGCAAAGAAGGAGACGGTC, PAM: TGG) gRNA를 제조하였다(제조예 1 참조).In one manufacturing example, in the case of HLA-A, G0002-HLA-A-g1(+, ACAGCGACGCCGCGAGCCAG, PAM: AGG) gRNA was manufactured with the ACAGCGACGCCGCGAGCCAG base sequence located within codons 37 to 43 in exon 2 as the target sequence and the AGG sequence as the PAM sequence, and in the case of HLA-B, G0002-HLA-B-g1(-, GCTGTCGAACCTCACGAACT, PAM: GGG) gRNA was manufactured with the GCTGTCGAACCTCACGAACT base sequence located within codons 31 to 38 in exon 2 as the target sequence and the GGG sequence as the PAM sequence, and in the case of HLA-DRA, TGGCAAAGAAGGAGACGGTC base sequence located within codons 36 to 42 in exon 2 as the target sequence and the TGG sequence as the PAM sequence. G0002-HLA-DRA-g2(+, TGGCAAAGAAGGAGACGGTC, PAM: TGG) gRNA was prepared (see Preparation Example 1).

일 실시예에서는, 상기 디자인한 gRNA 서열의 유효성을 확인하기 위해 ICE 분석을 수행하였으며, 그 결과 G0002-HLA-A-g1가 91%, G0002-HLA-B-g1이 78%, G0002-HLA-DRA-g2가 86%의 효율성을 갖는 것을 확인하였다(실시예 1 참조).In one embodiment, ICE analysis was performed to confirm the effectiveness of the designed gRNA sequences, and as a result, it was confirmed that G0002-HLA-A-g1 had an efficiency of 91%, G0002-HLA-B-g1 had an efficiency of 78%, and G0002-HLA-DRA-g2 had an efficiency of 86% (see Example 1).

상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 감소되도록 인위적으로 수행하는 유전자 조작은 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상이 본래의 기능을 갖는 단백질 형태로 발현되지 않도록 하는 것일 수 있다. 상기 유전자의 조작은 다음 중 하나 이상에 의하여 유도된 것일 수 있다:The genetic manipulation that is artificially performed to reduce the expression or activity of one or more selected from the group consisting of the human leukocyte antigen class I and human leukocyte antigen class II or the gene encoding the same may be such that one or more selected from the group consisting of the human leukocyte antigen class I and human leukocyte antigen class II is not expressed in the form of a protein having its original function. The manipulation of the gene may be induced by one or more of the following:

1) HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 전부 또는 일부 결실, 예컨대, HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 1bp 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드의 결실,1) Deletion of all or part of the gene encoding HLA class I and/or HLA class II, e.g., deletion of 1 bp or more of the nucleotides of the gene encoding HLA class I and/or HLA class II, e.g., deletion of 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide,

2) HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 1bp 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드의 원래(모세포)와 상이한 뉴클레오티드로의 치환,2) Substitution of 1 bp or more of the nucleotides of the gene encoding HLA class I and/or HLA class II, for example, 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide, with a nucleotide different from the original (parent cell),

3) 하나 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드 (각각 독립적으로 A, T, C 및 G 중에서 선택됨)의 타겟 유전자의 임의의 위치에의 삽입, 및3) insertion of one or more nucleotides, e.g., 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide (each independently selected from A, T, C, and G) into any position of the target gene, and

4) 상기 1) 내지 3) 중에서 선택된 2 가지 이상의 조합.4) A combination of two or more selected from 1) to 3) above.

상기 HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 변형되는 일부 ('타겟 부위')는 상기 유전자 중의 1bp 이상, 3bp 이상, 5bp 이상, 7bp 이상, 10bp 이상, 12bp 이상, 15bp 이상, 17bp 이상, 20bp 이상, 예컨대, 1bp 내지 30bp, 3bp 내지 30bp, 5bp 내지 30bp, 7bp 내지 30bp, 10bp 내지 30bp, 12bp 내지 30bp, 15bp 내지 30bp, 17bp 내지 30bp, 20bp 내지 30bp, 1bp 내지 27bp, 3bp 내지 27bp, 5bp 내지 27bp, 7bp 내지 27bp, 10bp 내지 27bp, 12bp 내지 27bp, 15bp 내지 27bp, 17bp 내지 27bp, 20bp 내지 27bp, 1bp 내지 25bp, 3bp 내지 25bp, 5bp 내지 25bp, 7bp 내지 25bp, 10bp 내지 25bp, 12bp 내지 25bp, 15bp 내지 25bp, 17bp 내지 25bp, 20bp 내지 25bp, 1bp 내지 23bp, 3bp 내지 23bp, 5bp 내지 23bp, 7bp 내지 23bp, 10bp 내지 23bp, 12bp 내지 23bp, 15bp 내지 23bp, 17bp 내지 23bp, 20bp 내지 23bp, 1bp 내지 20bp, 3bp 내지 20bp, 5bp 내지 20bp, 7bp 내지 20bp, 10bp 내지 20bp, 12bp 내지 20bp, 15bp 내지 20bp, 17bp 내지 20bp, 21bp 내지 25bp, 18bp 내지 22bp, 또는 21bp 내지 23bp의 연속하는 염기 서열 부위일 수 있다.The modified portion ('target region') of the gene encoding the HLA class I and/or HLA class II is at least 1 bp, at least 3 bp, at least 5 bp, at least 7 bp, at least 10 bp, at least 12 bp, at least 15 bp, at least 17 bp, at least 20 bp of the gene, for example, 1 bp to 30 bp, 3 bp to 30 bp, 5 bp to 30 bp, 7 bp to 30 bp, 10 bp to 30 bp, 12 bp to 30 bp, 15 bp to 30 bp, 17 bp to 30 bp, 20 bp to 30 bp, 1 bp to 27 bp, 3 bp to 27 bp, 5 bp to 27 bp, 7 bp to 27 bp, 10bp to 27bp, 12bp to 27bp, 15bp to 27bp, 17bp to 27bp, 20bp to 27bp, 1bp to 25bp, 3bp to 25bp, 5bp to 25bp, 7bp to 25bp, 10bp to 25bp, 12bp to 25bp, 15bp to 25bp, 17bp to 25bp, 20bp to 25bp, 1bp to 23bp, 3bp to 23bp, 5bp to 23bp, 7bp to 23bp, 10bp to 23bp, 12bp to 23bp, 15bp to 23bp, It may be a continuous base sequence region of 17bp to 23bp, 20bp to 23bp, 1bp to 20bp, 3bp to 20bp, 5bp to 20bp, 7bp to 20bp, 10bp to 20bp, 12bp to 20bp, 15bp to 20bp, 17bp to 20bp, 21bp to 25bp, 18bp to 22bp, or 21bp to 23bp.

다른 실시예에서, 백만개의 유도만능줄기세포와 CRISPR/CAS9 시스템을 혼합하여 전기 천공법을 실시해 세포 내 형질 주입을 유도하였다. 이 후 약 28일 간 배양하면서 PCR 및 생어 염기서열 분석을 수행하여 수집한 클론을 스크리닝하고 유전자형 분석을 수행한 결과, 최종적으로 HLA-A, HLA-B 및 HLA-DRA 유전자가 조작된 세포를 선정하였다. 상기 세포의 시퀀싱을 검증한 결과, HLA-A 영역에서는 1 bp가 삽입되고 HLA-B 영역에서는 28 bp가 결실되었으며, HLA-DRA 영역에서는 1 bp의 결실이 발생한 것을 확인할 수 있었다(실시예 2 참조).In another embodiment, one million induced pluripotent stem cells were mixed with the CRISPR/CAS9 system and electroporated to induce intracellular transfection. After approximately 28 days of culture, PCR and Sanger sequencing were performed to screen the collected clones, and genotyping was performed to ultimately select cells in which the HLA-A, HLA-B, and HLA-DRA genes were manipulated. Sequencing of the cells revealed that a 1 bp insertion occurred in the HLA-A region, a 28 bp deletion occurred in the HLA-B region, and a 1 bp deletion occurred in the HLA-DRA region (see Example 2).

용어 "키메라 RNA", "키메라 가이드 RNA", "가이드 RNA", "단일의 가이드 RNA(single guide RNA, sgRNA)" 및 "합성 가이드 RNA"는 상호교환가능하게 사용되며, 가이드 서열, tracr 서열 및/또는 tracr 메이트 서열을 포함하는 폴리뉴클레오티드 서열을 지칭한다. 용어 "가이드 서열"은 표적 부위를 지정하는 가이드 RNA 내의 약 20bp 서열을 지칭하며, 용어 "가이드" 또는 "스페이서"와 상호교환가능하게 사용될 수 있다. 또한, 용어 "tracr 메이트 서열"은 용어 "직접 반복부(들)"와 상호교환가능하게 사용될 수 있다. 상기 가이드 RNA는 두 개의 RNA, 즉, CRISPR RNA (crRNA) 및 트랜스 활성화 crRNA (transactivating crRNA, tracrRNA)로 이루어져 있는 것일 수 있으며, 또는 crRNA 및 tracrRNA의 부분을 포함하고 상기 표적 DNA와 혼성화하는 단일 사슬 RNA (single-chain RNA, sgRNA)일 수 있다.The terms "chimeric RNA", "chimeric guide RNA", "guide RNA", "single guide RNA (sgRNA)" and "synthetic guide RNA" are used interchangeably and refer to a polynucleotide sequence comprising a guide sequence, a tracr sequence and/or a tracr mate sequence. The term "guide sequence" refers to a sequence of about 20 bp within a guide RNA that directs a target site and may be used interchangeably with the term "guide" or "spacer". The term "tracr mate sequence" may also be used interchangeably with the term "direct repeat(s)". The guide RNA may be composed of two RNAs, namely a CRISPR RNA (crRNA) and a transactivating crRNA (tracrRNA), or may be a single-chain RNA (sgRNA) comprising portions of crRNA and tracrRNA and hybridizing to the target DNA.

일반적으로, 가이드 서열은 표적 서열과 혼성화하고, 표적 서열로의 CRISPR 복합체의 서열-특이적 결합을 유도하기에 충분한, 표적 폴리뉴클레오티드 서열과의 상보성을 갖는 임의의 폴리뉴클레오티드 서열이다. 또한 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성을 감소시키기 위한 유전자 조작에 이용할 수 있는 염기 서열이라면 제한 없이 가이드 RNA로 이용할 수 있으며, 예컨대 상기 염기 서열은 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자와 혼성화할 수 있는 서열일 수 있다. 또한 상기 가이드 RNA의 기능을 변형/증진시키기 위하여 가이드 RNA 염기 서열의 일부분을 변형할 수 있다. 또한 일 구체예에서, 가이드 서열과 그의 상응하는 표적 서열 간의 상보성의 정도는 적절한 정렬 알고리즘을 사용하여 최적으로 정렬되는 경우, 약 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99% 이상이다. 최적의 정렬은 서열을 정렬하기에 적절한 임의의 알고리즘의 사용으로 결정될 수 있으며, 그의 비제한적인 예는 스미스-워터만(Smith-Waterman) 알고리즘, 니들만-분쉬(Needleman-Wunsch) 알고리즘, 버로우즈-휠러 트랜스폼(Burrows-Wheeler Transform)에 기초한 알고리즘(예를 들어, 버로우즈 휠러 얼라이너(Burrows Wheeler Aligner)), ClustalW, Clustal X, BLAT, 노보얼라인(Novoalign)(노보크라프트 테크놀로지즈(Novocraft Technologies), ELAND(일루미나(Illumina), 미국 캘리포니아주 샌디에고), SOAP(soap.genomics.org.cn에서 이용가능) 및 Maq(maq.sourceforge.net에서 이용가능)를 포함할 수 있다. 일 구체예에서, 가이드 서열은 예컨대 약 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75개 이상의 뉴클레오티드 길이이다. 일 구체예에서, 가이드 서열은 약 75, 50, 45, 40, 35, 30, 25, 20, 15, 12개 이하의 뉴클레오티드 길이이다. 표적 서열로의 CRISPR 복합체의 서열-특이적 결합을 유도하는 가이드 서열의 능력은 임의의 적절한 검정에 의해 평가될 수 있다. 예를 들어, 시험되는 가이드 서열을 포함하는 CRISPR 복합체를 형성하기에 충분한 CRISPR 시스템의 성분은 예를 들어, CRISPR 서열의 성분을 인코딩하는 벡터로의 트랜스펙션 후에, 예를 들어, 본원에 기술된 바와 같은 서베이어 검정에 의한 표적 서열 내의 우선적인 절단의 평가에 의해서와 같이, 상응하는 표적 서열을 갖는 숙주 세포로 제공될 수 있다. 유사하게, 표적 폴리뉴클레오티드 서열의 절단은 표적 서열, 시험되는 가이드 서열 및 시험 가이드 서열과 상이한 대조군 가이드 서열을 포함하는 CRISPR 복합체의 성분을 제공하고, 표적 서열에서 시험 및 대조군 가이드 서열 반응 간의 결합 또는 절단 비율을 비교함으로써 시험관에서 평가될 수 있다. 다른 검정이 가능하며, 당업자에게 용이하게 사용될 수 있을 것이다.In general, a guide sequence is any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and induce sequence-specific binding of a CRISPR complex to the target sequence. In addition, any base sequence that can be used for genetic manipulation to reduce the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be used as a guide RNA without limitation. For example, the base sequence may be a sequence that can hybridize with a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II. In addition, a portion of the guide RNA base sequence can be modified to modify/enhance the function of the guide RNA. Also, in one specific embodiment, the degree of complementarity between the guide sequence and its corresponding target sequence is about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99% or more when optimally aligned using an appropriate alignment algorithm. An optimal alignment can be determined by the use of any suitable algorithm for aligning sequences, non-limiting examples of which include the Smith-Waterman algorithm, the Needleman-Wunsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g., the Burrows Wheeler Aligner), ClustalW, Clustal X, BLAT, Novoalign (Novocraft Technologies), ELAND (Illumina, San Diego, CA), SOAP (available at soap.genomics.org.cn), and Maq (available at maq.sourceforge.net). In one embodiment, the guide sequence comprises, for example, about 5, 10, 11, 12, 13, 14, 15, The guide sequence is at least about 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75 nucleotides in length. In one embodiment, the guide sequence is no more than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12 nucleotides in length. The ability of a guide sequence to direct sequence-specific binding of a CRISPR complex to a target sequence can be assessed by any suitable assay. For example, components of a CRISPR system sufficient to form a CRISPR complex comprising the tested guide sequence can be assessed by, for example, assessing preferential cleavage within the target sequence by, for example, a Surveyor assay as described herein, after transfection with a vector encoding components of the CRISPR sequence. A host cell containing the corresponding target sequence can be provided. Similarly, cleavage of the target polynucleotide sequence can be assessed in vitro by providing components of a CRISPR complex comprising the target sequence, the test guide sequence, and a control guide sequence different from the test guide sequence, and comparing the binding or cleavage rates between the test and control guide sequence reactions at the target sequence. Other assays are possible and will be readily available to those skilled in the art.

가이드 서열은 임의의 표적 서열을 표적화하도록 선택될 수 있다. 일 구체예에서, 표적 서열은 세포의 게놈 내의 서열이다. 예시적인 표적 서열은 표적 게놈에서 독특한 것들을 포함할 수 있다. 예를 들어, 스트렙토코커스 피오게네스 유래 Cas9에 대하여, 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMNNNNNNNNNNNNXGG의 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNNXGG (N은 A, G, T 또는 C이며; X는 임의의 것일 수 있음)는 게놈 내에 단일의 존재를 갖는다. 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMMNNNNNNNNNNNXGG의 스트렙토코커스 피오게네스 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNXGG (N은 A, G, T 또는 C이며; X는 임의의 것일 수 있음)는 게놈 내에 단일의 존재를 갖는다. 스트렙토코커스 써모필러스 CRISPR1 Cas9에 대하여, 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMNNNNNNNNNNNNXXAGAAW의 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNNXXAGAAW (N은 A, G, T 또는 C이고; X는 임의의 것일 수 있으며; W는 A 또는 T임)는 게놈 내에 단일의 존재를 갖는다. 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMMNNNNNNNNNNNXXAGAAW의 스트렙토코커스 써모필러스 CRISPR1 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNXXAGAAW(N은 A, G, T 또는 C이고; X는 임의의 것일 수 있으며; W는 A 또는 T임)는 게놈 내에 단일의 존재를 갖는다. 스트렙토코커스 피오게네스 Cas9에 대하여, 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMNNNNNNNNNNNNXGGXG의 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNNXGGXG (N은 A, G, T 또는 C이고; X는 임의의 것일 수 있음)는 게놈 내에 단일의 존재를 갖는다. 게놈 내의 독특한 표적 서열은 형태 MMMMMMMMMNNNNNNNNNNNXGGXG의 스트렙토코커스 피오게네스 Cas9 표적 부위를 포함할 수 있으며, 여기서, NNNNNNNNNNNXGGXG (N은 A, G, T 또는 C이고; X는 임의의 것일 수 있음)는 게놈 내에 단일의 존재를 갖는다. 이들 서열 각각에서, "M"은 A, G, T 또는 C일 수 있다.The guide sequence can be selected to target any target sequence. In one embodiment, the target sequence is a sequence within the genome of the cell. Exemplary target sequences can include those that are unique within the target genome. For example, for a Cas9 from Streptococcus pyogenes, a unique target sequence within the genome can include a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXGG, wherein NNNNNNNNNNNNXGG (N is A, G, T, or C; X can be anything) has a single occurrence within the genome. A unique target sequence within the genome can include a Streptococcus pyogenes Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXGG, wherein NNNNNNNNNNNXGG (N is A, G, T, or C; X can be anything) has a single occurrence within the genome. For Streptococcus thermophilus CRISPR1 Cas9, the unique target sequence within the genome can comprise a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXXAGAAW, wherein NNNNNNNNNNNNXXAGAAW (N is A, G, T, or C; X can be anything; and W is A or T) has a single occurrence within the genome. The unique target sequence within the genome can comprise a Streptococcus thermophilus CRISPR1 Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXXAGAAW, wherein NNNNNNNNNNNXXAGAAW (N is A, G, T, or C; X can be anything; and W is A or T) has a single occurrence within the genome. For Streptococcus pyogenes Cas9, the unique target sequence within the genome can comprise a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXGGXG, wherein NNNNNNNNNNNNXGGXG (N is A, G, T, or C; and X can be anything) has a single occurrence within the genome. The unique target sequence within the genome can comprise a Streptococcus pyogenes Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXGGXG, wherein NNNNNNNNNNNXGGXG (N is A, G, T, or C; and X can be anything) has a single occurrence within the genome. In each of these sequences, "M" can be A, G, T, or C.

일 구체예에서, 상기 CRISPR/Cas 시스템은 서열번호 1의 염기서열로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어진 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어진 폴리뉴클레오티드로 이루어진 군에서 선택되는 하나 이상의 폴리뉴클레오티드를 포함할 수 있다. In one specific example, the CRISPR/Cas system may include one or more polynucleotides selected from the group consisting of a polynucleotide comprising a base sequence of SEQ ID NO: 1, a polynucleotide comprising a base sequence of SEQ ID NO: 2, and a polynucleotide comprising a base sequence of SEQ ID NO: 3.

일 구체예에서, 상기 CRISPR/Cas 시스템은 서열번호 1의 염기서열로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어진 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어진 폴리뉴클레오티드를 모두 포함할 수 있다.In one specific example, the CRISPR/Cas system may include a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3.

일 구체예에서, 상기 폴리뉴클레오티드는 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자에 상보적으로 결합할 수 있으며, 구체적으로는 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 엑손 1 내지 엑손 3으로 이루어진 부위에서 선택되는 하나 이상의 부위에 결합하는 것일 수 있다.In one specific example, the polynucleotide can complementarily bind to a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, and specifically, can bind to at least one site selected from a site consisting of exon 1 to exon 3 of the gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II.

일 구체예에서, 상기 폴리뉴클레오티드는 서열번호 1의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 예를 들어, 서열번호 1의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다.In one specific example, the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 1, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 1.

일 구체예에서, 상기 폴리뉴클레오티드는 서열번호 2의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 예를 들어, 서열번호 2의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다.In one specific example, the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 2, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 2.

일 구체예에서, 상기 폴리뉴클레오티드는 서열번호 3의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 예를 들어, 서열번호 3의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다.In one specific example, the polynucleotide may include a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 3, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 3.

일 구체예에서, 상기 서열번호 1 내지 3의 염기서열로 이루어진 폴리뉴클레오티드를 포함하는 염기서열은 CRISPR/CAS 시스템의 gRNA 역할을 수행할 수 있고, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자를 표적으로 할 수 있다. 구체적으로는, HLA-A를 암호화하는 유전자의 엑손 1 내지 3 부위, HLA-B를 암호화하는 유전자의 엑손 1 내지 3 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 1 내지 3 부위로 이루어진 부위에서 선택되는 하나 이상의 부위를 표적으로 할 수 있으며, 보다 구체적으로는 HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-A를 암호화하는 유전자의 엑손 3 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위, HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 3 부위로 이루어진 부위에서 선택되는 하나 이상의 부위를 표적으로 할 수 있다. In one specific example, a base sequence comprising a polynucleotide consisting of the base sequence of SEQ ID NOs: 1 to 3 can serve as a gRNA of the CRISPR/CAS system and can target a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II. Specifically, one or more regions selected from regions consisting of exons 1 to 3 of the gene encoding HLA-A, exons 1 to 3 of the gene encoding HLA-B, and exons 1 to 3 of the gene encoding HLA-DRA may be targeted, and more specifically, one or more regions selected from regions consisting of exon 2 of the gene encoding HLA-A, exon 3 of the gene encoding HLA-A, exon 2 of the gene encoding HLA-B, exon 3 of the gene encoding HLA-B, exon 2 of the gene encoding HLA-DRA, and exon 3 of the gene encoding HLA-DRA may be targeted.

일 구체예에서는, HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 2 부위를 모두 표적으로 할 수 있다. In one specific example, all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be targeted.

일 구체예에서, 상기 HLA-A를 암호화하는 유전자의 엑손 2 부위는 서열번호 4의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 상기 HLA-A를 암호화하는 유전자의 엑손 2 부위 및 엑손 3 부위는 서열번호 5의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다. In one specific example, the exon 2 region of the gene encoding HLA-A may include a polynucleotide consisting of a base sequence of SEQ ID NO: 4, and the exon 2 region and exon 3 region of the gene encoding HLA-A may include a polynucleotide consisting of a base sequence of SEQ ID NO: 5.

일 구체예에서, 상기 서열번호 4의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 4의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 4의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide consisting of the base sequence of SEQ ID NO: 4 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 4, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 4.

일 구체예에서, 상기 서열번호 5의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 5의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 5의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide consisting of the base sequence of SEQ ID NO: 5 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 5, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 5.

일 구체예에서, 상기 HLA-B를 암호화하는 유전자의 엑손 2 부위는 서열번호 6의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 상기 HLA-B를 암호화하는 유전자의 엑손 2 부위 및 엑손 3 부위는 서열번호 7의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다. In one specific example, the exon 2 region of the gene encoding HLA-B may include a polynucleotide consisting of a base sequence of SEQ ID NO: 6, and the exon 2 region and exon 3 region of the gene encoding HLA-B may include a polynucleotide consisting of a base sequence of SEQ ID NO: 7.

일 구체예에서, 상기 서열번호 6의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 6의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 6의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide consisting of the base sequence of SEQ ID NO: 6 may be a polynucleotide consisting of a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 6, for example, a polynucleotide consisting of a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 6.

일 구체예에서, 상기 서열번호 7의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 7의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 7의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide comprising the base sequence of SEQ ID NO: 7 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 7, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 7.

일 구체예에서, 상기 HLA-DRA를 암호화하는 유전자의 엑손 2 부위는 서열번호 8의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있고, 상기 HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 엑손 3 부위는 서열번호 9의 염기서열로 이루어진 폴리뉴클레오티드를 포함할 수 있다. In one specific example, the exon 2 region of the gene encoding the HLA-DRA may include a polynucleotide consisting of the base sequence of SEQ ID NO: 8, and the exon 2 region and the exon 3 region of the gene encoding the HLA-DRA may include a polynucleotide consisting of the base sequence of SEQ ID NO: 9.

일 구체예에서, 상기 서열번호 8의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 8의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 8의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide comprising the base sequence of SEQ ID NO: 8 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 8, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 8.

일 구체예에서, 상기 서열번호 9의 염기서열로 이루어진 폴리뉴클레오티드는 서열번호 9의 염기서열과 70% 이상의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있고, 예를 들어, 서열번호 9의 염기 서열과 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% 또는 99% 이상 또는 100%의 상동성을 갖는 염기 서열로 이루어진 폴리뉴클레오티드일 수 있다.In one specific example, the polynucleotide comprising the base sequence of SEQ ID NO: 9 may be a polynucleotide comprising a base sequence having 70% or more homology with the base sequence of SEQ ID NO: 9, for example, a polynucleotide comprising a base sequence having 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more or 100% homology with the base sequence of SEQ ID NO: 9.

일 구체예에서, 상기 CRISPR/Cas 시스템은 Cas9(CRISPR associated protein 9) 단백질 또는 Cas9 단백질을 코딩하는 유전자 및 NLS(Nuclear Localization Signal) 단백질 또는 NLS 단백질을 코딩하는 유전자를 포함할 수 있다.In one specific example, the CRISPR/Cas system may include a Cas9 (CRISPR associated protein 9) protein or a gene encoding a Cas9 protein and a NLS (Nuclear Localization Signal) protein or a gene encoding an NLS protein.

일 구체예에서, 상기 "시스템"은 상기 서열번호 1의 염기서열로 이루어지는 폴리뉴크레오티드, 서열번호 2의 염기서열로 이루어지는 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어지는 폴리뉴클레오티드로 이루어진 군에서 선택되는 하나 이상을 포함하는 핵산 및 Cas 단백질의 복합체를 포함할 수 있고 구체적으로는, 서열번호 1로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어지는 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어지는 폴리뉴클레오티드; 및 Cas 단백질의 복합체를 포함할 수 있다. 보다 구체적으로는, 상기 Cas 단백질은 Cas9 단백질일 수 있다.In one specific example, the "system" may include a complex of a nucleic acid and a Cas protein, wherein the nucleic acid includes at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3, and specifically, a complex of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a Cas protein. More specifically, the Cas protein may be a Cas9 protein.

일 구체예에서, RNP와 관련하여, pre-formed RNP는 세포로 도입되어 타겟 게놈 DNA 편집을 시작할 수 있다. 이러한 RNP 형태의 전달은 벡터 전달에 비해 오프 타겟 효과를 감소시킬 수 있고, 세포의 핵으로 Cas 단백질과 gRNA를 직접 전달 가능하다는 장점이 있다.In one specific example, with respect to RNPs, preformed RNPs can be introduced into cells to initiate target genomic DNA editing. This RNP-based delivery has the advantage of reducing off-target effects compared to vector delivery and enabling direct delivery of the Cas protein and gRNA to the nucleus of the cell.

RNP (ribonucleoprotein) 형성을 위해 Cas 단백질과 gRNA 각각을 합성 및 정제하여 RNP 형태로 제작할 수 있다. 경우에 따라서 Cas 단백질 및 절단인자 RNA를 플라스미드를 통해 직접 발현 후 self-assembly 형태로 조립된 RNP를 정제하여 제작할 수 있다To form ribonucleoproteins (RNPs), Cas proteins and gRNAs can be synthesized and purified individually to produce RNPs. In some cases, Cas proteins and cleavage factor RNAs can be directly expressed via plasmids, and then the self-assembled RNPs can be purified.

또한, 상기 "시스템"은, 상기 서열번호 1의 염기서열로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어지는 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어지는 폴리뉴클레오티드로 이루어진 군에서 선택되는 어느 하나 이상을 암호화하는 폴리뉴클레오티드; 및 Cas 단백질을 암호화하는 폴리뉴클레오티드를 포함하는 벡터일 수 있고, 상기 서열번호 1의 염기서열로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어지는 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어지는 폴리뉴클레오티드를 암호화하는 폴리뉴클레오티드; 및 Cas 단백질을 암호화하는 폴리뉴클레오티드를 포함하는 벡터일 수 있다. 보다 구체적으로는, 상기 Cas 단백질은 Cas9 단백질일 수 있다.In addition, the "system" may be a vector including a polynucleotide encoding at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein; and a vector including a polynucleotide encoding a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein. More specifically, the Cas protein may be a Cas9 protein.

일 구체예에서, 상기 서열번호 1의 염기서열로 이루어지는 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어지는 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어지는 폴리뉴클레오티드로 이루어진 군에서 선택되는 어느 하나 이상을 암호화하는 폴리뉴클레오티드; 및 Cas 단백질을 암호화하는 폴리뉴클레오티드는 동일한 벡터에 포함되어 있을 수 있고, 상이한 벡터에 각각 포함되어 있을 수 있다.In one specific example, a polynucleotide encoding at least one selected from the group consisting of a polynucleotide comprising the base sequence of SEQ ID NO: 1, a polynucleotide comprising the base sequence of SEQ ID NO: 2, and a polynucleotide comprising the base sequence of SEQ ID NO: 3; and a polynucleotide encoding a Cas protein may be contained in the same vector, or may be contained in different vectors.

상기 용어 "벡터(vector)"는 숙주 세포에서 목적 유전자를 발현시키기 위한 수단을 의미한다. 예를 들어, 플라스미드 벡터, 코즈미드 벡터 및 박테리오파아지 벡터, 아데노바이러스 벡터, 레트로바이러스 벡터 및 아데노-연관 바이러스 벡터와 같은 바이러스 벡터를 포함할 수 있다. 상기 재조합 벡터로 사용될 수 있는 벡터는 당업계에서 종종 사용되는 플라스미드 (예를 들면, V1k_GE, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX 시리즈, pET 시리즈 및 pUC19 등), 파지 또는 바이러스 (예를 들면, SV40 등)를 조작하여 제작될 수 있다.The above term "vector" refers to a means for expressing a target gene in a host cell. For example, it may include a plasmid vector, a cosmid vector, a bacteriophage vector, an adenovirus vector, a retrovirus vector, and a viral vector such as an adeno-associated virus vector. A vector that can be used as the above recombinant vector can be produced by manipulating a plasmid (e.g., V1k_GE, pSC101, pGV1106, pACYC177, ColE1, pKT230, pME290, pBR322, pUC8/9, pUC6, pBD9, pHC79, pIJ61, pLAFR1, pHV14, pGEX series, pET series, and pUC19, etc.), a phage, or a virus (e.g., SV40, etc.) that is frequently used in the art.

상기 벡터에서 상기 인간 백혈구 항원 클래스 Ⅰ 및 인간 백혈구 항원 클래스 Ⅱ로 이루어지는 군에서 선택되는 하나 이상을 암호화하는 유전자에 결합하는 가이드 RNA를 코딩하는 유전자, 상기 Cas 단백질을 코딩하는 유전자 및 상기 NLS를 코딩하는 유전자는 프로모터에 작동 가능하게 연결될 수 있다. 상기 용어 "작동 가능하게 연결된(operatively linked)"은 뉴클레오티드 발현 조절 서열(예를 들어, 프로모터 서열)과 다른 뉴클레오티드 서열 사이의 기능적인 결합을 의미한다. 상기 조절 서열은 "작동 가능하게 연결(operatively linked)"됨으로써 다른 뉴클레오티드 서열의 전사 및/또는 해독을 조절할 수 있다.In the above vector, a gene encoding a guide RNA that binds to a gene encoding at least one selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II, a gene encoding the Cas protein, and a gene encoding the NLS may be operably linked to a promoter. The term "operably linked" refers to a functional linkage between a nucleotide expression regulatory sequence (e.g., a promoter sequence) and another nucleotide sequence. The regulatory sequence can regulate transcription and/or translation of another nucleotide sequence by being "operably linked."

일 구체예에서, 상기 CRISPR/Cas 시스템은 전기천공법(electroporation) 방법으로 세포로 전달되는 것일 수 있다.In one specific example, the CRISPR/Cas system may be delivered to cells by electroporation.

상기 용어 "저면역원성"은 세포가 다른 개체에게 이식 시, 야생형과 비교할 때 감소하거나 제거된 면역 거부 반응을 나타내는 것을 의미한다. 면역 거부 반응은 20% 이상, 약 30% 이상, 약 40% 이상, 약 50% 이상, 약 55% 이상, 약 60% 이상, 약 70% 이상, 약 75% 이상, 약 80% 이상, 약 85% 이상, 약 90% 이상, 약 95% 이상, 또는 약 100% 감소된 것일 수 있다.The term "hypoimmunogenicity" above means that the cells, when transplanted into another organism, exhibit a reduced or eliminated immune rejection response compared to the wild type. The immune rejection response may be reduced by at least 20%, at least about 30%, at least about 40%, at least about 50%, at least about 55%, at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%.

또 다른 실시예에서, 유전자가 조작된 줄기세포의 면역원성을 분석한 결과, IFNγ 자극이 주어진 경우와 주어지지 않은 경우 모두 HLA-A, HLA-B 및 HLA-DRA의 발현량이 모세포에 비하여 현저하게 감소하는 것을 확인할 수 있었으며, central memory T cell(TCM)과 effector memory T cell(TEM)의 증식이 되지 않는 것 또한 확인할 수 있어 유전적으로 조작된 줄기세포가 면역원성을 보이지 않는 다는 것을 나타낸다(실시예 4 참조). In another example, the immunogenicity of genetically engineered stem cells was analyzed, and it was confirmed that the expression levels of HLA-A, HLA-B, and HLA-DRA were significantly reduced compared to the parent cells both when IFNγ stimulation was given and when it was not given, and it was also confirmed that central memory T cells (TCM) and effector memory T cells (TEM) did not proliferate, indicating that the genetically engineered stem cells do not exhibit immunogenicity (see Example 4).

상기 "줄기세포"는 미분화된 세포로서 자기 복제 능력을 가지면서 두 개 이상의 서로 다른 종류의 세포로 분화하는 능력을 갖는 세포를 말한다. 상기 줄기세포는 자가 또는 동종 유래 줄기세포일 수 있다. The above "stem cell" refers to an undifferentiated cell that has the ability to self-replicate and differentiate into two or more different types of cells. The stem cell may be an autologous or allogeneic stem cell.

이때 상기 줄기세포는 유도만능줄기세포, 배아줄기세포, 체세포 핵치환 배아줄기세포 또는 성체 줄기세포일 수 있고, 구체적으로는 유도만능줄기세포일 수 있다.At this time, the stem cell may be an induced pluripotent stem cell, an embryonic stem cell, a somatic cell nuclear transfer embryonic stem cell, or an adult stem cell, and specifically, may be an induced pluripotent stem cell.

상기 "유도만능줄기세포"는 분화된 세포들로부터 인위적인 역분화 과정을 통해 다능성 분화능을 가지도록 유도된 세포들을 일컫는 말로서 역분화 줄기세포 라고도 한다. 인위적인 역분화 과정은 레트로바이러스, 렌티바이러스 및 센다이바이러스를 이용한 바이러스 - 매개 또는 비바이러스성 벡터 이용, 단백질 및 세포 추출물 등을 이용하는 비바이러스 - 매개 역분화 인자의 도입에 의해 수행되거나, 줄기세포 추출물, 화합물 등에 의한 역분화 과정을 포함할 수 있다. 유도만능줄기세포는 배아줄기세포와 거의 같은 특성을 가지며, 구체적으로는 비슷한 세포 모양을 보여주고, 유전자, 단백질 발현 패턴이 유사하며, in vitroin vivo에서 전분화능을 가지고, 테라토마(teratoma)를 형성하며, 생쥐의 배반포(blastocyst)에 삽입시켰을 때, 키메라(chimera) 생쥐를 형성하고, 유전자의 생식선 전이 (germline transmission)가 가능하다. The above "induced pluripotent stem cells" refer to cells that are induced to have pluripotent differentiation capacity through an artificial dedifferentiation process from differentiated cells, and are also called induced stem cells. The artificial dedifferentiation process may be performed by using viral or non-viral vectors such as retrovirus, lentivirus, and Sendai virus, or by introducing non-viral-mediated dedifferentiation factors such as proteins and cell extracts, or may include a dedifferentiation process using stem cell extracts, compounds, etc. Induced pluripotent stem cells have almost the same characteristics as embryonic stem cells, specifically, they show a similar cell shape, have similar gene and protein expression patterns, have pluripotency in vitro and in vivo , form teratomas, form chimera mice when inserted into a mouse blastocyst, and are capable of germline transmission of genes.

유도만능줄기세포로는 인간, 원숭이, 돼지, 말, 소, 양, 개, 고양이, 마우스, 랫트 또는 토끼 등의 모든 종 유래의 유도만능줄기세포를 포함하나, 구체적으로는 인간 유래의 유도만능줄기세포일 수 있다. Induced pluripotent stem cells include induced pluripotent stem cells derived from any species, including humans, monkeys, pigs, horses, cows, sheep, dogs, cats, mice, rats, or rabbits, but may specifically be induced pluripotent stem cells derived from humans.

상기 유도만능줄기세포가 역분화 되기 전의 체세포는 제대, 제대혈, 혈액, 골수, 지방, 근육, 신경, 피부, 양막, 양수 또는 태반 등으로부터 유래된 체세포일 수 있다. 구체적으로 상기 체세포는 말초혈액단핵세포(Peripheral blood mononuclear cell), 섬유아세포(fibroblast), 간세포(hepatocyte), 지방세포(adipose cell), 상피세포(epithelial cell), 표피세포(epidermal cell), 연골세포(chondrocyte), 근세포(muscle cell), 심근세포(cardiac muscle cell), 멜라노사이트(melaonocyte), 신경세포(neural cell), 교세포(glial cell), 성상교세포(astroglial cell), 단핵구 (monocyte), 대식세포(macrophage) 등을 포함할 수 있고, 보다 구체적으로는 말초혈액단핵세포 유래 유도만능줄기세포일 수 있다.The somatic cells before the above-mentioned induced pluripotent stem cells are dedifferentiated may be somatic cells derived from umbilical cord, umbilical cord blood, blood, bone marrow, fat, muscle, nerve, skin, amniotic membrane, amniotic fluid, or placenta. Specifically, the somatic cells may include peripheral blood mononuclear cells, fibroblasts, hepatocytes, adipocytes, epithelial cells, epidermal cells, chondrocytes, muscle cells, cardiac muscle cells, melanocytes, neural cells, glial cells, astroglial cells, monocytes, macrophages, and the like, and more specifically, may be induced pluripotent stem cells derived from peripheral blood mononuclear cells.

일 구체예에서, 상기 인간 백혈구 항원 클래스 I은 HLA-A 및 HLA-B로 이루어진 군에서 선택되는 하나 이상일 수 있으며, HLA-A 및 HLA-B를 모두 포함하는 것일 수 있다.In one specific example, the human leukocyte antigen class I may be at least one selected from the group consisting of HLA-A and HLA-B, and may include both HLA-A and HLA-B.

일 구체예에서, 상기 인간 백혈구 항원 클래스 II는 HLA-DRA일 수 있다. In one specific example, the human leukocyte antigen class II may be HLA-DRA.

일 구체예에서, 상기 줄기세포는 OCT4, SSEA4, NANOG, SOX2, ESRRB, TRA-1-60, SOX17, BRACHYURY 및 PAX 6으로 이루어진 군으로부터 선택되는 하나 이상의 단백질을 모세포와 동일한 수준으로 발현할 수 있고, 구체적으로는 OCT4, SSEA4, NANOG, TRA-1-60, SOX17, BRACHYURY 및 PAX 6으로 이루어진 군으로부터 선택되는 하나 이상의 단백질을 모세포와 동일한 수준으로 발현할 수 있다.In one specific example, the stem cell can express at least one protein selected from the group consisting of OCT4, SSEA4, NANOG, SOX2, ESRRB, TRA-1-60, SOX17, BRACHYURY and PAX 6 at the same level as the parent cell, and specifically, can express at least one protein selected from the group consisting of OCT4, SSEA4, NANOG, TRA-1-60, SOX17, BRACHYURY and PAX 6 at the same level as the parent cell.

또 다른 실시예에서, 유전적으로 조작한 줄기세포의 다능성 마커를 발현하는 정도를 분석한 결과, 유전적 조작을 하지 않은 모세포와 유사한 수준으로 다능성 마커 OCT4, SSEA4, NANOG 및 TRA-1-60을 발현하는 것을 확인할 수 있었다. 또한, 3배엽으로 분화할 수 있는 분화능을 나타내는 마커인 SOX17, BRACHYURY 및 PAX 6의 마커도 모세포와 유사한 수준으로 발현하는 것을 확인할 수 있었다(실시예 3 참조).In another example, the degree of expression of pluripotency markers of genetically engineered stem cells was analyzed, and it was confirmed that the pluripotency markers OCT4, SSEA4, NANOG, and TRA-1-60 were expressed at a level similar to that of the parent cells that had not been genetically engineered. In addition, it was confirmed that the markers SOX17, BRACHYURY, and PAX 6, which indicate differentiation potential for differentiation into three germ layers, were expressed at a level similar to that of the parent cells (see Example 3).

또 다른 실시예에서, 유전적으로 조작한 줄기세포의 중배엽 계통 내피세포로 분화능을 확인한 결과, 모세포와 동일하게 중배엽을 거쳐 내피 세포로 분화되었으며, 내피 세포 마커인 CD31 및 VE-Cadherin을 모두 발현하는 것을 확인할 수 있었다(실시예 5).In another example, the differentiation potential of genetically engineered stem cells into mesodermal lineage endothelial cells was confirmed, and it was confirmed that they differentiated into endothelial cells through mesoderm, similar to the parent cells, and expressed both endothelial cell markers CD31 and VE-Cadherin (Example 5).

다른 양상은 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 줄기세포를 유전적으로 조작하는 단계를 포함하는 저면역원성 줄기세포의 제조 방법을 제공한다.Another aspect provides a method for producing hypoimmunogenic stem cells, comprising the step of genetically engineering stem cells to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II.

상기 "인간 백혈구 항원", "줄기세포" 및 "저면역원성"은 전술한 범위 내일 수 있다.The above “human leukocyte antigen”, “stem cell” and “low immunogenicity” may be within the ranges described above.

일 구체예에서, 상기 유전적으로 조작하는 단계는 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위, HLA-B를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위 및 HLA-Dra를 코딩하는 유전자의 엑손 1 내지 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 유전적으로 조작하는 것일 수 있고, 구체적으로는, 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-A를 코딩하는 유전자의 엑손 3 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 3 부위, HLA-DRA를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 유전적으로 조작하는 것일 수 있다. 보다 구체적으로는, 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 2 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 유전적으로 조작하는 것일 수 있다.In one specific example, the genetically manipulating step may be genetically manipulating one or more regions selected from the group consisting of exon 1 to exon 3 of a gene encoding HLA-A, exon 1 to exon 3 of a gene encoding HLA-B, and exon 1 to exon 3 of a gene encoding HLA-Dra of the stem cell, and specifically, may be genetically manipulating one or more regions selected from the group consisting of exon 2 of a gene encoding HLA-A, exon 3 of a gene encoding HLA-A, exon 2 of a gene encoding HLA-B, exon 3 of a gene encoding HLA-B, exon 2 of a gene encoding HLA-DRA, and exon 3 of a gene encoding HLA-DRA of the stem cell. More specifically, it may be a method of genetically manipulating one or more regions selected from the group consisting of the exon 2 region of the gene encoding HLA-A of the stem cell, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA.

일 구체예에서, 상기 유전적으로 조작하는 단계는 상기 줄기세포의 HLA-A를 코딩하는 유전자의 엑손 2 부위, HLA-B를 코딩하는 유전자의 엑손 2 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 2 부위를 모두 유전적으로 조작하는 것일 수 있다.In one specific example, the genetically manipulating step may be genetically manipulating all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA of the stem cells.

상기 용어 "유전적으로 조작(genetic engineering)" 또는 "유전적으로 조작된(genetically engineered)"은 세포에 대하여 하나 이상의 유전적 변형(genetic modification)을 도입하는 행위 또는 그에 의하여 만들어진 세포를 나타낸다.The term "genetic engineering" or "genetically engineered" refers to the act of introducing one or more genetic modifications into a cell or a cell produced thereby.

또한, 상기 유전적으로 조작은 유전자 편집 시스템에 의해 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자의 염기 서열 내 변형으로 유도되는 것일 수 있다. 상기 유전자 편집 시스템은 예를 들어, 메가뉴클레아제(Meganuclease) 시스템, 징크핑거 뉴클레아제(Zinc finger nuclease) 시스템, 탈렌(TALEN; Transcription Activator-Like Effector Nuclease) 시스템, CRISPR/Cas 시스템 등일 수 있다.Additionally, the genetic manipulation may be induced by a gene editing system to modify the base sequence of a gene encoding one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II. The gene editing system may be, for example, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, or the like.

상기 Cas 단백질은 CRISPR RNA(crRNA) 및 트랜스-활성화 crRNA(trans-activating crRNA, tracrRNA)로 불리는 두 RNA와 복합체를 형성할 때, 활성 엔도뉴클레아제 또는 니카아제(nickase)를 형성할 수 있다. 상기 Cas 단백질의 비제한적인 예는 Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9(Csn1 및 Csx12로도 알려짐), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, 그의 상동체 또는 그의 변형된 버전을 포함할 수 있고, 구체적으로는 Cas9일 수 있다. 이들 효소가 알려져 있으며; 예를 들어, 스트렙토코커스 피오게네스 Cas9 단백질의 아미노산 서열은 수탁 번호 Q99ZW2 하에 스위스프로트(SwissProt) 데이터베이스에서 얻을 수 있다. 일 구체예에서, 비변형 CRISPR 효소, 예를 들어, Cas9는 DNA 절단 활성을 갖는다.The above Cas protein can form an active endonuclease or nickase when it forms a complex with two RNAs called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA). Non-limiting examples of the above Cas proteins include Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, a homolog thereof or a homolog thereof. It may include a modified version, specifically Cas9. These enzymes are known; for example, the amino acid sequence of the Streptococcus pyogenes Cas9 protein is available from the SwissProt database under accession number Q99ZW2. In one embodiment, the unmodified CRISPR enzyme, e.g., Cas9, has DNA cleavage activity.

일 구체예에서, CRISPR 효소는 Cas9 단백질일 수 있고, 상기 Cas9 단백질은 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질, 캄필로박터 제주니(Campylobacter jejuni) 유래 Cas9 단백질, 스트렙토코커스 써모필러스(Streptococcus thermophiles) 유래 Cas9 단백질, 스트렙토코커스 아우레우스(Streptocuccus aureus) 유래 Cas9 단백질 및 네이세리아 메닝기디티스(Neisseria meningitidis) 유래 Cas9 단백질로 이루어진 군에서 선택되는 하나 이상의 Cas9 단백질일 수 있고, 구체적으로 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질일 수 있다. 일 구체예에서, Cas9 단백질은 진핵 세포에서의 발현을 위해 코돈-최적화되며, 상기 스트렙토코커스 피요게네스(Streptococcus pyogenes) 유래 Cas9 단백질을 사용하는 경우, 상기 인간 백혈구 항원 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상 또는 이를 암호화하는 유전자의 발현 또는 활성이 최대로 감소될 수 있다.In one specific example, the CRISPR enzyme may be a Cas9 protein, and the Cas9 protein may be one or more Cas9 proteins selected from the group consisting of a Cas9 protein derived from Streptococcus pyogenes , a Cas9 protein derived from Campylobacter jejuni , a Cas9 protein derived from Streptococcus thermophiles, a Cas9 protein derived from Streptococcus aureus , and a Cas9 protein derived from Neisseria meningitidis , and specifically, a Cas9 protein derived from Streptococcus pyogenes . In one embodiment, the Cas9 protein is codon-optimized for expression in a eukaryotic cell, and when using the Streptococcus pyogenes derived Cas9 protein, the expression or activity of one or more selected from the group consisting of human leukocyte antigen class I and human leukocyte antigen class II or a gene encoding the same can be maximally reduced.

상기 용어 “핵 위치화 서열 또는 신호(Nuclear localization sequence or signal, NLS)"는 특정물질(예컨대, 단백질)을 세포 핵 내로 운반하는 역할을 하는 아미노산 서열을 의미하며, 대체적으로 핵공(Nuclear Pore)을 통하여 세포 핵 내로 운반하는 작용을 한다. 상기 핵 위치화 서열은 진핵 생물에서 CRISPR 복합체 활성에 필요하지 않지만, 이러한 서열을 포함하여, 시스템의 활성을 증진시켜, 특히 핵 내의 핵산 분자를 표적화하는 것으로 여겨진다.The term “nuclear localization sequence or signal (NLS)” refers to an amino acid sequence that functions to transport a specific substance (e.g., a protein) into the cell nucleus, and generally functions to transport it into the cell nucleus through a nuclear pore. Although the nuclear localization sequence is not required for the activity of the CRISPR complex in eukaryotes, it is believed that including such a sequence enhances the activity of the system, particularly targeting nucleic acid molecules in the nucleus.

또한 RNA 유전자 가위(RNA-guided CRISPR)(clustered regularly interspaced short palindrome repeats)-연관된 뉴클레아제 Cas9는 표적 유전자의 넉아웃, 전사 활성화 및 single guide RNA(sgRNA)(즉, crRNA-tracrRNA 융합 전사체)를 이용한 억제에 대한 획기적인 기술을 제공하며, 이 기술은 수많은 유전자 위치를 타겟팅하는 것으로 알려져 있다.Additionally, RNA-guided CRISPR (clustered regularly interspaced short palindrome repeats)-associated nuclease Cas9 offers a breakthrough technology for knockout, transcriptional activation, and repression of target genes using a single guide RNA (sgRNA) (i.e., crRNA-tracrRNA fusion transcript), and this technology is known to target numerous genetic loci.

Cas9 (또는 Cpf1) 단백질은 CRISPR/Cas9 시스템에서 필수적인 단백질 요소를 의미하고, 상기 Cas9 (또는 Cpf1) 유전자 및 단백질의 정보는 국립생명공학정보센터(national center for biotechnology information, NCBI)의 GenBank에서 구할 수 있다. Cas (또는 Cpf1)단백질을 암호화하는 CRISPR - 연관 유전자는 약 40 개 이상의 서로 다른 Cas (또는 Cpf1) 단백질 패밀리가 존재하는 것으로 알려져 있으며, Cas 유전자 및 반복 구조(repeat structure)의 특정 조합에 따라 8개의 CRISPR 하위 유형 (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, 및 Mtube)을 정의할 수 있다. 따라서 상기 각 CRISPR 하위 유형이 반복단위를 이루어 폴리리보뉴클레오티드-단백질 복합체를 형성할 수 있다.The Cas9 (or Cpf1) protein refers to an essential protein component in the CRISPR/Cas9 system, and information on the Cas9 (or Cpf1) gene and protein can be obtained from GenBank of the National Center for Biotechnology Information (NCBI). CRISPR-associated genes encoding Cas (or Cpf1) proteins are known to exist in more than 40 different Cas (or Cpf1) protein families, and eight CRISPR subtypes (Ecoli, Ypest, Nmeni, Dvulg, Tneap, Hmari, Apern, and Mtube) can be defined according to specific combinations of Cas genes and repeat structures. Therefore, each of the above CRISPR subtypes can form a repeat unit to form a polyribonucleotide-protein complex.

상기 인간 백혈구 항체 클래스 I 및 HLA 인간 백혈구 항체 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자 또는 인간 백혈구 항체 클래스 I 및 인간 백혈구 항체 클래스 II로 이루어진 군에서 선택되는 어느 하나 이상의 발현 또는 활성이 감소되도록 인위적으로 수행하는 유전자 조작은 인간 백혈구 항체 클래스 I 및 인간 백혈구 항체 클래스 II로 이루어진 군에서 선택되는 하나 이상이 본래의 기능을 갖는 단백질 형태로 발현되지 않도록 하는 것일 수 있다. 상기 유전자의 조작은 다음 중 하나 이상에 의하여 유도된 것일 수 있다:The genetic manipulation that is artificially performed so that the expression or activity of at least one gene encoding at least one selected from the group consisting of human leukocyte antibody class I and HLA human leukocyte antibody class II is reduced may be such that at least one selected from the group consisting of human leukocyte antibody class I and human leukocyte antibody class II is not expressed in a protein form having its original function. The manipulation of the gene may be induced by at least one of the following:

1) HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 전부 또는 일부 결실, 예컨대, HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 1bp 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드의 결실,1) Deletion of all or part of the gene encoding HLA class I and/or HLA class II, e.g., deletion of 1 bp or more of the nucleotides of the gene encoding HLA class I and/or HLA class II, e.g., deletion of 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide,

2) HLA 클래스 I 및/또는 HLA 클래스 II를 암호화하는 유전자의 1bp 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드의 원래(모세포)와 상이한 뉴클레오티드로의 치환,2) Substitution of 1 bp or more of the nucleotides of the gene encoding HLA class I and/or HLA class II, for example, 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide, with a nucleotide different from the original (parent cell),

3) 하나 이상의 뉴클레오티드, 예컨대, 1 내지 30개, 1 내지 27개, 1 내지 25개, 1 내지 23개, 1 내지 20개, 1 내지 15개, 1 내지 10개, 1 내지 5개, 1 내지 3개, 또는 1개의 뉴클레오티드 (각각 독립적으로 A, T, C 및 G 중에서 선택됨)의 타겟 유전자의 임의의 위치에의 삽입, 및3) insertion of one or more nucleotides, e.g., 1 to 30, 1 to 27, 1 to 25, 1 to 23, 1 to 20, 1 to 15, 1 to 10, 1 to 5, 1 to 3, or 1 nucleotide (each independently selected from A, T, C, and G) into any position of the target gene, and

4) 상기 1) 내지 3) 중에서 선택된 2 가지 이상의 조합.4) A combination of two or more selected from 1) to 3) above.

일 구체예에서, 상기 유전자 편집 시스템은 전기천공법, 유전자총, 초음파천공법, 자기주입법(magnetofection), 일시적인 세포 압축 또는 세포 스퀴징으로 세포 내로 도입될 수 있다.In one embodiment, the gene editing system can be introduced into cells by electroporation, a gene gun, sonoporation, magnetofection, transient cell compression, or cell squeezing.

상기 용어 "도입" 이란, 핵산 및 단백질 복합체를 세포 내로 삽입하는 것일 수 있고, 핵산 및 단백질을 암호화하는 폴리뉴클레오티드를 포함하는 벡터를 세포 내로 삽입하는 것일 수 있다. The term "introduction" above may refer to inserting a nucleic acid and protein complex into a cell, or may refer to inserting a vector comprising a polynucleotide encoding a nucleic acid and a protein into a cell.

일 구체예에 있어서, 상기 유전적으로 조작하는 단계는 서열번호 1의 염기서열로 이루어진 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어진 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어진 폴리뉴클레오티드로 이루어진 군에서 선택되는 하나 이상을 포함하는 CRISPR/Cas 시스템을 줄기세포에 도입하는 것에 의해 수행되는 것일 수 있다.In one specific example, the genetically manipulating step may be performed by introducing into the stem cell a CRISPR/Cas system comprising at least one selected from the group consisting of a polynucleotide comprising a base sequence of SEQ ID NO: 1, a polynucleotide comprising a base sequence of SEQ ID NO: 2, and a polynucleotide comprising a base sequence of SEQ ID NO: 3.

일 구체예에서, 상기 서열번호 1 내지 3의 염기서열로 이루어진 폴리뉴클레오티드를 포함하는 염기서열은 CRISPR/CAS 시스템의 gRNA 역할을 수행할 수 있고, 상기 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상을 암호화하는 유전자를 표적으로 할 수 있다. 구체적으로는, HLA-A를 암호화하는 유전자의 엑손 1 내지 3 부위, HLA-B를 암호화하는 유전자의 엑손 1 내지 3 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 1 내지 3 부위로 이루어진 부위에서 선택되는 하나 이상의 부위를 표적으로 할 수 있으며, 보다 구체적으로는 HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-A를 암호화하는 유전자의 엑손 3 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위, HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 3 부위로 이루어진 부위에서 선택되는 하나 이상의 부위를 표적으로 할 수 있다.In one specific example, a base sequence comprising a polynucleotide consisting of the base sequences of SEQ ID NOS: 1 to 3 can serve as a gRNA of the CRISPR/CAS system and can target a gene encoding one or more selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II. Specifically, one or more regions selected from regions consisting of exons 1 to 3 of the gene encoding HLA-A, exons 1 to 3 of the gene encoding HLA-B, and exons 1 to 3 of the gene encoding HLA-DRA may be targeted, and more specifically, one or more regions selected from regions consisting of exon 2 of the gene encoding HLA-A, exon 3 of the gene encoding HLA-A, exon 2 of the gene encoding HLA-B, exon 3 of the gene encoding HLA-B, exon 2 of the gene encoding HLA-DRA, and exon 3 of the gene encoding HLA-DRA may be targeted.

일 구체예에서, HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 2 부위를 모두 표적으로 할 수 있다. In one specific example, all of the exon 2 region of the gene encoding HLA-A, the exon 2 region of the gene encoding HLA-B, and the exon 2 region of the gene encoding HLA-DRA may be targeted.

일 양상에 따른, 저면역원성의 범용성 유도만능줄기세포는 질환의 치료 목적으로 동종이식(allogeneic transplantation)되는 경우에 면역원성이 저하되어 수혜자의 체내에서 면역 거부반응을 나타내지 않아 이를 원천으로 하는 세포치료제를 개발하는데 유용하게 활용할 수 있다.In one aspect, low immunogenicity, universal induced pluripotent stem cells can be useful in developing cell therapy products using them as a source because they have low immunogenicity and do not cause immune rejection in the recipient's body when used for allogeneic transplantation for the purpose of treating diseases.

도 1은 삼중 HLA 유전자를 넉아웃 시킨 iPSC를 제조하는 방법 및 그 결과를 전체적으로 보여주는 모식도이다.Figure 1 is a schematic diagram showing the overall method for producing iPSCs with triple HLA gene knockout and the results thereof.

도 2는 넉아웃에 사용되는 gRNA의 유효성 평가 결과 및 유전자가 넉아웃 된 것을 확인한 결과와 제조한 iPSC에 IFN γ 자극을 가한 경우, HLA의 유전자와 단백질의 발현 여부를 나타내는 도이다.Figure 2 is a diagram showing the results of evaluating the effectiveness of gRNA used for knockout and confirming that the gene was knocked out, and whether the HLA gene and protein were expressed when IFN γ stimulation was applied to the manufactured iPSC.

도 3은 제조한 iPSC의 형태와 핵형 및 이의 다능성을 확인할 수 있는 실험 결과를 나타내는 도이다.Figure 3 is a diagram showing the results of an experiment that can confirm the morphology and karyotype of the manufactured iPSC and its pluripotency.

도 4는 제조한 iPSC에 IFN γ 자극을 가한 경우, PBMC 또는 NK 세포를 이용한 체외 면역원성 평가를 확인한 실험 결과를 나타내는 도이다.Figure 4 is a diagram showing the results of an experiment confirming the in vitro immunogenicity evaluation using PBMC or NK cells when IFN γ stimulation was applied to manufactured iPSCs.

도 5는 제조한 iPSC의 내피세포로의 분화능 및 HLA 단백질의 발현 여부를 확인한 실험 결과를 나타내는 도이다.Figure 5 is a diagram showing the results of an experiment confirming the differentiation ability of manufactured iPSCs into endothelial cells and the expression of HLA proteins.

이하, 본 발명을 구체적으로 설명하기 위해 실시예를 들어 상세하게 설명하기로 한다. Hereinafter, the present invention will be described in detail by way of examples to specifically explain the present invention.

제조예Manufacturing example

제조예 1. 유도만능줄기세포의 준비Manufacturing Example 1. Preparation of induced pluripotent stem cells

본 출원인의 독자적인 유도만능줄기세포(induced pluripotent stem cell, iPSC)를 준비하였다. 상기 iPSC의 HLA 유형(type) 분석결과는 하기 [표 1]과 같다.The applicant prepared his own induced pluripotent stem cells (iPSCs). The results of HLA type analysis of the iPSCs are shown in [Table 1] below.

LocusLocus Allele 1Allele 1 Allele 2Allele 2 HLA AHLA A 11:01:01:0111:01:01:01 29:01:01:0129:01:01:01 HLA BHLA B 13:02:01:0113:02:01:01 58:01:01:0158:01:01:01 HLA CHLA C 03:02:02:0103:02:02:01 02:02:02:0102:02:02:01 HLA DRB 1HLA DRB 1 07:01:01:0107:01:01:01 15:01:01:0215:01:01:02 HLA DRAHLA DRA 01:02:0101:02:01 01:01:0201:01:02

제조예 2. 삼중 HLA 유전자가 넉아웃 된 iPSC 제조 - g RNA 디자인Manufacturing Example 2. Manufacturing of iPSCs with triple HLA gene knockout - g RNA design

면역 거부반응이 없는 저면역원성 iPSC를 만들기 위해 염색체 6번 내 HLA 유전자가 다른 종류로 이루어진 대립 유전자를 가지고 있는 PBMC(Peripheral blood mononuclear cell, 말초혈액단핵세포) 유래 iPSC인 YiP3를 이용하였다. YiP3 라인의 경우 각각의 대립 유전자에서 HLA-A는 HLA-A 11:01:01:01 와 HLA-A 29:01:01:01를 HLA-B는 HLA-B 13:02:01:01, HLA-B 58:01:01:01를 HLA-C의 경우는 HLA-C 03:02:02 와 HLA-C 02:02:02를 HLA-DRA의 경우는 HLA-DRA 01:02:01, HLA-DRA 01:01:02를 가지고 있었다. 넉아웃 하는 영역으로서 HLA class 1에서 다형성을 나타내는 HLA-A와 HLA-B를 CRISPR/CAS9으로 넉아웃하고 HLA class 2에서 다형성을 나타내는 HLA-DR을 없애기 위해 HLA-DRA를 넉아웃하고 다형성에서 마이너하다고 알려져 있는 HLA-C는 남겨두도록 하는 넉아웃 전략을 수립하였다(도 1a 및 1b). To create hypoimmunogenic iPSCs without immune rejection, we used YiP3, a PBMC (Peripheral Blood Mononuclear Cell)-derived iPSC line that has different alleles of the HLA gene on chromosome 6. In the case of the YiP3 line, each allele of HLA-A had HLA-A 11:01:01:01 and HLA-A 29:01:01:01, HLA-B had HLA-B 13:02:01:01 and HLA-B 58:01:01:01, HLA-C had HLA-C 03:02:02 and HLA-C 02:02:02, and HLA-DRA had HLA-DRA 01:02:01 and HLA-DRA 01:01:02. A knockout strategy was established to knock out HLA-A and HLA-B, which exhibit polymorphism in HLA class 1, using CRISPR/CAS9 as the knockout region, knock out HLA-DRA to eliminate HLA-DR, which exhibits polymorphism in HLA class 2, and leave HLA-C, which is known to be minor in polymorphism (Figures 1a and 1b).

유전자를 넉아웃하는 시스템으로서 CRISPR/CAS9을 이용하였으며, HLA-A 유전자 내 heterogeneous한 영역은 제외한 부분에서 PAM site(NGG)가 존재하고 20 염기쌍이 이중대립유전자로 작용(biallelic) 할 수 있게 디자인하였다. 각각의 유전자에 대해 IPD - IMGT/HLA DATA base를 이용하여 각 대립 유전자를 alignment를 하였고 HLA-A의 경우 엑손 2 내 코돈 37 - 43내에 위치하는 g RNA, G0002 - HLA-A - g1(+, ACAGCGACGCCGCGAGCCAG, PAM: AGG)를 HLA-B의 경우는 엑손 2 내 코돈 31 - 38 내 위치하는 g RNA G0002 - HLA-B - g1(-, GCTGTCGAACCTCACGAACT, PAM: GGG), HLA-DRA의 경우는 엑손 2 내 코돈 36 - 42 내 위치하는 G0002-HLA-DRA-g2(+, TGGCAAAGAAGGAGACGGTC, PAM: TGG)으로 디자인하였다(도 1a 내지 1e).CRISPR/CAS9 was used as a gene knockout system, and the PAM site (NGG) was designed to exist in the part excluding the heterogeneous region in the HLA-A gene and to allow 20 base pairs to function as biallelic genes. For each gene, each allele was aligned using the IPD-IMGT/HLA DATA base, and for HLA-A, the g RNA, G0002-HLA-A-g1 (+, ACAGCGACGCCGCGAGCCAG, PAM: AGG), located within codons 37 to 43 in exon 2, for HLA-B, the g RNA, G0002-HLA-B-g1 (-, GCTGTCGAACCTCACGAACT, PAM: GGG), located within codons 31 to 38 in exon 2, and for HLA-DRA, the g RNA, G0002-HLA-DRA-g2 (+, TGGCAAAGAAGGAGACGGTC, PAM: TGG), located within codons 36 to 42 in exon 2, were designed (Figs. 1a to 1e).

GeneGene Guide RNA sequenceGuide RNA sequence 서열번호Sequence number HLA-AHLA-A ACAGCGACGCCGCGAGCCAGACAGCGACGCCGCGAGCCAG 11 HLA-BHLA-B GCTGTCGAACCTCACGAACTGCTGTCGAACCTCACGAACT 22 HLA-DRAHLA-DRA TGGCAAAGAAGGAGACGGTCTGGCAAAGAAGGAGACGGTC 33

실시예Example

실시예 1. gRNA의 유효성 확인Example 1. Validation of gRNA

상기 제조예 1. 에서 제조한 gRNA의 유효성을 확인하기 위한 실험을 수행하였다. Day 0에는 10000개의 iPSCs와 CRISPR/CAS9 Reagent(RNP complex)를 섞어서 전기 천공법을 실시하였다. 그 다음 이를 플레이트에 시딩하여 37 ℃의 인큐베이터 내에서 배양하였다. Day 3 에는 Flowcytometry sorting를 통하여 트랜스펙션 된 세포를 각각 후보 g RNA 별로 수집하여 총 10000개의 세포를 수집하여 분석을 진행하였다. 트랜스펙션 된 bulk iPSCs는 PCR과 생어 염기서열 분석법으로 분석하였다. 그후 g RNA 유효성 평가를 위해 ICE(interlaced chain reaction) 분석을 진행하였다.An experiment was conducted to confirm the effectiveness of the gRNA manufactured in Manufacturing Example 1 above. On Day 0, 10,000 iPSCs were mixed with CRISPR/CAS9 Reagent (RNP complex) and electroporated. These were then seeded on plates and cultured in an incubator at 37°C. On Day 3, transfected cells were collected for each candidate g RNA through flow cytometry sorting, and a total of 10,000 cells were collected for analysis. The transfected bulk iPSCs were analyzed by PCR and Sanger sequencing. Afterwards, ICE (interlaced chain reaction) analysis was performed to evaluate the effectiveness of g RNA.

실험 결과, HLA-A의 g RNA의 후보 중 G0002-HLA-A-g1가 91 %, HLA-B의 g RNA인 G0002-HLA-B-g 1의 경우 78 %로, HLA-DRA의 경우 G0002-HLA-DRA-g2의 경우 86 %의 효율을 보였다(도 2a 내지 2c).As a result of the experiment, among the candidates for g RNA of HLA-A, G0002-HLA-A-g1 showed an efficiency of 91%, G0002-HLA-B-g 1, which is a g RNA of HLA-B, showed an efficiency of 78%, and G0002-HLA-DRA-g2, which is a g RNA of HLA-DRA, showed an efficiency of 86% (Figs. 2a to 2c).

실시예 2. iPSC의 삼중 HLA 유전자 넉아웃 확인Example 2. Confirmation of triple HLA gene knockout in iPSCs

Day 0에 백만개의 iPSCs와 CRISPR/CAS9 Reagent(RNP complex)를 섞어서 전기 천공법을 실시하였다. 그 전 RNP complex의 조건을 잡기 위해 40 μg each gRNA, 80 μg each gRNA인 두 가지 조건으로 수행한 후 각 유전자의 KO 정도를 ICE 분석을 통해 분석하였다. On Day 0, one million iPSCs were mixed with CRISPR/CAS9 Reagent (RNP complex) and electroporated. To determine the RNP complex conditions, two conditions of 40 μg each gRNA and 80 μg each gRNA were performed, and the degree of KO of each gene was analyzed using ICE analysis.

Day 3 - 5에는 트랜스펙션 된 세포를 수집하여 팩스 분석을 통하여 single cell cloning을 실시하였으며, 약 10000개의 세포를 수집하여 EP(Electrostatic Potential) pool 분석을 실시하였다. Day 5에서 Day 11 - 13 까지는 NGS(Next Generation Sequencing)를 통해 트랜스펙션된 EP pool의 유전자형을 분석하였다. 그 다음 96 웰 플레이트에 시딩하여 single clone을 48개 수집하였다. Day 11 - 13에서 Day 25 - 28에는 수집한 clone을 PCR과 생어 염기서열 분석을 통해 각각의 clone을 스크리닝하고 유전자형 분석을 수행하였다. 이 후 6개의 3중 HLA 유전자가 넉아웃된 clones를 수집하였으며, NGS 분석을 통해 최종 HLA-A, HLA-B, HLA-DRA 유전자가 조작된 세포를 최종적으로 선정하였다.On Days 3-5, transfected cells were collected and single cell cloning was performed through fax analysis. Approximately 10,000 cells were collected and EP (Electrostatic Potential) pool analysis was performed. From Day 5 to Day 11-13, the genotype of the transfected EP pool was analyzed through Next Generation Sequencing (NGS). Then, 48 single clones were collected by seeding in 96-well plates. From Days 11-13 to Days 25-28, each clone was screened and genotyped through PCR and Sanger sequencing. After this, clones with six triple HLA genes knocked out were collected, and cells with the final HLA-A, HLA-B, and HLA-DRA genes manipulated were finally selected through NGS analysis.

3중 HLA 유전자가 넉아웃 된 세포의 유전자형 분석 결과를 추가 시퀀싱 검증(further sequencing verification)을 수행하여 최종적으로 확인하였고, 시퀀싱 검증 결과 HLA-A 영역에서는 Homo type으로 1 bp가 삽입되고 HLA-B 영역에서는 28 bp가 결실되고 HLA-DRA 영역에서는 1 bp의 결실이 발생한 것을 확인할 수 있었다(도 2d 내지 도 2f).Added genotyping results of cells with triple HLA gene knockout Further sequencing verification was performed to confirm the final result, and the sequencing verification results confirmed that 1 bp was inserted as a Homo type in the HLA-A region, 28 bp was deleted in the HLA-B region, and 1 bp was deleted in the HLA-DRA region (Fig. 2d to Fig. 2f).

PCR을 통해 확인하였을 때도 유전적으로 조작한 세포에서 HLA-A, HLA -B, HLA-DRA에 대한 각 유전자의 발현이 감소하는 것을 확인할 수 있었다(도 2g).When confirmed through PCR, it was confirmed that the expression of each gene for HLA-A, HLA-B, and HLA-DRA was reduced in genetically engineered cells (Fig. 2g).

유세포 분석을 통해 iPSC 상태에서 아무 자극 없이 유전자 조작을 진행한 HLA-A, HLA-B, HLA-DR의 발현을 확인한 결과, 단백질 수준에서는 발현하지 않았고, 유전자 조작을 하지 않은 부분인 HLA-C 도 발현하지 않았다. HLA 단백질을 활성화시키기 위해 이틀간 iPSC에 IFNγ 자극을 주었을 때 유전자 조작전인 YiP3의 경우 HLA-A, HLA-B, HLA-C의 단백질의 발현이 증가됨을 확인하였고 HLA-A에서는 99.03 %, HLA-B에서는 91.61 %, HLA-C에서는 88.35 %, HLA-DR에서는 0.04 % 증가함을 보였다. 그에 반해 유전자 조작된 iPSC의 경우 HLA-A는 0.07%, HLA-B는 0.15 %, HLA-DR은 0.02 %로 현저히 감소함을 확인할 수 있었고. 유전자 조작 영역이 아니었던 HLA-C의 경우는 97.34 %로 단백질 발현이 감소되지 않음을 확인할 수 있었다(도 2h). 이는 유전자 조작 결과 HLA-A, HLA-B, HLA-DRA가 선택적으로 넉아웃 되었음을 단백질 수준에서 확인할 수 있음을 나타낸다.Flow cytometry analysis confirmed the expression of HLA-A, HLA-B, and HLA-DR in iPSCs that had been genetically modified without any stimulation. As a result, they were not expressed at the protein level, and HLA-C, which was not genetically modified, was not expressed either. When IFNγ was stimulated in iPSCs for two days to activate HLA proteins, it was confirmed that the protein expression of HLA-A, HLA-B, and HLA-C increased in YiP3 before genetic modification, and HLA-A increased by 99.03%, HLA-B by 91.61%, HLA-C by 88.35%, and HLA-DR by 0.04%. In contrast, in the case of genetically modified iPSCs, HLA-A was significantly reduced by 0.07%, HLA-B by 0.15%, and HLA-DR by 0.02%. In the case of HLA-C, which was not a genetically modified region, protein expression was confirmed to be not reduced by 97.34% (Fig. 2h). This indicates that HLA-A, HLA-B, and HLA-DRA were selectively knocked out as a result of genetic manipulation, and this can be confirmed at the protein level.

실시예 3. 유전자를 조작한 세포의 특성 분석Example 3. Characterization of genetically engineered cells

유전자를 조작한 세포의 특성을 분석하기 위하여 유전자가 조작되기 전인 YiP3과의 비교 실험을 수행하였다.To analyze the characteristics of genetically modified cells, comparative experiments were performed with YiP3 before genetic modification.

형태 및 분화능을 확인하기 위하여 Alkaline Phosphatase (AP) 염색을 수행하였다. iPSCs를 VTN - N 코팅된 6칸 배양 접시에 저밀도로 시딩한 뒤, 콜로니를 형성하게 하기 위해 5일 간 배양 후, 용기는 1 mL의 0.05 % PBST로 세척하고 1 mL의 4 % PFA로 10분 동안 RT에서 고정하였다. 세포를 1 mL의 0.05 % PBST로 한 번 세척한 후 Fast Red Violet, Naphthol AS-BI phosphate 용액 및 TDW(Sigma, SCR004) 이 섞인 1 mL의 염색 용액을 첨가한 뒤 30분 동안 어두운 실온에 방치하였다. 이 후, 세포를 0.05 % PBST로 한 번, 1 x DPBS로 두 번 세척하였다. Alkaline Phosphatase (AP) staining was performed to confirm morphology and differentiation potential. iPSCs were seeded at low density on VTN-N-coated 6-well culture dishes and cultured for 5 days to form colonies. The dishes were washed with 1 mL of 0.05% PBST and fixed with 1 mL of 4% PFA for 10 min at RT. The cells were washed once with 1 mL of 0.05% PBST, and 1 mL of a staining solution containing Fast Red Violet, Naphthol AS-BI phosphate solution, and TDW (Sigma, SCR004) was added and incubated in the dark at room temperature for 30 min. Afterwards, the cells were washed once with 0.05% PBST and twice with 1 × DPBS.

Alkaline Phosphatase(AP) 염색 형태를 분석한 결과, 유전자를 조작한 세포가 콜로니 형상을 띄고 미분화 상태임을 확인하였다(도 3a 및 3b). Analysis of the alkaline phosphatase (AP) staining pattern confirmed that the genetically modified cells had a colony shape and were undifferentiated (Figures 3a and 3b).

RNA 분리는 TRIZOL 시약 (Invitrogen, 15596026)을 사용하여 수행하였다. 분리된 RNA로부터 RevertAid First Strand cDNA 합성 키트 (ThermoFisher, K1622)를 사용하여 cDNA를 합성하였다. qRT-PCR(Quantitative Realtime PCR)은 QuantStudio 3 장비 (Applied Biosystems)를 사용하여 수행하고 Power SYBR Green PCR Master Mix (Applied Biosystems, 4367659)를 사용하였다. mRNA 레벨에서 다능성 마커인 OCT4, SOX2, KLF4, LIN28, NANOG가 발현함을 확인하였고, 내배엽 분화 마커인 SOX17, 중배엽 분화 마커인 BRACHYUARY, 외배엽 분화 마커인 PAX6는 발현하지 않음을 알 수 있었다(도 3c). RNA isolation was performed using TRIZOL reagent (Invitrogen, 15596026). cDNA was synthesized from the isolated RNA using the RevertAid First Strand cDNA synthesis kit (ThermoFisher, K1622). qRT-PCR (Quantitative Real-Time PCR) was performed using a QuantStudio 3 instrument (Applied Biosystems) and Power SYBR Green PCR Master Mix (Applied Biosystems, 4367659) . At the mRNA level, the pluripotency markers OCT4, SOX2, KLF4, LIN28, and NANOG were expressed, while the endoderm differentiation marker SOX17, the mesoderm differentiation marker BRACHYUARY, and the ectoderm differentiation marker PAX6 were not expressed (Fig. 3c).

각 Clone의 단백질 수준에서 다능성 마커 발현하는 정도를 YiP3와 비교하기 위해 유세포 분석을 시행하여 OCT4, SSEA4. NANOG, TRA-1-60의 발현과 Negative marker인 CD34를 비교하였다. FACS 분석을 위해 세포를 1 X TrypLE Express Enzyme를 사용하여 분리하였고, Fixation and Permeabilization 용액 (BD, 554722)으로 20분 동안 4 ℃에서 고정하였다. 1 X Perm/Wash 용액 (BD, 554723)으로 두 번 세척하고, 항체 APC - conjugated HLA-A (BD, 568024), BV510 - conjugated HLA-B(BD, 752615), PE - conjugated HLA-C(BD, 566372), FITC - conjugated HLA-DR (Invitrogen, 11-9956-42), 그리고 각각의 isotype control로 APC - conjugated mouse Ig G (BD, 554681), BV510 - conjugated Ig G (BD, 563039), PE - conjugated mouse Ig G(BD, 555058), FITC - conjugated mouse Ig G (Invitrogen, 11-4732-42)를 4 ℃에서 40분 동안 반응시킨다. 이 후 세포를 1X Perm/Wash 용액으로 두 번 세척한 다음 Attune NxT 기기 (Invitrogen)를 사용하여 분석하였다. To compare the expression of pluripotency markers at the protein level of each clone with YiP3, flow cytometry was performed to compare the expression of OCT4, SSEA4, NANOG, TRA-1-60, and the negative marker CD34. For FACS analysis, cells were dissociated using 1 X TrypLE Express Enzyme and fixed with Fixation and Permeabilization solution (BD, 554722) for 20 minutes at 4°C. Wash twice with 1 X Perm/Wash solution (BD, 554723), and react with antibodies APC - conjugated HLA-A (BD, 568024), BV510 - conjugated HLA-B (BD, 752615), PE - conjugated HLA-C (BD, 566372), FITC - conjugated HLA-DR (Invitrogen, 11-9956-42), and APC - conjugated mouse Ig G (BD, 554681), BV510 - conjugated Ig G (BD, 563039), PE - conjugated mouse Ig G (BD, 555058), FITC - conjugated mouse Ig G (Invitrogen, 11-4732-42) as respective isotype controls at 4 ℃ for 40 minutes. Afterwards, cells were washed twice with 1X Perm/Wash solution and analyzed using an Attune NxT instrument (Invitrogen).

유세포 분석 결과, 유전자를 조작한 세포가 다능성 마커를 YiP3와 유사한 수준으로 발현하는 것을 확인하였다(도 3d 및 3e).Flow cytometry analysis confirmed that the genetically engineered cells expressed pluripotency markers at levels similar to YiP3 (Figures 3d and 3e).

유전자 조작 전 iPSC 와 조작 후 iPSC의 3 배엽 계통으로의 분화 능력을 알아보기 위해 STEMdiff Trilineage 분화 키트(STEMCELL technology, 05230)을 사용하여 분화를 유도하였다. 세포를 1 ug/mL iMatrix-511MG (Nippi, MX892012)로 코팅된 24 웰 플레이트에 2.0 x 105 cells/well(외배엽, 내배엽) 또는 1.0 x 105 cells/well (중배엽)의 밀도로 배양하며, 10 ㎛ Y-27632를 함께 첨가하였다. 외배엽으로의 분화를 위해 매일 배지를 1주일 동안 교환하고, 중배엽이나 내배엽으로 분화를 위해서는, 배지를 매일 5일 동안 교환하였다. 면역 염색을 위해 세포를 4 % 파라포름알데하이드로 고정하고 면역 형광 염색을 실시하였다. To investigate the differentiation capacity of iPSCs before and after genetic manipulation into the three germ layer lineages, differentiation was induced using the STEMdiff Trilineage Differentiation Kit (STEMCELL technology, 05230). Cells were cultured at a density of 2.0 x 105 cells/well (ectoderm, endoderm) or 1.0 x 105 cells/well (mesoderm) in 24-well plates coated with 1 ug/mL iMatrix-511MG (Nippi, MX892012), and 10 μm Y-27632 was also added. For differentiation into ectoderm, the medium was changed daily for 1 week, and for differentiation into mesoderm or endoderm, the medium was changed daily for 5 days. For immunostaining, cells were fixed with 4% paraformaldehyde and immunofluorescence staining was performed.

그 결과 유전자 조작 후 iPSC에서 SOX17, BRACHYURY 및 PAX6의 마커 발현을 확인하였으며, 이로써 유전자 조작된 iPSC의 분화능이 YiP3 와 동일함을 확인하였다(도 3f).As a result, we confirmed the expression of markers SOX17, BRACHYURY, and PAX6 in iPSCs after genetic manipulation, thereby confirming that the differentiation potential of genetically manipulated iPSCs was the same as that of YiP3 (Fig. 3f).

다음은 핵형 분석을 실행하여 YiP3와 비교하여 유전자 조작된 iPSC가 유전자 조작에도 불구하고 수적, 구조적 이상이 없는 정상 핵형을 가지고 있음을 확인하였다(도 3g).Next, we performed karyotype analysis to confirm that the genetically modified iPSCs had a normal karyotype with no numerical or structural abnormalities despite the genetic manipulation compared to YiP3 (Fig. 3g).

실시예 4. 체외(Example 4. In vitro ( in vitroin vitro ) 면역원성 분석) Immunogenicity analysis

또한, 유전자 조작된 iPSC가 면역 회피성을 가지는지 알아보기 위해 YiP3와 HLA Type이 다른 PBMC의 T 세포를 분리하여 공배양을 실시하여 effector memory T cell(TEM)과 central memory T cell(TCM)의 증식 정도를 알아보았다.In addition, to determine whether genetically engineered iPSCs have immune evasion, T cells from PBMCs with different YiP3 and HLA types were isolated and co-cultured to determine the degree of proliferation of effector memory T cells (TEM) and central memory T cells (TCM).

우선 donor로는 YiP3와 HLA 타입이 각 대립 유전자마다 모두 다른 타입을 선별하였고 대립 유전자1 은 HLA-A 02:01, HLA-B 15:01, HLA-C 01:02, HLA-DRB1 11:01, 대립 유전자 2는 HLA-A 02:07, HLA-B 46:01, HLA-C 04:01, HLA-DRB1 15:02을 가지고 있는 donor의 PBMC를 선별하였다(도 4a). 유전자 조작 전인 YiP3 와 조작 후의 iPSC 모두 PBMC와 공배양 전 IFN γ 자극을 이틀동안 준 뒤 배양하였다. HLA 타입이 다른 donor의 PBMC에서 T 세포를 제거한 후 PBMC 내 항원 제시세포가 제시하는 항원에 의해 활성화된 T 세포의 증식을 분석하였다. YiP3, 유전자 조작 후의 iPSC와 T cell이 제거된 PBMC, CFSE - 표지 CD4+ T 세포의 공배양을 통하여 자극을 유도한 후 CD4+ TCM(CD3+CD4+CD45RO+CD62L+)과 CD4+ TEM(CD3+CD4+CD45RO+CD62L-) 세포의 증식 여부를 유세포 분석기로 분석하여 평가하였다. First, donors with different YiP3 and HLA types for each allele were selected, and PBMCs from donors with allele 1 of HLA-A 02:01, HLA-B 15:01, HLA-C 01:02, and HLA-DRB1 11:01 and allele 2 of HLA-A 02:07, HLA-B 46:01, HLA-C 04:01, and HLA-DRB1 15:02 were selected (Fig. 4a). Both YiP3 before genetic manipulation and iPSCs after manipulation were cultured after IFN γ stimulation for two days before co-culture with PBMCs. After removing T cells from PBMCs from donors with different HLA types, the proliferation of T cells activated by antigens presented by antigen-presenting cells in PBMCs was analyzed. After stimulation through co-culture of YiP3, genetically engineered iPSCs and T cell-depleted PBMCs, and CFSE-labeled CD4 + T cells, proliferation of CD4 + TCM (CD3 + CD4 + CD45RO + CD62L + ) and CD4 + TEM (CD3 + CD4 + CD45RO + CD62L - ) cells was evaluated by flow cytometry analysis.

CD4+ T 세포 반응을 평가하기 위하여 면역원성 분석을 수행하였다. PBMCs를 37 걥, 5 % CO2 배양기에서 1일 동안 AIM - V 배지 (Gibco, 12055-083)로 배양하였다. CD4+ T 세포를 분류하기 위해 CD4+ T 세포 분리 키트 (Miltenyi, 130-096-533)를 사용하였다. 수집된 CD4+ T 세포에 0.5 ㎛ CFSE (Invitrogen, C34554)를 첨가하고 암실에서 37 ℃ 배양기에서 10분 동안 배양하였고, 1 X DPBS로 한 번 세척하였다. An immunogenicity assay was performed to evaluate CD4 + T cell responses. PBMCs were cultured in AIM-V medium (Gibco, 12055-083) for 1 day in a 5% CO2 incubator at 37 °C. A CD4 + T cell isolation kit (Miltenyi, 130-096-533) was used to sort CD4 + T cells. 0.5 μm CFSE (Invitrogen, C34554) was added to the collected CD4 + T cells, and they were incubated for 10 minutes in a darkened 37 °C incubator, and washed once with 1 X DPBS.

CD4- PBMCs에서 CD3-/CD4- 세포를 획득하기 위해 CD3 마이크로비드(Miltenyi, 130-050-101)를 사용하였다. 획득한 CD3-/CD4- 세포에 10 ㎍/㎖ Mitomycin C를 넣고 20분 동안 배양한 다음, AIM - V 배지로 세척하였다. CD3-/CD4- PBMC 3.0 x 105 세포와 CFSE로 표지된 CD4+ T, 4 x 105 세포를 24 웰 플레이트에 공동 배양하였다. 직접 동종 항원 자극을 유도하기 위해 각각의 종류의 3 x 103 iPSCs를 각 웰에 첨가하였고 1 ㎎/㎖의 IL - 2를 처리해 37 ℃의 배양기에서 1주일 동안 배양하였다. iPSCs의 면역원성을 확인하기 위해 CFSE로 표지된 CD4+ T 세포의 증식을 유세포 분석기로 분석하였다. 남은 세포에 추가적인 자극으로 7일 동안 CD3-/CD4- PBMCs, 3.0 x 105 세포와 iPSCs, 3.0 x 103 세포를 첨가하여 공배양하였으며 장기적인 면역원성을 평가하기 위해 이를 총 3주 동안 매주 분석하였다.CD3 microbeads (Miltenyi, 130-050-101) were used to obtain CD3 - /CD4 - cells from CD4 - PBMCs. The obtained CD3 - /CD4 - cells were added with 10 μg/mL Mitomycin C and incubated for 20 minutes, then washed with AIM - V medium. CD3 - /CD4 - PBMCs (3.0 x 10 5 cells) and CFSE-labeled CD4 + T cells (4 x 10 5 cells) were co-cultured in 24-well plates. To induce direct alloantigen stimulation, 3 x 10 3 iPSCs of each type were added to each well and cultured in an incubator at 37 °C for 1 week after treatment with 1 mg/mL IL-2. To confirm the immunogenicity of iPSCs, the proliferation of CFSE-labeled CD4 + T cells was analyzed by flow cytometry. As an additional stimulus to the remaining cells, co-culture was performed with CD3 - /CD4 - PBMCs (3.0 x 10 5 cells) and iPSCs (3.0 x 10 3 cells) for 7 days, and analyzed weekly for a total of 3 weeks to assess long-term immunogenicity.

공배양 첫 날 PBMC, CFSE가 표지된 CD4+ 및 TCM과 7일간 공배양 한 후 분석한 결과 유전자 조작을 하지 않은 YiP3의 경우 CD4+ TCM 이 소량 증가하였고 7일째 다시 T 세포 제거 - PBMC와 IFNγ 자극이 된 YiP3와 유전자 조작 후의 iPSC를 자극시킨 후 14일, 21일째 세포를 수집하여 분석하였을 때 CFSE가 표지된 CD4+ TCM은 YiP3와 함께 증가한 반면 HLA 유전자가 조작된 iPSC에서는 YiP3와 비교하여 증가하지 않은 것을 확인할 수 있었다. CFSE가 표지된 CD4+ TEM과의 공배양 결과에서도 유사한 양상을 보였다. 이로써, HLA-A, HLA-B, HLA-DRA 유전자가 조작된 iPSC의 HLA타입이 다른 PBMC 와 공배양 되었을 때 면역원성을 보이지 않는다는 것을 확인할 수 있었다 (도 4b 내지 도 4d). On the first day of co-culture, after co-culture with PBMC, CFSE-labeled CD4 + and TCM for 7 days, analysis showed that in the case of YiP3 without genetic modification, CD4 + TCM increased slightly, and on day 7, T cell depletion was performed again. After stimulating PBMC, IFNγ-stimulated YiP3, and genetically modified iPSCs, cells were collected and analyzed on days 14 and 21. CFSE-labeled CD4 + TCM increased with YiP3, whereas in HLA-modified iPSCs, no increase was observed compared to YiP3. A similar pattern was observed in the co-culture results with CFSE-labeled CD4 + TEM. This confirmed that iPSCs with modified HLA-A, HLA-B, and HLA-DRA genes did not show immunogenicity when co-cultured with PBMCs of different HLA types (Figs. 4b to 4d).

유전자가 조작된 iPSC가 현저하게 낮은 면역원성을 갖는지 여부를 확인하기 위하여 NK 세포 활성화 마커인 CD107a의 발현 정도를 확인하였다.To determine whether genetically engineered iPSCs have significantly lower immunogenicity, the expression level of CD107a, a NK cell activation marker, was examined.

우선, HLA 유형이 상이한 다른 Donor(HLA-A 30:04, HLA-A 02:03, HLA-B 38:02, HLA-B 14:01, HLA-DRB1 15:02, HLA-DRB1 07:01)의 말초혈액단핵세포(도 4e)에서 NK 세포를 분리한 뒤, IL-2로 자극한 후, 유전자가 조작된 iPSC와 공동 배양하였다. 공동 배양 6시간 후, NK 세포의 활성화 마커인 CD107a를 발현하는 세포의 비율을 유세포 분석기로 측정한 결과, CD107a를 발현하는 세포의 비율은 유전적으로 조작되지 않은 iPSC에서 6.71 %인 반면, 유전자가 조작된 iPSC에서는 1.37 %로 확인되었다(도 4f).First, NK cells were isolated from peripheral blood mononuclear cells (Fig. 4e) of different donors with different HLA types (HLA-A 30:04, HLA-A 02:03, HLA-B 38:02, HLA-B 14:01, HLA-DRB1 15:02, HLA-DRB1 07:01), stimulated with IL-2, and co-cultured with genetically engineered iPSCs. After 6 hours of co-culture, the proportion of cells expressing CD107a, an activation marker of NK cells, was measured by flow cytometry. The proportion of cells expressing CD107a was 6.71% in non-genetically engineered iPSCs, whereas it was 1.37% in genetically engineered iPSCs (Fig. 4f).

즉, NK 세포의 활성정도는 유전적으로 조작되지 않은 줄기세포와 비교하여 유전자가 조작된 iPSC에서 현저하게 감소(p < 0.01, 유전적으로 조작되지 않은 줄기세포 대비)한 것을 확인할 수 있다(도 4g).That is, it can be confirmed that the activity level of NK cells was significantly reduced in genetically modified iPSCs compared to non-genetically modified stem cells (p < 0.01, compared to non-genetically modified stem cells) (Fig. 4g).

실시예 5. 분화능 확인 및 웨스턴 블롯Example 5. Confirmation of differentiation potential and Western blot

유전자가 조작된 iPSC의 중배엽 계통인 내피세포로 분화 정도를 확인하기 위하여 분화를 유도하였다. 분화를 유도한 결과, YiP3과 유전자가 조작된 iPSC는 혈 생성 중배엽을 거쳐 내피 세포(EC)로 분화가 되었으며 분화된 형태는 primary EC과 유사한 모양을 띄었다. EC 마커인 CD31과 VE - Cadherin을 모두 발현하는 세포의 population을 유세포 분석으로 확인해 보았을 때 YiP3의 경우 95.69 %, 유전자 조작된 iPSC의 경우 91.9 %로 확인되었다. 이 결과로, 유전자가 조작된 iPSC가 YiP3과 같이 내피 세포로 분화율이 90 % 이상임을 보여주었고 그 분화능에 있어서 이상이 없음을 확인할 수 있었다(도 5a 및 5b). To confirm the degree of differentiation of genetically engineered iPSCs into endothelial cells, a mesodermal lineage, differentiation was induced. As a result of differentiation induction, YiP3 and genetically engineered iPSCs differentiated into endothelial cells (ECs) through hematopoietic mesoderm, and the differentiated morphology was similar to primary ECs. When the population of cells expressing both the EC markers CD31 and VE-Cadherin was confirmed by flow cytometry, it was confirmed to be 95.69% for YiP3 and 91.9% for genetically engineered iPSCs. These results showed that the genetically engineered iPSCs had a differentiation rate of over 90% into endothelial cells, similar to YiP3, and it was confirmed that there was no abnormality in their differentiation potential (Figures 5a and 5b).

iPSC에서 분화된 EC 세포에 IFNγ을 자극 유무에 따른 HLA-A, HLA-B, HLA-C에 대한 단백질 발현을 확인하기 위한 웨스턴 블롯을 수행하였다. Western blot was performed to confirm protein expression of HLA-A, HLA-B, and HLA-C in EC cells differentiated from iPSCs with or without IFNγ stimulation.

세포를 용해하기 위해 1 mL 당 5.0 x 106세포의 RIPA 라이시스 버퍼(Invitrogen, 89901)에 1 X 단백질 분해효소 칵테일 (Invitrogen, 78430)을 첨가하고 냉장한 상태에서 10분 동안 배양하였다. 그 후 13,200 xg에서 20 분간 원심분리한 후, 용해물을 수집하였으며 단백질 농도는 BCA 분석 키트 (Invitrogen, 23227)를 사용하여 측정하였다. 4X LDS sample buffer (Invitrogen, B0007), 10 X reducing agent (Invitrogen, B0009)를 샘플 농도에 따라 세포 용해물에 혼합한 후, 5 분간 가열하고 각 샘플은 4 - 12 % Bis-Tris PAGE gels (Invitrogen, NW04125BOX)에서 전기영동을 통해 분리하였다. 그런 다음, 단백질은 i Blot을 통해 PVDF 멤브레인으로 옮기고, 비특이적 결합을 차단하기 위해 5 % skim milk 로 1시간 동안 blocking하였다. 1차 항체 HLA-A (Abcam, ab52922, 1:5,000), HLA-B (Abcam, ab193415, 1:1,000), HLA-C (Abcam, ab126722, 1:1,000) HLA-DR (Abcam, ab92511, 1:1,000), HLA-DRA (Proteintech, PTG-17221-1-AP, 1:1:000), β - Actin (SantaCruz, sc-47778 HRP, 1:1,000)는 5% skim milk 로 희석하였고, 멤브레인을 4 ℃에서 하룻밤 동안 배양하였다. 멤브레인은 오비탈 쉐이커에서 1X PBS-T 버퍼로 세 번 세척한 후, 1:10,000로 희석한 2차 항체로 1시간 동안 배양하였다. 그런 다음 멤브레인을 세 번 세척한 후, Advansta사의 ECL 시약 (K-12045)를 이용하여 iBright 1500 장비 (Invitrogen)를 사용하여 검출한다. To lyse the cells, 1 X protease cocktail (Invitrogen, 78430) was added to RIPA lysis buffer (Invitrogen, 89901) at 5.0 x 10 6 cells per mL and incubated for 10 minutes in a refrigerated state. After centrifugation at 13,200 x g for 20 minutes, the lysate was collected and the protein concentration was measured using a BCA assay kit (Invitrogen, 23227). 4X LDS sample buffer (Invitrogen, B0007) and 10 X reducing agent (Invitrogen, B0009) were mixed with the cell lysate according to the sample concentration, boiled for 5 minutes, and each sample was separated by electrophoresis on 4-12% Bis-Tris PAGE gels (Invitrogen, NW04125BOX). Then, the proteins were transferred to PVDF membranes via i-Blot and blocked with 5% skim milk for 1 hour to block nonspecific binding. Primary antibodies HLA-A (Abcam, ab52922, 1:5,000), HLA-B (Abcam, ab193415, 1:1,000), HLA-C (Abcam, ab126722, 1:1,000), HLA-DR (Abcam, ab92511, 1:1,000), HLA-DRA (Proteintech, PTG-17221-1-AP, 1:1:000), β-Actin (SantaCruz, sc-47778 HRP, 1:1,000) were diluted in 5% skim milk, and the membranes were incubated overnight at 4°C. The membrane was washed three times with 1X PBS-T buffer on an orbital shaker and then incubated with secondary antibody diluted 1:10,000 for 1 hour. The membrane was then washed three times and detected using Advansta ECL reagent (K-12045) on an iBright 1500 instrument (Invitrogen).

웨스턴 블롯 결과 IFNγ을 자극이 없을 때는 HLA-A, HLA-B, HLA-C의 단백질이 발현하지 않았고 IFNγ을 자극이 있을 때는 유전자가 조작되지 않은 YiP3에서는 HLA-A, HLA-B 단백질이 발현하고, 유전자가 조작된 iPSC의 경우, HLA-A, HLA-B 단백질이 발현하지 않았다. 그리고 유전자 조작을 하지 않은 HLA-C에 대해서는 YiP3와 함께 유전자가 조작된 iPSC에서도 그대로 존재함을 확인할 수 있었다(도 5c).Western blot results showed that when there was no IFNγ stimulation, HLA-A, HLA-B, and HLA-C proteins were not expressed, but when there was IFNγ stimulation, HLA-A and HLA-B proteins were expressed in YiP3 that had not been genetically modified, and in the case of genetically modified iPSCs, HLA-A and HLA-B proteins were not expressed. In addition, it was confirmed that HLA-C, which had not been genetically modified, was present in both YiP3 and genetically modified iPSCs (Fig. 5c).

이 결과로서 HLA-A, HLA-B, HLA-DRA가 KO된 iPSC가 EC로 분화하였을 때 EC 마커를 발현하는 세포 분화할 수 있고, HLA-A, HLA-B의 단백질을 발현하지 않음을 확인할 수 있었다. 참고적으로 HLA-DRA의 단백질은 분화된 EC 세포에서는 확인할 수 없었다. As a result, it was confirmed that when iPSCs with HLA-A, HLA-B, and HLA-DRA knockout were differentiated into ECs, they could differentiate into cells expressing EC markers and did not express HLA-A and HLA-B proteins. For reference, HLA-DRA protein could not be confirmed in differentiated EC cells.

Claims (13)

모세포에 비하여 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 유전적으로 조작된 저면역원성 줄기세포.A hypoimmunogenic stem cell genetically engineered to have reduced expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II compared to the parent cell. 청구항 1에 있어서, 상기 인간 백혈구 항원 클래스 I은 HLA-A, HLA-B 및 HLA-C로 이루어진 군에서 선택되는 하나 이상인, 줄기세포.A stem cell according to claim 1, wherein the human leukocyte antigen class I is at least one selected from the group consisting of HLA-A, HLA-B, and HLA-C. 청구항 1에 있어서, 상기 인간 백혈구 항원 클래스 II는 HLA-DR, HLA-DP 및 HLA-DQ로 이루어진 군에서 선택되는 하나 이상인, 줄기세포.A stem cell according to claim 1, wherein the human leukocyte antigen class II is at least one selected from the group consisting of HLA-DR, HLA-DP, and HLA-DQ. 청구항 1에 있어서, 상기 줄기세포는 HLA-A를 코딩하는 유전자의 엑손 부위, HLA-B를 코딩하는 유전자의 엑손 부위 및 HLA-DRA를 코딩하는 유전자의 엑손 부위로 이루어진 군에서 선택되는 하나 이상의 부위가 유전적으로 조작된 것인, 줄기세포.In claim 1, the stem cell is a stem cell in which at least one region selected from the group consisting of an exon region of a gene encoding HLA-A, an exon region of a gene encoding HLA-B, and an exon region of a gene encoding HLA-DRA is genetically engineered. 청구항 4 있어서, 상기 줄기세포는 HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-A를 암호화하는 유전자의 엑손 3 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위, HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위가 유전적으로 조작된 것인, 줄기세포.In claim 4, the stem cell is a stem cell in which at least one region selected from the group consisting of an exon 2 region of a gene encoding HLA-A, an exon 3 region of a gene encoding HLA-A, an exon 2 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-B, an exon 3 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-DRA, and an exon 3 region of a gene encoding HLA-DRA is genetically engineered. 청구항 1에 있어서, 상기 줄기세포는 RNA 간섭(RNAi; RNA interference) 시스템, 메가뉴클레아제(Meganuclease) 시스템, 징크핑거 뉴클레아제(Zinc finger nuclease) 시스템, 탈렌(TALEN; Transcription Activator-Like Effector Nuclease) 시스템, CRISPR/Cas 시스템, X-선 조사, 감마선 조사, 에틸 메탄설포네이트(ethyl methanesulfonate) 처리 또는 다이메틸 설페이트(dimethyl sulfate) 처리를 통해 유전적으로 조작된 것인, 줄기세포.A stem cell according to claim 1, wherein the stem cell is genetically engineered through an RNA interference (RNAi) system, a meganuclease system, a zinc finger nuclease system, a TALEN (Transcription Activator-Like Effector Nuclease) system, a CRISPR/Cas system, X-ray irradiation, gamma ray irradiation, ethyl methanesulfonate treatment, or dimethyl sulfate treatment. 청구항 6에 있어서, 상기 CRISPR/Cas 시스템은 서열번호 1의 염기 서열로 이루어진 폴리뉴클레오티드, 서열번호 2의 염기서열로 이루어진 폴리뉴클레오티드 및 서열번호 3의 염기서열로 이루어진 폴리뉴클레오티드로 이루어진 군에서 선택되는 하나 이상의 폴리뉴클레오티드를 포함하는 것인, 줄기세포.A stem cell according to claim 6, wherein the CRISPR/Cas system comprises at least one polynucleotide selected from the group consisting of a polynucleotide consisting of a base sequence of sequence number 1, a polynucleotide consisting of a base sequence of sequence number 2, and a polynucleotide consisting of a base sequence of sequence number 3. 청구항 1에 있어서, 상기 줄기세포는 유도만능 줄기세포인, 줄기세포.In claim 1, the stem cell is an induced pluripotent stem cell. 청구항 1에 있어서, 상기 줄기세포는 제대, 제대혈, 혈액, 골수, 지방, 근육, 신경, 피부, 양막, 양수 및 태반으로 이루어진 군에서 선택되는 하나 이상의 유래인, 줄기세포.In claim 1, the stem cell is a stem cell derived from one or more selected from the group consisting of umbilical cord, umbilical cord blood, blood, bone marrow, fat, muscle, nerve, skin, amniotic membrane, amniotic fluid, and placenta. 모세포에 비하여 인간 백혈구 항원(Human Leukocyte Antigen, HLA) 클래스 I 및 인간 백혈구 항원 클래스 II로 이루어진 군에서 선택되는 하나 이상의 인간 백혈구 항원의 발현이 감소되도록 줄기세포를 유전적으로 조작하는 단계를 포함하는 저면역원성 줄기세포의 제조 방법.A method for producing hypoimmunogenic stem cells, comprising the step of genetically engineering stem cells so that the expression of one or more human leukocyte antigens selected from the group consisting of human leukocyte antigen (HLA) class I and human leukocyte antigen class II is reduced compared to parent cells. 청구항 10 있어서, 상기 유전적으로 조작하는 단계는 상기 줄기세포의 HLA-A를 암호화하는 유전자의 엑손 부위, HLA-B를 암호화하는 유전자의 엑손 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 유전적으로 조작하는 것인, 방법.A method according to claim 10, wherein the genetically manipulating step genetically manipulates at least one region selected from the group consisting of an exon region of a gene encoding HLA-A, an exon region of a gene encoding HLA-B, and an exon region of a gene encoding HLA-DRA of the stem cell. 청구항 11에 있어서, 상기 유전적으로 조작하는 단계는 상기 줄기세포의 HLA-A를 암호화하는 유전자의 엑손 2 부위, HLA-A를 암호화하는 유전자의 엑손 3 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 2 부위, HLA-B를 암호화하는 유전자의 엑손 3 부위, HLA-DRA를 암호화하는 유전자의 엑손 2 부위 및 HLA-DRA를 암호화하는 유전자의 엑손 3 부위로 이루어진 군에서 선택되는 하나 이상의 부위를 유전적으로 조작하는 것인, 방법.A method according to claim 11, wherein the genetically manipulating step genetically manipulates at least one region selected from the group consisting of an exon 2 region of a gene encoding HLA-A, an exon 3 region of a gene encoding HLA-A, an exon 2 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-B, an exon 3 region of a gene encoding HLA-B, an exon 2 region of a gene encoding HLA-DRA, and an exon 3 region of a gene encoding HLA-DRA. 청구항 10에 있어서, 상기 유전적으로 조작하는 단계는 서열번호 1의 염기 서열로 이루어진 폴리뉴클레오티드, 서열번호 2의 염기 서열로 이루어진 폴리뉴클레오티드 및 서열번호 3의 염기 서열로 이루어진 폴리뉴클레오티드로 이루어진 군에서 선택되는 하나 이상을 포함하는 CRISPR/Cas 시스템을 줄기세포에 도입하는 것에 의해 수행되는 것인, 방법. A method according to claim 10, wherein the genetically manipulating step is performed by introducing into a stem cell a CRISPR/Cas system comprising at least one selected from the group consisting of a polynucleotide consisting of a base sequence of SEQ ID NO: 1, a polynucleotide consisting of a base sequence of SEQ ID NO: 2, and a polynucleotide consisting of a base sequence of SEQ ID NO: 3.
PCT/KR2025/003036 2024-04-22 2025-03-07 Hypoimmunogenic universal induced pluripotent stem cells Pending WO2025225866A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2024-0053682 2024-04-22
KR1020240053682A KR20250155134A (en) 2024-04-22 2024-04-22 Hypoimmunogenic universal induced pluripotent stem cells

Publications (1)

Publication Number Publication Date
WO2025225866A1 true WO2025225866A1 (en) 2025-10-30

Family

ID=97490674

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2025/003036 Pending WO2025225866A1 (en) 2024-04-22 2025-03-07 Hypoimmunogenic universal induced pluripotent stem cells

Country Status (2)

Country Link
KR (1) KR20250155134A (en)
WO (1) WO2025225866A1 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102459911B1 (en) 2020-12-11 2022-10-28 한국한의학연구원 Composition for inducing selective apoptosis undifferentiated human induced pluripotent stem cells comprising Phellodendri Cortex

Also Published As

Publication number Publication date
KR20250155134A (en) 2025-10-30

Similar Documents

Publication Publication Date Title
JP7590015B2 (en) Method for producing low antigenic cells
US11197935B2 (en) Talen targeting blood coagulation factor VIII intron 1 inversion gene and composition for treating hemophilia comprising same
US20230272429A1 (en) Modification of blood type antigens
WO2016021972A1 (en) Immune-compatible cells created by nuclease-mediated editing of genes encoding hla
US20240060047A1 (en) Cells with sustained transgene expression
TW202242101A (en) Compositions and methods for genetically modifying ciita in a cell
WO2020047300A1 (en) Nucleic acid constructs comprising gene editing multi-sites and uses thereof
WO2018030630A1 (en) Nonviral minicircle vector carrying sox gene and construction method therefor
WO2025225866A1 (en) Hypoimmunogenic universal induced pluripotent stem cells
CA3125629A1 (en) Genetically modified cells, tissues, and organs for treating disease
KR101658135B1 (en) Endo-Nucleases Targeting the Blood Coagulation Factor VIII Gene and Uses Thereof
WO2023167575A1 (en) Low immunogenic stem cells, low immunogenic cells differentiated or derived from stem cells, and production method therefor
Vassena et al. Accumulation of instability in serial differentiation and reprogramming of parthenogenetic human cells
WO2023008918A1 (en) Genetically modified stem cell having inhibited b2m gene expression, and method for using same
US20230242894A1 (en) Novel transplantation cells having reduced immunogenicity
TW202532430A (en) Cell therapy
EP4112720A1 (en) Genetically modified megakaryocyte, modified platelet, and methods respectively for producing said genetically modified megakaryocyte and said modified platelet
WO2025008763A1 (en) Modified cell
CN116804185A (en) Universal cell and preparation method thereof
Vassena et al. HMG Advance Access published April 30, 2012

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: 25794866

Country of ref document: EP

Kind code of ref document: A1