WO2021037234A1 - Method for efficiently repairing gene mutation of ring sideroblastic anemia - Google Patents

Method for efficiently repairing gene mutation of ring sideroblastic anemia Download PDF

Info

Publication number
WO2021037234A1
WO2021037234A1 PCT/CN2020/112227 CN2020112227W WO2021037234A1 WO 2021037234 A1 WO2021037234 A1 WO 2021037234A1 CN 2020112227 W CN2020112227 W CN 2020112227W WO 2021037234 A1 WO2021037234 A1 WO 2021037234A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
gene
alas
sgrna
cells
Prior art date
Application number
PCT/CN2020/112227
Other languages
French (fr)
Chinese (zh)
Inventor
方日国
袁鹏飞
张英驰
杨卉慧
于玲玲
Original Assignee
广州辑因医疗科技有限公司
中国医学科学院血液病医院(中国医学科学院血液学研究所)
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 广州辑因医疗科技有限公司, 中国医学科学院血液病医院(中国医学科学院血液学研究所) filed Critical 广州辑因医疗科技有限公司
Priority to CN202080056107.1A priority Critical patent/CN114222816A/en
Publication of WO2021037234A1 publication Critical patent/WO2021037234A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses

Definitions

  • the present invention relates to the field of gene editing therapy. Specifically, the present invention relates to a method for efficiently repairing gene mutations that cause ring sideroblastic anemia by using gene editing technology, which includes using gene editing technology to efficiently and safely genetically modify human hematopoiesis
  • the specific point mutation of the ALAS-2 gene of the stem cells restores the expression of the ALAS-2 gene and achieves the purpose of treating diseases.
  • Hereditary sideroblastic anemia (Congenital sideroblastic anemia, CSA) is a group of inherited diseases of iron utilization disorders. It is characterized by the appearance of a large number of ring sideroblasts in the bone marrow, ineffective production of red blood cells, excessive iron reserves in the tissues, and small cell hypochromic anemia in the peripheral blood. At present, molecular level testing has found 7 disease mutations, which mainly lead to iron synthesis, iron-sulfur complex, and mitochondrial protein synthesis disorders. Among them, X-chain sideroblastic anemia is the most common type of disease (Kaneko K, et al. Haematologica. 2014).
  • X-linked sideroblastic anemia is a rare genetic disease of the blood system, which is a kind of CSA with insufficient incidence
  • ALAS-2 is a key regulator that regulates the synthesis of catalyzed heme in red blood cells.
  • the treatment plan of gene editing therapy is to use gene editing tools, such as CRISPR/Cas9, zinc finger nuclease (Zinc Finer Nulease, ZFN), and transcription activator-like effector nuclease (transcription activator-like effector nucleases, TALEN), etc.
  • Gene editing tools such as CRISPR/Cas9, zinc finger nuclease (Zinc Finer Nulease, ZFN), and transcription activator-like effector nuclease (transcription activator-like effector nucleases, TALEN), etc. Edit the patient’s autologous hematopoietic stem cells, repair the ALAS-2 gene mutation to restore the expression of the ALAS-2 gene, and then return the genetically modified autologous hematopoietic stem cells to the patient to make the patient’s heme synthesis and red blood cell status Return to normal level to achieve the purpose of curing diseases.
  • Gene editing technology is a genetic recombination technology that artificially uses encoded nucleases to insert, knock out, and mutate at specific sites in the DNA sequence to change the gene sequence.
  • Gene editing tools will first identify specific sequences in the genome, generate DNA double-strand break gaps through nucleases, and rely on endogenous repair mechanisms.
  • Non-homologous end joining (NHEJ) and homologous recombination (Homology-directed) repair, HDR) two repair mechanisms to repair.
  • the former introduces insertions and deletions (INDELs) into the genome through cell replication and repair, resulting in mutations; the latter precisely repairs the genome sequence due to the addition of foreign donor nucleic acid as a template (Dever, et al. Nature). .2016).
  • CRISPR/Cas9 has the advantages of simple operation, low cost, and large development space, which greatly improves the operability and work efficiency of gene editing (Cong, et al. Science. 2013; Jinek, et al. .Science.2012).
  • the present invention uses gene editing technology for the first time, such as CRISPR/Cas9 gene editing technology, to develop a new generation of hematopoietic stem cells, and successfully and efficiently repair the point mutation of the ALAS-2 gene in the hematopoietic stem cells derived from the bone marrow of XLSA patients.
  • the gene repair efficiency is as high as approx. 30%-40%, the ALAS-2 gene expression of the gene repaired cells reaches about 50% of the ALAS-2 expression of healthy donors, thereby promoting the differentiation of hematopoietic stem cells into mature red blood cells.
  • the present application provides a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations in stem cell chromosomes through gene editing, wherein the gene editing includes:
  • the present application provides a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2, wherein the gene editing includes:
  • the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing functional ALAS-2 expression, thereby increasing heme production in cells derived from the hematopoietic stem cells, wherein
  • the gene editing includes:
  • the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing functional ALAS-2 expression, thereby increasing heme production in cells derived from the hematopoietic stem cells, thereby promoting The method for maturation of hematopoietic stem cells, wherein the gene editing includes:
  • the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of hemoglobin in the cells derived from the hematopoietic stem cells, and promoting the A method for the maturation of hematopoietic stem cells to treat anemia (such as XLSA) in an individual, wherein the gene editing includes:
  • the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing and increasing functional ALAS-2 expression, wherein said gene editing does not cause off-target or off-target rate in the genome of said hematopoietic stem cells Less than 1%, such as less than 0.5% or less than 0.1%, wherein the gene editing includes:
  • the hematopoietic stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
  • HSPC CD34 + hematopoietic stem and progenitor cells
  • hiPSC human induced pluripotent stem cells
  • the hematopoietic stem cells are obtained from anemia patients, such as sideroblastic anemia patients, specifically, hereditary sideroblastic anemia patients, more specifically, XLSA patients.
  • the mutation is located in exon 5-11 or intron-1 of the ALAS-2 gene.
  • the mutation is located in intron-1 of the ALAS-2 gene.
  • the mutation is Int-1-GATA.
  • sequence-specific nuclease is selected from the group consisting of RNA guide nuclease, zinc finger nuclease (ZFN), and transcription activator-like effector nuclease (TALEN).
  • the sequence-specific nuclease is an RNA guide nuclease.
  • the RNA guide nuclease is Cas.
  • the RNA guide nuclease is Cas9.
  • it further comprises introducing a guide RNA (sgRNA) that recognizes the ALAS-2 gene into the CD34 + HSPC.
  • sgRNA guide RNA
  • the nuclease cleavage site is not more than about 20 nucleotides away from the mutation site, for example, about 15, 13, 12, 11, or about 10 nucleotides. About 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, about 1 nucleotide, or nuclease cleavage site and the The mutation sites overlap.
  • the sgRNA is complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome.
  • the guide sequences in the sgRNA are about 10 to about 25, about 12 to about 24, about 14 to about 23, about 16 to about 22, about 17 to about 21 nucleotides long. In some specific embodiments of the above method, the guide sequence in the sgRNA is 20 nucleotides long.
  • the sgRNA is chemically modified.
  • the sgRNA is modified by nucleotide ribose 2'-O-methylation and/or internucleotide 3'phosphorothioate modification (also referred to as phosphorothioate modification) .
  • nucleotide ribose 2'-O-methylation and/or internucleotide 3'phosphorothioate modification also referred to as phosphorothioate modification
  • one, two and/or three bases before the 5'end of the sgRNA and/or the last nucleotide ribose base at the 3'end are 2'-O-methylated modifications.
  • the sgRNA comprises the first 3 nucleotides at the 5'end and the 3 nucleotides after the 3'end.
  • the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end The connection between the 3 nucleotides after the 3'end contains phosphorothioate modification.
  • the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end
  • the last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
  • the sgRNA comprises a nucleic acid sequence complementary to a sequence in intron-1 of the ALAS-2 gene.
  • the nucleic acid sequence that is complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA is selected from the following group: SEQ ID NO: 1-SEQ ID NO: 3.
  • the sgRNA is introduced into the hematopoietic stem cells by electroporation and transduction.
  • the donor DNA is circular.
  • the donor DNA is linear.
  • the donor DNA is ssODN.
  • the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end. .
  • the donor DNA includes a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, And the first three nucleotides at the 5'end and the last three nucleotides at the 3'end also contain 2'-O-methylation modification of ribose.
  • the correction sequence has the same length as the mutation sequence. In some embodiments of the above methods, the donor DNA and the correction sequence are equal in length. In some embodiments of the above methods, the donor DNA is longer than the correction sequence.
  • the correction sequence is about 50 to about 300, about 60 to about 250, about 60 to about 240, about 60 to about 230, about 60 to about About 220, about 60 to about 210, about 60 to about 200 nucleotides long.
  • the correction sequence comprises a 5'arm that is substantially complementary to a target region located at the 3'end of the mutation site, and a 5'arm that is substantially complementary to a target region located at the 5'end of the mutation site. Complementary 3'arms on top.
  • the 5'arm or 3'arm of the correction sequence has at least about 85% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively, at least about 85%. 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% homology.
  • the 5'arm or 3'arm of the correction sequence has 100% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively.
  • the 5'arms of the correction sequence are about 30 to about 100 nucleotides long, for example, about 35 to about 80, about 40 to about 70, about 40. From about 60 nucleotides in length.
  • the 3'arms of the correction sequence are about 20 to about 100 nucleotides long, for example, about 20 to about 80, about 20 to about 70, about 20. From about 20 to about 50 nucleotides in length.
  • the 5'arm of the correction sequence is longer than the 3'arm of the correction sequence.
  • the 3'arm of the correction sequence is longer than the 5'arm of the correction sequence.
  • the 5'arm of the correction sequence and the 3'arm of the correction sequence have the same length.
  • the correction sequence is complementary to the target sequence at ChrX:55028172-55028268 except for the mutation site.
  • the correction sequence when the correction sequence includes a coding sequence, it encodes an amino acid sequence that is the same as the amino acid sequence encoded by the mutation sequence except for the mutation site.
  • the correction sequence corresponding to the mutation is SEQ ID NO: 4.
  • the donor DNA is introduced into the hematopoietic stem cells by electroporation.
  • introducing the sequence-specific nuclease includes introducing mRNA encoding the sequence-specific nuclease into stem cells.
  • the mRNA encoding the sequence-specific nuclease is introduced into the hematopoietic stem cell by electroporation.
  • the mRNA encoding the sequence-specific nuclease and the donor DNA are simultaneously introduced into the stem cell.
  • sgRNA is introduced into stem cells, and wherein the mRNA encoding the sequence-specific nuclease and the sgRNA are simultaneously introduced into the stem cells.
  • the sgRNA and the donor DNA are simultaneously introduced into the stem cell.
  • the sgRNA, the mRNA encoding the sequence-specific nuclease, and the donor DNA are separately or simultaneously introduced into the stem cells by means of electroporation or transduction.
  • the weight ratio of the sgRNA to the donor DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11:1, about 1:10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1.
  • the weight ratio of the mRNA encoding the sequence-specific nuclease to the single-stranded DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11:1 , About 1:10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1 .
  • the hematopoietic stem cells are obtained from a male individual.
  • the CD34 + HSPC is obtained from a male individual.
  • the human induced pluripotent stem cells are obtained from male individuals.
  • the hematopoietic stem cells are obtained from a female individual.
  • the CD34 + HSPC is obtained from a female individual.
  • the human induced pluripotent stem cells are obtained from a female individual.
  • This application also relates to a gene-edited CD34 + HSPC or human induced pluripotent stem cell (hiPSC) obtained by the above method, wherein the CD34 + HSPC or human induced pluripotent stem cell (hiPSC) is derived from an anemia patient and has been gene-edited,
  • the mutation of the ALAS-2 gene has been corrected.
  • the anemia is sideroblast anemia, such as hereditary sideroblast anemia, specifically, XLSA.
  • the ALAS-2 gene mutation is located in exon 5-11 or intron-1 of the ALAS-2 gene.
  • the mutation is Int-1-GATA.
  • a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations of hematopoietic stem cells by CRISPS/Cas9 gene editing comprising: adding a single-stranded correction sequence corresponding to the ALAS-2 mutation sequence
  • the donor DNA, the sgRNA that recognizes the ALAS-2 mutation sequence, and the nucleic acid sequence encoding the Cas9 protein are introduced into the hematopoietic stem cell, whereby the correction sequence in the donor DNA replaces the ALAS-2 mutation sequence in the hematopoietic stem cell.
  • ALAS-2 mutant sequence is a mutant sequence in exons 5-11 of the ALAS-2 gene and/or a mutant sequence in intron-1 of the ALAS-2 gene.
  • sequence of the sgRNA is selected from the following group: SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3.
  • correction sequence comprises a 5'arm complementary to the target region 3'of the mutation site, and a 3'arm complementary to the target region 5'of the mutation site.
  • 'Arm wherein the 5'arm of the calibration sequence is about 40 to about 60 nucleotides long, and the 3'arm of the calibration sequence is about 20 to about 50 nucleotides long.
  • Figure 1 Point mutations in intron-1 of ALAS-2 gene on human X chromosome (X:55054635[ChrX(GRCh37/hg19):g.55054635A>G,NM 000032.4:c.-15-2187T> C) Schematic diagram of multiple sgRNAs and donor template single-stranded DNA designed at nearby locations.
  • Figure 2 Point mutations in intron-1 of ALAS-2 gene on human X chromosome (X:55054635 [Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15-2187 T> C) Sequence information of multiple sgRNAs and donor template single-stranded DNA designed at nearby locations.
  • Electrotransform Cas9 mRNA, sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN enter the hiPSCs derived from XLSA patients according to the addition amount of Cas9, sgRNA and ssODN, and expand after 4 days Increase the target fragment and NGS, and analyze the ratio of NHEJ and HDR through bioinformatics methods.
  • NHEJ non-homologous end joining, representing the ratio of indels
  • HDR Homology-directed repair, representing the ratio of gene repair
  • n 3 experimental replicates.
  • FIG. 8 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+HSPC derived from the bone marrow of XLSA patients for 2 days, and in vitro clone formation experiment (CFU detection), 14 days later Count the number of clones in different blood systems, BFU-E, CFU-GM, CFU-E, CFU-GEMM represent the formation of clones of different blood system lineages such as erythroid, myeloid, and lymphatic systems.
  • healthy donors represent healthy donors that have not undergone gene editing
  • blank control represent cells that have not undergone gene editing
  • gene repair represent cells that have undergone gene repair
  • n 3 experimental replicates.
  • Figure 9 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. The 7th day after differentiation was detected. The expression ratio of human CD71 and human CD235a membrane proteins on the 13th and 18th days represents the efficiency of erythroid differentiation.
  • healthy donors represent healthy donors that have not undergone gene editing
  • blank control represent cells that have not undergone gene editing
  • gene repair represent cells that have undergone gene repair.
  • Figure 10 Electrotransformation of Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from bone marrow of XLSA patients for red blood cell differentiation. 18 days later, Figure A is the post-differentiation Photograph of the posterior cell.
  • Figure B is a schematic diagram of Benzidine staining bright field.
  • Panel C is a schematic diagram of Wright-Giemsa staining bright field. Ruler: 20um.
  • healthy donors represent healthy donors that have not undergone gene editing
  • blank control represent cells that have not undergone gene editing
  • gene repair represent cells that have undergone gene repair.
  • FIG. 11 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. 18 days later, perform Benzidine staining and statistics Analyze the proportion of Benzidine positive.
  • healthy donors represent healthy donors that have not undergone gene editing
  • blank control represent cells that have not undergone gene editing
  • gene repair represent cells that have undergone gene repair
  • n 3 experimental replicates.
  • FIG. 12 Electrotransform Cas9 mRNA, sgRNA-1 near the point mutation of ALAS-2 intron-1 and donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation, detected after 18 days, and quantified by fluorescence PCR was used to detect the mRNA expression of ALAS-2, GATA-1 and GAPDH genes.
  • healthy donors represent healthy donors that have not undergone gene editing
  • blank control represent cells that have not undergone gene editing
  • gene repair represent cells that have undergone gene repair
  • n 3 experimental replicates.
  • ALAS-2 gene and GATA-1 were normalized with GAPDH and healthy donors.
  • FIG. 13 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. After 18 days, the test was performed by Western Blot experiment. Detect the protein level expression of ALAS-2, GATA-1 and GAPDH genes. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, and gene repair: represent cells that have undergone gene repair.
  • FIG. 14 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+HSPC derived from the bone marrow of XLSA patients.
  • the transplantation has undergone gene repair and has not undergone gene editing.
  • the cells from the irradiator enter the 6-week-old NPG immunodeficiency mouse model. After 10 weeks, 12 weeks, and 16 weeks, the proportion of human CD45-positive cells in the peripheral blood of the mouse is detected. At the same time, the proportion of human CD45 positive cells is detected in the mouse after 16 weeks of transplantation.
  • the proportion of CD45 positive cells is calculated as human CD45 positive cells%/(human CD45 positive cells%+mouse CD45 positive cells%), human CD45 positive cells% and small
  • the percentages of mouse CD45 positive cells were measured by flow cytometry experiments.
  • Blank control represents cells that have not undergone gene editing
  • gene repair represents cells that have undergone gene repair.
  • n 6 mice.
  • FIG. 15 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients.
  • the transplantation has undergone gene repair and has not undergone gene editing.
  • the cells entered into a 6-week-old NPG immunodeficiency mouse model irradiated with an irradiator. After 16 weeks, the ratio of human cell membrane proteins such as CD3, CD33, CD56, and CD19 to human CD45 protein was detected in the mouse bone marrow and spleen.
  • Blank control represents cells that have not undergone gene editing
  • gene repair represents cells that have undergone gene repair.
  • n 6 mice.
  • FIG. 16 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. The cells entered the 6-week-old NPG immunodeficiency mouse model irradiated by the irradiator. After 16 weeks, flow cytometry analysis of human CD45 in bone marrow, spleen and peripheral blood of 1 mouse in the blank control group and the gene repair group The proportion of positive cells.
  • FIG. 17 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients.
  • the transplantation has undergone gene repair and has not undergone gene editing.
  • the cells enter the 6-week-old NPG immunodeficiency mouse model that has been irradiated by the irradiator. After 16 weeks, flow cytometry analysis of CD3, CD33, and CD56 in the bone marrow and spleen of 1 mouse in the blank control group and the gene repair group , CD19 and other human cell membrane proteins accounted for the proportion of human CD45 protein.
  • blank control represents cells that have not undergone gene editing
  • gene repair represents cells that have undergone gene repair.
  • FIG. 18 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients.
  • the transplantation has undergone gene repair and has not undergone gene editing.
  • Cells from the irradiator enter the 6-week-old NPG immunodeficiency mouse model, extract the genome of the cells before transplantation and the bone marrow 16 weeks after transplantation, amplify the target fragment and NGS, and analyze the NHEJ and HDR by bioinformatics methods. proportion.
  • NHEJ non-homologous end joining
  • Indels represents the ratio of Indels
  • HDR Homology-directed repair
  • Figure 19 Isolation of bone marrow from NPG immunodeficient mice 16 weeks after one transplantation, and transplantation into new irradiated NPG immunodeficient mice for two transplantation experiments. Isolate bone marrow cells at 12 weeks after transplantation and detect the proportion of human CD45-positive cells. The proportion of CD45-positive cells is calculated as human CD45-positive cells%/(human CD45-positive cells%+mouse CD45-positive cells%), human CD45-positive cells % And mouse CD45 positive cells% are the results measured by flow cytometry experiments. Blank control: represents cells that have not undergone gene editing, and gene repair: represents cells that have undergone gene repair.
  • Figure 20 Isolation of bone marrow 16 weeks after one transplantation of NPG immunodeficiency mice, and then transplantation into new irradiated NPG immunodeficiency mice for two transplantation experiments.
  • the bone marrow cells were isolated 12 weeks after transplantation to test the gene repair efficiency and analyze the ratio of NHEJ and HDR.
  • NHEJ non-homologous end connection, representing the ratio of Indels
  • HDR homologous recombination repair, representing the ratio of gene repair.
  • FIG. 21 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 into hiPSCs derived from XLSA patients, extract the genome 2 days after electrotransformation, and amplify through sequence similarity prediction analysis and unbiased whole-genome analysis Methods
  • the target fragments of 32 potential off-target sites predicted by the Digenome-Seq method were analyzed by NGS sequencing, and the mutation frequency of each off-target site was analyzed by bioinformatics methods.
  • gene repair represents cells that have undergone gene editing
  • POT potential off-target
  • On-target represents gene editing efficiency.
  • This application provides a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations through gene editing, which can efficiently repair the ALAS-2 gene mutations in hiPSC and CD34+HSPC derived from XLSA patients , Significantly increase the expression of ALAS-2 gene and protein, thereby significantly increasing the synthesis of heme in differentiated red blood cells, promoting the maturation of red blood cells and the ability to carry oxygen, improving the symptoms of anemia patients, so as to overcome the shortcomings of traditional treatment methods and meet the clinical needs Treatment requirements.
  • ALAS-2 5-aminolevulinic acid synthase 2
  • the "gene editing” mentioned in this application refers to the technique of site-specific modification of the genome to achieve site-specific deletion, insertion, and/or replacement of specific nucleotides and nucleotide fragments at the gene level.
  • well-known gene editing technologies include artificial nuclease-mediated zinc finger nuclease (ZFN) technology, transcription activator-like effector nuclease (TALEN) technology, and RNA-guided CRISPR/Cas nuclease (CRISPR/Cas RGNs) technology. They can specifically recognize the target site, and after precise cutting of its single or double strand, the cell's endogenous repair mechanism completes the knockout and replacement of the target gene.
  • CRISPR/Cas technology is an emerging gene editing technology, which uses sgRNA complementary to the target sequence to guide Cas enzyme to cut DNA at a specific point.
  • a “mutated sequence” is a gene sequence whose nucleotide sequence has been changed compared to a normal natural sequence.
  • the nucleotide sequence that replaces the mutated sequence to achieve correction of the mutated sequence is called a "correction sequence”.
  • "Donor DNA” is DNA containing a "correction sequence”. After the donor DNA molecule containing the correction sequence is introduced into the cell by means of electroporation or transduction, homologous recombination can occur with the mutant sequence, so that the correction sequence can replace the mutant sequence to realize gene editing.
  • Stem cell refers to a cell population with vigorous proliferation potential, multi-differentiation ability and self-renewal ability.
  • Hematopoietic stem cells refer to cell populations with vigorous proliferation potential, multidirectional differentiation into blood cells, and self-renewal capabilities. Hematopoietic stem cells can not only differentiate and supplement various blood cells, but also maintain the characteristics and quantity of stem cells through self-renewal. The degree of differentiation and proliferation ability of hematopoietic stem cells are different and heterogeneous.
  • Pluripotent hematopoietic stem cells are the most primitive, and first differentiate into directed pluripotent hematopoietic stem cells, such as myeloid hematopoietic stem cells that can produce granulocytes, erythroid, mononuclear and megakaryocyte-platelet lines, and lymphoids that can produce B lymphocytes and T lymphocytes stem cell. These two types of stem cells not only maintain the basic characteristics of hematopoietic stem cells, but are also slightly differentiated. They are responsible for the occurrence of "bone marrow components" and lymphocytes, so they are called directed pluripotent hematopoietic stem cells. They further differentiate into hematopoietic progenitor cells.
  • directed pluripotent hematopoietic stem cells such as myeloid hematopoietic stem cells that can produce granulocytes, erythroid, mononuclear and megakaryocyte-platelet lines, and lymphoids that can produce B lymphocytes and
  • hematopoietic progenitor cells Although this cell is also a primitive blood cell, it has lost many of the basic characteristics of hematopoietic stem cells. Lost the ability of repeated self-renewal, and rely on the proliferation and differentiation of hematopoietic stem cells to supplement the number; the proliferation potential is limited and can only divide several times. According to the number of blood cell lines that hematopoietic progenitor cells can differentiate, they are divided into unipotent hematopoietic progenitor cells (differentiated into only one blood cell line) and oligopotent hematopoietic progenitor cells (differentiated into 2 to 3 blood cell lines).
  • hematopoietic stem cells refer to cell populations that can form granulocytes, erythroid, monocytes, megakaryocytes-platelet cells and/or lymphoid cells through differentiation or directed differentiation. They are pluripotent hematopoietic stem cells and are multipotent. Generic term for hematopoietic stem cells and hematopoietic progenitor cells.
  • Hematopoietic stem cells can be derived from bone marrow (bone marrow hematopoietic stem cells), peripheral blood (peripheral hematopoietic stem cells), umbilical cord blood (umbilical cord blood hematopoietic stem cells), and can also be derived from placental stem cells or hiPSC.
  • flow cytometry and fluorescently labeled anti-CD34 antibodies can be used to detect and count CD34-positive hematopoietic stem/progenitor cells (HSPC).
  • CRISPR/Cas is a gene editing technology, including but not limited to various naturally occurring or artificially designed CRISPR/Cas systems, such as the CRISPR/Cas9 system.
  • the naturally occurring CRISPR/Cas system (Naturally occurring CRISPR/Cas system) is an adaptive immune defense formed during the long-term evolution of bacteria and archaea, which can be used to fight invading viruses and foreign DNA.
  • a simple CRISPR/Cas9 system includes three components: Cas9 enzyme, crRNA (CRISPR-derived RNA) and tracrRNA (trans-activating crRNA).
  • crRNA contains a guide sequence and a sequence partially complementary to tracrRNA.
  • tracrRNA is trans-activating RNA (trans-activating RNA), which contains a long constant base sequence and provides a "stem-loop" structure bound by CRISPR nuclease (such as Cas9 enzyme).
  • crRNA combines with tracrRNA (trans-activating RNA) through base pairing to form a tracrRNA/crRNA complex, through which crRNA and the target sequence are complementary, and through the "stem-loop" structure in tracrRNA, Cas9 nuclease can be guided Cut the double-stranded DNA to the target site of the target sequence.
  • tracrRNA and crRNA can be combined to transform into a guiding sgRNA (single guide RNA), so that the sgRNA is sufficient to guide Cas9's targeted cleavage of DNA.
  • a guiding sgRNA single guide RNA
  • Cas9 nuclease can co-localize RNA, DNA and protein, and has great potential for transformation.
  • the CRISPR/Cas system can use type 1, type 2 or type 3 Cas proteins. In some embodiments of the invention, the method uses Cas9.
  • CRISPR/Cas systems include but are not limited to the systems and methods described in WO2013176772, WO2014065596, WO2014018423, US8,697,359, PCT/CN2018/112068, PCT/CN2018/112027.
  • Cell “differentiation” refers to the process in which cells from the same source gradually produce cell groups with different morphological structures and functional characteristics.
  • the "differentiation" from hematopoietic stem cells to erythrocytes includes hematopoietic stem cell stage, erythroid progenitor cell stage, erythroid precursor cell (primary red blood cell to late red blood cell) proliferation and differentiation stage, reticulocyte proliferation and maturation process, and reticulum Red blood cells are released from peripheral blood to mature into red blood cells.
  • Hematopoietic stem cell stage It is currently known that hematopoietic stem cells mainly exist in bone marrow, spleen, liver and other hematopoietic tissues. There is also a small amount of circulation in the peripheral blood.
  • Erythroid progenitor cell stage In the progenitor cell stage, cells are a cell group between hematopoietic stem cells and erythroid precursor cells. Hematopoietic stem cells differentiate into erythroid progenitor cells under the influence of bone marrow hematopoietic microenvironment. The hematopoietic microenvironment includes the microvascular system, nervous system and hematopoietic interstitium. Humoral factors and cytokines have a special effect and influence on the differentiation of hematopoietic stem cells. Erythroid precursor cell stage: including primitive red blood cells, early young red blood cells, middle young red blood cells, late young red blood cells and reticulocytes to reach mature red blood cells.
  • Non-homologous end joining is also abbreviated as NHEJ (Non-homologous end joining), which refers to eukaryotic cells that do not rely on DNA homology to avoid the retention of DNA or chromosome breaks and the resulting DNA The effect of degradation is a DNA double-strand break repair mechanism that forcibly connects two DNA ends to each other.
  • NHEJ may produce insertions and deletions (Indels, Insertions and deletions), leading to gene mutations.
  • HDR Homology-directed repair
  • HDR homology-mediated double-stranded DNA repair
  • “Anemia” refers to a clinical symptom in which the volume of human peripheral blood red blood cells decreases below the lower limit of the normal range.
  • “Sideroblastic anemia” is a disorder of iron utilization. It is characterized by the appearance of a large number of ring sideroblasts in the bone marrow, ineffective production of red blood cells, excessive tissue iron reserves, and small cell hypochromic anemia in the peripheral blood. Sideroblastic anemia is mainly divided into acquired and hereditary sideroblastic anemia. Among them, hereditary sideroblastic anemia is mostly adolescents, males and have family history. Poor iron utilization, impaired heme synthesis and ineffective production of red blood cells are the main links in the pathogenesis of this disease.
  • the result of poor iron utilization and heme synthesis disorder is the formation of hypochromic anemia.
  • a large amount of iron accumulates in red blood cells and various tissues, which damages the morphology and function of red blood cells and causes premature destruction of red blood cells.
  • a large amount of iron deposits in various tissues, forming hemochromatosis, affecting the functions of various tissues and organs.
  • This application relates to genetic repair of specific mutations in the ALAS-2 gene, such as mutations in exons 5-11 or intron-1 of the ALAS-2 gene, such as Int-1-GATA point mutations, to improve the source of iron Myeloblastic anemia patients, such as hereditary sideroblast anemia patients, specifically, CD34 + HSPC or human induced pluripotent stem cells (hiPSC) in XLSA patients with ALAS-2 gene and protein expression, so that the treatment includes hereditary sideroblasts Immature red blood cell anemia, such as sideroblast anemia of XLSA.
  • specific mutations in the ALAS-2 gene such as mutations in exons 5-11 or intron-1 of the ALAS-2 gene, such as Int-1-GATA point mutations
  • iron Myeloblastic anemia patients such as hereditary sideroblast anemia patients, specifically, CD34 + HSPC or human induced pluripotent stem cells (hiPSC) in XLSA patients with ALAS-2 gene and
  • the present invention relates to a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations in the chromosomes of hematopoietic stem cells through gene editing, wherein the gene editing includes: (a) A donor DNA containing a single-stranded correction sequence corresponding to the mutant sequence is introduced into the hematopoietic stem cell; (b) a sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the donor The correction sequence on the somatic DNA replaces the mutation sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
  • the gene editing includes: (a) A donor DNA containing a single-stranded correction sequence corresponding to the mutant sequence is introduced into the hematopoietic stem cell; (b) a sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic
  • the invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2.
  • the present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing and increasing the expression of functional ALAS-2, thereby increasing the production of heme in cells derived from the hematopoietic stem cells.
  • the present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of heme in the cells derived from the hematopoietic stem cells, thereby promoting the maturation of the hematopoietic stem cells .
  • the present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of heme in the cells derived from the hematopoietic stem cells, and promoting the maturation of the hematopoietic stem cells, thereby Methods of treating individuals include hereditary sideroblast anemia, such as XLSA sideroblast anemia.
  • the hematopoietic stem cells are obtained from patients with sideroblast anemia, including hereditary sideroblast anemia, such as XLSA.
  • the patient is a male individual or a female individual, and the obtained stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
  • HSPC hematopoietic stem and progenitor cells
  • hiPSC human induced pluripotent stem cells
  • CD34-positive hematopoietic stem/progenitor cells are isolated from an organism (individual) containing cells of hematopoietic origin. "Separate” means to remove from its original environment. For example, a cell is isolated if it is separated from some or all of the components that normally accompany it in its natural state. Hematopoietic stem cells/progenitor cells can be obtained or isolated from unfractionated or fractionated bone marrow of adults, including femurs, hip bones, ribs, sternum and other bones.
  • Hematopoietic stem cells and progenitor cells can be directly obtained or separated from the hip bone using a needle and syringe, or obtained from the blood, usually obtained from the blood after pretreatment with a hematopoietic stem cell mobilizer such as G-CSF (granulocyte colony stimulating factor) .
  • a hematopoietic stem cell mobilizer such as G-CSF (granulocyte colony stimulating factor)
  • G-CSF granulocyte colony stimulating factor
  • Other sources of hematopoietic stem and progenitor cells include cord blood, placental blood, and peripheral blood of mobilized individuals.
  • the cell population After the cell population is isolated from an individual (such as bone marrow or peripheral blood), it can be further purified to obtain CD34-positive hematopoietic stem cells/progenitor cells.
  • mature lineage-directed cells in an isolated cell population can be removed by immunization, for example, by using antibodies that bind to a set of "lineage" antigens (eg CD2, CD3, CD11b, CD14, CD15, CD16, CD19, CD56, CD123, and CD235a)
  • the solid matrix is labeled, and then the original hematopoietic stem cells and progenitor cells are separated with antibodies that bind to CD34 positive antigen.
  • Kits for purifying hematopoietic stem and progenitor cells from a variety of cell sources are commercially available, and in specific embodiments, these kits can be used with the methods of the present invention.
  • CD34 positive hematopoietic stem cells/progenitor cells can represent at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% of the cell population rich in CD34 positive cells Or 100% CD34-positive hematopoietic stem/progenitor cells (HSPC).
  • the gene editing includes introducing a donor DNA containing a single-stranded correction sequence corresponding to the mutant sequence into the hematopoietic stem cell.
  • the single-stranded correction sequence replaces the mutant sequence in the ALAS-2 gene.
  • the donor DNA has the same nucleotide composition and length as the single-stranded correction sequence. In some embodiments, the donor DNA is longer than the single-stranded correction sequence, for example, one or more nucleotides are added to one or both ends of the single-stranded correction sequence. In some embodiments, the sequence composed of the added nucleotides is a nuclease specific recognition site. In some embodiments, both ends of the calibration sequence in the donor DNA may further include protective bases for the specific recognition site of the nuclease. In some embodiments, the donor DNA further comprises one or more LNA nucleosides. In some embodiments, the donor DNA is single-stranded. In some embodiments, the donor DNA is circular.
  • the donor DNA is provided in the form of a plasmid or viral vector.
  • the donor DNA is ssODN (single-stranded donor oligonucleotides).
  • the donor DNA is shown in SEQ ID NO: 4.
  • the donor DNA is chemically modified, such as 2'-O-methylation modification on nucleotide ribose, 3'phosphorothioate modification between nucleotides, and 5' End phosphorylation modification.
  • the chemical modification is a 2'-O-methylation modification and/or a ribose 3 nucleotides before the 5'end and 3 nucleotides after the 3'end of the donor DNA. 3'phosphorothioate modification between glycidyl acids.
  • the chemical modification is one, two and/or three bases before the 5'end of the donor DNA and/or the 2'-O of the last nucleotide ribose at the 3'end.
  • the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose in the 3 nucleotides before the 5'end and the 3 nucleotides after the 3'end, and the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose before the 5'end.
  • the three internucleotide linkages after the 3 and 3'ends contain phosphorothioate modification.
  • the donor DNA contains a 2'-O-methylation modification in the ribose of the first 5 nucleotides at the 5'end and the last 5 nucleotides at the 3'end, and the ribose at the 5'end
  • the first 5 and the last 5 internucleotide linkages at the 3'end contain phosphorothioate modification.
  • the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end.
  • the donor DNA contains a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, and The first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end also contain the 2'-O-methylation modification of ribose.
  • sequence-specific nucleases include RNA guide nuclease, zinc finger nuclease (ZFN), and transcription activator-like effector nuclease (TALEN).
  • ZFN zinc finger nuclease
  • TALEN transcription activator-like effector nuclease
  • the sequence-specific nuclease may be, for example, an RNA guide nuclease, that is, a Cas nuclease, and specifically may be Cas9.
  • the nuclease cleavage site is no more than about 11 nucleotides away from the mutation site.
  • mRNA encoding Cas9 such as mRNA containing an ARCA cap, is introduced into stem cells (e.g., by electroporation or other means of gene transduction).
  • the nucleotide encoding the Cas nuclease e.g., Cas9
  • a viral vector e.g., a lentiviral vector.
  • the sgRNA and the Cas9-encoding nucleic acid are present in the same vector.
  • the sgRNA and the Cas9-encoding nucleic acid are in different vectors.
  • it further includes introducing sgRNA that recognizes the ALAS-2 gene into the hematopoietic stem cells, such as CD34 + HSPC.
  • the "guide sequence" in sgRNA is any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and direct the sequence-specific binding of the CRISPR complex to the target sequence.
  • the degree of complementarity between the guide sequence and its corresponding target sequence is about or greater than about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more.
  • the optimal alignment can be determined using any appropriate algorithm for aligning sequences.
  • Non-limiting examples include Smith-Waterman algorithm, Needleman-Wimsch algorithm, Burrows-Wheeler Transform-based algorithms (such as Burrows Wheeler Aligner), ClustalW, Clustai X, BLAT, Novoalign (Novocraft Technologies, ELAND ((Illumina, San Diego, CA), SOAP (available at soap.genomics.org.cn) and Maq (available at maq.sourceforge.net).
  • the guide sequence length can be about or greater than about 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, 55, 60, 65, 70, 75 or more nucleotides.
  • the guide sequence length is less than about 75, 70, 65, 60, 55 , 50, 45, 40, 35, 30, 25, 20, 15, 12 or fewer nucleotides.
  • the ability of the guide sequence to direct the sequence-specific binding of the CRISPR complex to the target sequence can be assessed by any appropriate assay method
  • a host cell with the corresponding target sequence can be provided with the components of the CRISPR system (including the guide sequence to be tested) sufficient to form a CRISPR complex, for example, by transfection with a vector encoding the CRISPR sequence component, and then evaluate the target sequence.
  • the preferential cleavage (such as by the Surveyor assay as described herein) is carried out.
  • the cleavage of the target polynucleotide sequence can be performed in the test tube by providing the target sequence, the CRISPR complex (containing the guide sequence to be tested and the other The evaluation is carried out by comparing the binding or cleavage rate of the test and the control guide sequence in the target sequence.
  • Other methods known to those skilled in the art can also be used for the above determination and evaluation.
  • the sgRNA may be modified, for example, it may be chemically modified, specifically, the sgRNA is modified by nucleotide ribose 2'-O-methylation and/ Or internucleotide 3'phosphorothioate modified.
  • “Chemically modified sgRNA” refers to the special chemical modification of sgRNA, such as the 2'-O-methylation modification of ribose with 3 nucleotides at the 5'and 3'ends and/or the 3'between nucleotides. 'Phosphorothioate modification.
  • the chemical modification is a 2'-O-methylation modification of one, two and/or three bases before the 5'end of the sgRNA and/or the last nucleotide ribose at the 3'end.
  • the sgRNA comprises 2'-O-methyl modification of ribose at the first 3 nucleotides at the 5'end and 3 nucleotides at the rear of the 3'end and/or an internucleotide 3'sulfur Phosphorylation modification.
  • the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end
  • the connection between the 3 nucleotides after the 3'end contains phosphorothioate modification.
  • the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end
  • the last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
  • the chemically modified sgRNA has at least the following two advantages.
  • chemical modification methods commonly used in the art can be used, as long as they can improve the stability of sgRNA (extend the half-life) and enhance the ability to enter the cell membrane.
  • it also includes the use of other modification methods, for example, Deleavey GF1, Damha MJ. Designing chemically modified oligonucleotides for targeted gene silencing. Chem Biol. 2012 Aug 24; 19(8): 937-54, and Hendel et al. Chemically modified guide RNAs enhancement CRISPR-Cas genome editing in human primary cells. Nat Biotechnol. 2015 Sep; 33(9):985-989 The chemical modification methods reported in the literature.
  • the sgRNA may be complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome.
  • the sgRNA may include a nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene.
  • the nucleic acid sequence that is complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA may be about 17 to about 20 nucleotides long.
  • the sgRNA is selected from the following group: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3.
  • This application uses CRISPR/Cas9 gene editing technology to develop a method for efficiently repairing ALAS-2 gene mutations.
  • the gene repair efficiency is as high as about 30%-40%.
  • the ALAS-2 gene expression of the cells after the gene repair reaches the healthy donor ALAS -2 expression is about 50%, which can significantly alleviate the clinical manifestations of patients with sideroblast anemia (such as XLSA).
  • using the method can produce extremely high mutation frequency at the target site (On-target), far exceeding the mutation frequency of the blank control group, and close to 100%, while at potential off-target sites Then the significant difference between the gene editing group and the blank control group cannot be measured.
  • the method will not cause off-target in the hematopoietic stem cell genome.
  • the off-target rate is less than 1%, for example, less than 0.5% or less than 0.1%. This extremely low off-target rate can improve the safety of the method for gene repair of hematopoietic stem cells.
  • the present application provides a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2, wherein the gene editing includes: (a) comprising: The donor DNA of the single-stranded correction sequence of the mutant sequence is introduced into the hematopoietic stem cell; (b) the sequence-specific nuclease that cuts the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction on the donor DNA The sequence replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
  • the hematopoietic stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
  • the sequence-specific nuclease is an RNA-guided nuclease, specifically Cas9.
  • the inventors found that the closer the nuclease cleavage site is to the mutation site, the more helpful it is to improve the repair efficiency.
  • the nuclease cleavage site is no more than about 11 nucleotides away from the mutation site, for example, about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, about 1 nucleotide, or nuclease cleavage site overlaps the mutation site.
  • the sgRNA is complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome.
  • the sgRNA comprises a nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene.
  • the nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA is 17-20 nucleotides long.
  • the sgRNA is selected from the following group: SEQ ID NO: 1-SEQ ID NO: 3, preferably SEQ ID NO: 1.
  • the sgRNA is chemically modified. Specifically, the sgRNA is modified by 2'-O-methylation of nucleotide ribose and/or internucleotide 3'phosphorothioate modification, for example, the chemical modification is that of the sgRNA 2'-O-methylation modification of one, two and/or three bases before the 5'end and/or the last nucleotide ribobase at the 3'end. In some embodiments, the sgRNA comprises the first 3 nucleotides at the 5'end and the 3 nucleotides after the 3'end.
  • the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end The connection between the 3 nucleotides after the 3'end contains phosphorothioate modification.
  • the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end
  • the last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
  • the donor sequence is longer than the correction sequence.
  • the donor sequence is the same length as the correction sequence, and is about 60 to about 200 nucleotides in length, for example, 60 to about 180, 60 to about 160, 60 to about 140. One, 60 to about 120, 60 to about 100, 60 to about 80 nucleotides long.
  • the correction sequence comprises a 5'arm that is substantially complementary to a target region located at the 3'end of the mutation site, and a 5'arm that is substantially complementary to a target region located at the 5'end of the mutation site. 3'arm.
  • the substantially complementary means that the 5'arm or the 3'arm of the correction sequence has high homology with the target region at the 3'end or the target region at the 5'end of the mutation site, for example, at least about 90%. , At least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% homology. Most preferably, the 5'arm or 3'arm of the correction sequence has 100% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively.
  • the 5'arm of the correction sequence is longer than the 3'arm of the correction sequence, the 3'arm of the correction sequence is longer than the 5'arm of the correction sequence, or the 5'arm of the correction sequence and the The 3'arms of the calibration sequence have the same length.
  • the sequence of the donor DNA is shown in SEQ ID NO: 4.
  • the donor DNA is chemically modified, such as 2'-O-methylation modification on nucleotide ribose, 3'phosphorothioate modification between nucleotides, and 5' End phosphorylation modification.
  • the chemical modification is a 2'-O-methylation modification and/or a ribose 3 nucleotides before the 5'end and 3 nucleotides after the 3'end of the donor DNA. 3'phosphorothioate modification between glycidyl acids.
  • the chemical modification is one, two and/or three bases before the 5'end of the donor DNA and/or the 2'-O of the last nucleotide ribose at the 3'end.
  • the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose in the 3 nucleotides before the 5'end and the 3 nucleotides after the 3'end, and the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose before the 5'end.
  • the three internucleotide linkages after the 3 and 3'ends contain phosphorothioate modification.
  • the donor DNA contains a 2'-O-methylation modification in the ribose of the first 5 nucleotides at the 5'end and the last 5 nucleotides at the 3'end, and the ribose at the 5'end
  • the first 5 and the last 5 internucleotide linkages at the 3'end contain phosphorothioate modification.
  • the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end.
  • the donor DNA contains a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, and The first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end also contain the 2'-O-methylation modification of ribose.
  • the donor DNA, sgRNA and/or mRNA encoding Cas9 are introduced sequentially or simultaneously into the hematopoietic stem cells by electroporation (or electrotransduction).
  • the weight ratio of the sgRNA to the donor DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11:1, about 1. :10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1.
  • the weight ratio of the mRNA encoding the sequence-specific nuclease to the single-stranded DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11: 1.
  • the Cas9 mRNA: sgRNA-1: ssODN is 6 ⁇ g: 4 ⁇ g: 6 ⁇ g, 6 ⁇ g: 4 ⁇ g: 8 ⁇ g, 6 ⁇ g: 4 ⁇ g: 10 ⁇ g, 6 ⁇ g: 4 ⁇ g: 12 ⁇ g.
  • the donor DNA, sgRNA and/or mRNA encoding Cas9 are introduced sequentially or simultaneously into the hematopoietic stem cells by electroporation (or electrotransduction).
  • the electrical transduction conditions are, for example, 250-360V, 0.5-1ms; 250-300V, 0.5-1ms; 250V, 1ms; 250V, 2ms; 300V, 0.5ms; 300V, 1ms; 360V, 0.5ms; or 360V, 1ms.
  • the hematopoietic stem cells gene-repaired by the above-mentioned method of this application that is, the ALAS-2 gene mutation (for example, the gene mutation in exons 5-11 or intron-1 of the gene, specifically the Int-1-GATA mutation) is corrected or
  • the repaired hematopoietic stem cells can be returned to patients with sideroblast anemia (for example, hereditary sideroblast anemia, specifically XLSA).
  • sideroblast anemia for example, hereditary sideroblast anemia, specifically XLSA
  • the hematopoietic stem cells genetically repaired by the above method of the present application are returned to the patient, the hematopoietic stem cells can be colonized in the bone marrow of the patient for a long time, and the hematopoietic system of the patient can be successfully reconstructed.
  • the ALAS-2 gene mutation repaired CD34 + HSPC is derived from the peripheral blood of the individual to be treated (with or without bone marrow hematopoietic stem cell mobilization) or obtained from the bone marrow of the individual.
  • the CD34+HSPC population is subjected to erythroidization using hematopoietic stem cell erythroid expansion and differentiation medium.
  • the hematopoietic stem cell erythroid expansion and differentiation medium includes a basal medium and a composition of growth factors, wherein the composition of growth factors includes stem cell growth factor (SCF); interleukin 3 (IL-3) and erythropoietin (EPO).
  • SCF stem cell growth factor
  • IL-3 interleukin 3
  • EPO erythropoietin
  • erythroid differentiation and denucleation medium for erythroid differentiation and denucleation of hematopoietic stem cells, the erythroid differentiation and denucleation medium comprising a basal medium, growth factors, and progesterone receptors and glucocorticoids Antagonists and/or inhibitors of hormone receptors.
  • the growth factor in the erythroid differentiation and denucleation medium includes erythropoietin (EPO), and the antagonist and/or inhibitor of the progesterone receptor and glucocorticoid receptor is selected From any one or two or more of the following compounds (I) to (IV):
  • the hematopoietic stem cell erythroid expansion and differentiation medium comprises a basal medium and growth factor additives, wherein the basal medium can be selected from any serum-free basal medium, such as STEMSPAN TM SFEM II (STEM CELLS TECHNOLOGY Inc.), IMDM (Iscove's Modified Dulbecco's Medium), optionally supplemented with ITS (Thermofisher), L-gulutamin (Thermofisher), vitamin C and/or bovine serum albumin; wherein the growth factor additive is selected from IL-3 A combination of one or more of, SCF and EPO.
  • serum-free basal medium such as STEMSPAN TM SFEM II (STEM CELLS TECHNOLOGY Inc.), IMDM (Iscove's Modified Dulbecco's Medium), optionally supplemented with ITS (Thermofisher), L-gulutamin (Thermofisher), vitamin C and/or bovine serum albumin; wherein
  • any commonly used basic medium can be used in the above hematopoietic stem cell erythroid expansion and differentiation medium, such as STEMSPAN TM SFEM II (purchased from STEM CELL TECHONOLOGIES); for example, IMDM, DF12, Knockout DMEM, RPMI 1640 from Thermo Fisher , Alpha MEM, DMEM, etc.
  • other components can be further added to these basic media as needed, for example, ITS (that is, mainly including insulin, human transferrin, and selenium), L-glutamine, vitamin C, and bovine serum albumin can be added.
  • ITS 2mM L-glutamine, 10-50 ⁇ g/ml vitamin C and 0.5-5 mass% BSA (bovine serum albumin)
  • BSA bovine serum albumin
  • DF12 can be added with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin.
  • Knockout DMEM can be supplemented with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin
  • RPMI 1640 can be supplemented with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin
  • Alpha MEM can be supplemented with the same concentration ITS, L-glutamine, vitamin C and bovine serum albumin
  • DMEM can also be added with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin.
  • the concentration of additional ITS in various basal media can be: insulin concentration is 0.1 mg/ml, human transferrin is 0.0055 mg/ml, selenium element is 6.7 ⁇ 10 -6 mg/ml.
  • the concentration of each component of ITS added can also be adjusted according to actual needs. ITS can be purchased from Thermofisher and adjusted to the appropriate final use concentration as required.
  • the above-mentioned hematopoietic stem cells repaired by gene editing can be directly or cultured for one or more days and then returned to the sideroblast anemia (for example, XLSA) patient for treatment.
  • the hematopoietic stem cells are cultured for one or more days (e.g., 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days) before being administered to the individual.
  • the hematopoietic stem cells are stored in a frozen condition for at least 24 hours before the hematopoietic stem cells are returned to the individual patient.
  • the cells are cultured for one or more days (e.g., 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days) before being stored under freezing conditions.
  • the treatment comprises administering to the individual (such as intravenous injection, comprising a single intravenous injection) ⁇ 2x10 6, ⁇ 5x10 6, ⁇ 1x10 7, ⁇ 2x10 7 cells / kg body weight above ALAS-2 Hematopoietic stem cells repaired by genetic mutations.
  • hematopoietic stem cells With the proliferation and differentiation of hematopoietic stem cells, the production of heme can be detected. It can be stained with Benzidine: benzidine staining. In the presence of hydrogen peroxide, benzidine can bind and react with the heme in hemoglobin to produce a brown or blue precipitate to detect the synthesis of heme. Assess the effect of gene therapy. The effect of gene therapy can also be evaluated by detecting the expression of ALAS-2 gene and/or protein by conventional methods in the art.
  • the differentiated cells can also be evaluated by, for example, Benzidine staining and Wright-Giemsa staining.
  • the red blood cells differentiated from healthy donors and hematopoietic stem cells that have undergone gene repair are mainly mature red blood cells and reticulocytes, while the red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are mainly promyelocytic red blood cells, indicating that differentiation is stagnant. In the early stage, mature red blood cells cannot be differentiated.
  • the detection methods known in the art can determine the differentiation of hematopoietic stem cells, so as to determine whether the ALAS-2 gene mutation of hematopoietic stem cells has been corrected.
  • Example 1 Efficient gene repair of ALAS-2 intron-1 point in hiPSC derived from XLSA patients mutation
  • This embodiment relates to the use of CRISPR/Cas9 system to edit XLSA patient-derived human induced pluripotent stem cells (Human induced pluripotent stem cells, hiPSC) to efficiently repair the ALAS-2 intron-1 point mutation, the location is (X: 55054635 [Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C), because this site is the junction of GATA-1 and ALAS-2 genes, so The point mutation is named Int-1-GATA.
  • sgRNA-1 aactctggcaactttacctg (SEQ ID NO: 1)
  • sgRNA-2 caactttacctgtggtctgc
  • sgRNA-3 gggctgagcctgcagaccac (SEQ ID NO: 3)
  • tcccacgccctggtctcagcttggggagtggtcagaccccaatggcgataaactctggcaactttacctgtggtctgcaggctcagccccaagtgct (SEQ ID NO: 4), the
  • Cas9 mRNA encoding information is as follows: gacaagaagtacagcatcggcctggacatcggcaccaactctgtgggctgggccgtgatcaccgacgagtacaaggtgcccagcaagaaattcaaggtgctgggcaacaccgaccggcacagcatcaagaagaacctgatcggagccctgctgttcgacagcggcgaaacagccgaggccacccggctgaagagaaccgccagaagaagatacaccagacggaagaaccggatctgctatctgcaagagatcttcagcaacgagatggccaaggtggacgacagcttctccacagactggaagagtccttcctggtggaagaggataagaagcacgagcggcaccccatctggtggaggaggagg
  • sgRNA refers to the modification of the ribose of the first three nucleotides of the 5'end and the last three nucleotides of the 3'end of the sgRNA with 2'-O-methylation and the 3'sulfur between the nucleotides. Phosphorylation modification. As shown in the following chemical formula, the left side is the chemically modified sgRNA, and the right side is the unmodified sgRNA.
  • the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end.
  • Cas9 mRNA and sgRNA were purchased from Trilink Biotechnologies, USA.
  • Reverse primer catatggcaacctccttcatc (SEQ ID NO: 7)
  • the Indels efficiency of sgRNA-1, sgRNA-2, and sgRNA-3 are about 35%, 10%, and 40%, respectively, as shown in FIG. 3.
  • efficient Indels efficiency is the prerequisite for gene repair, so we choose sgRNA-1 and sgRNA-3 as the preferred sgRNA;
  • second the closer the sgRNA cutting site is to the gene repair site, the closer the gene repair is. The higher the efficiency (Xiquan Liang, et al. Journal of Biotechnology. 2016; Mark A. et al. Science Translational Medicine. 2017).
  • sgRNA-1 we optimized the amount of Cas9 and sgRNA to electrotransform the same amount of hiPSC (1.0*10 ⁇ 6 cells), that is, electrotransform Cas9mRNA: sgRNA, 1 ⁇ g:1 ⁇ g, 2 ⁇ g:2 ⁇ g, 3 ⁇ g, respectively: 3 ⁇ g, 4 ⁇ g: 4 ⁇ g, 6 ⁇ g: 6 ⁇ g enter the hiPSC, 4 days after electroporation, extract the genome of the hiPSC, select about 450bp around the sgRNA cleavage site, and a total length of 905bp fragments for amplification and Sanger sequencing. Analyze the efficiency of Indels through the "Synthego ICE Analysis" online software. The results showed that with the increase of Cas9 mRNA and sgRNA, the efficiency of Indels increased, and the efficiency of 4ug:4ug was the highest, reaching about 50% gene editing efficiency, as shown in Figure 4.
  • the test volume of Cas9 mRNA:sgRNA:ssODN is 1 ⁇ g:1 ⁇ g:1 ⁇ g, 2 ⁇ g:2 ⁇ g:2 ⁇ g, 3 ⁇ g:3 ⁇ g:3 ⁇ g, 4 ⁇ g:4 ⁇ g:4 ⁇ g, 6 ⁇ g:6 ⁇ g:6 ⁇ g.
  • the results show that as the amount of addition increases, The gene repair efficiency HDR is improved.
  • the highest gene repair efficiency HDR is 6 ⁇ g:6 ⁇ g:6 ⁇ g, which is about 25%. This proves that we have successfully repaired the point mutations in hiPSC derived from XLSA patients at the Int-1-GATA position. Point successfully realized the repair from C to T.
  • Cas9 mRNA:sgRNA:ssODN is 6 ⁇ g:6 ⁇ g:6 ⁇ g, which induces a higher Indels efficiency (%NHEJ, as shown in Figure 5).
  • Example 2 Efficient gene repair of ALAS-2 in CD34+HSPC derived from bone marrow of XLSA patients Intron-1 point mutation
  • Example 1 we achieved an efficient repair of the Int-1-GATA point mutation of hiPSC derived from XLSA. Refer to the amount of Cas9 mRNA, sgRNA-1 and ssODN added in Example 1. In this experiment, we tried gene repair CD34+HSPC from bone marrow of XLSA patients.
  • Cas9 mRNA sgRNA-1: ssODN is 6 ⁇ g:4 ⁇ g: 12 ⁇ g, 4 days later, the HSPC genome was extracted, and the gene repair efficiency (HDR) and Indels efficiency (NHEJ) were analyzed by the next-generation sequencing bioinformatics method. As shown in Figure 7, the gene repair efficiency reached about 40%.
  • Example 2 Refer to the addition amount of Cas9 mRNA, sgRNA-1 and ssODN found in Example 2 (6 ⁇ g:4 ⁇ g:12 ⁇ g), select 300v 1ms electroporation conditions, and electrotransform Cas9 mRNA, sgRNA-1 and ssODN into HSPC derived from the bone marrow of XLSA patients, respectively , Use the following "two-step method" differentiation protocol for red blood cell differentiation experiments. In addition, our erythroid differentiation of healthy donors mobilized CD34+HSPC derived from peripheral blood as a positive control.
  • the two-step method of differentiation is to use HSPC erythroid amplification and differentiation medium for differentiation, and then use HSPC erythroid differentiation and denucleation medium for differentiation.
  • the erythroid expansion and differentiation medium of hematopoietic stem cells is StemSpan TM SFEM II, the growth factor is 50-200ng/ml SCF, 10-100ng/ml IL-3, 1-10U EPO/ml, culture conditions: use Hematopoietic stem cell erythroid expansion and differentiation medium culture hematopoietic stem cells 1.0 ⁇ 10 ⁇ 5 cells/ml for 7 days.
  • the erythroid differentiation and denucleation medium for hematopoietic stem cells is STEMSPAN TM SFEM II, the growth factor is 1-10 U EPO, 100-1000 ⁇ g/ml human transferrin, and the chemical small molecule is 0.5-10 ⁇ m mifepristone.
  • the 1.0 ⁇ 10 ⁇ 6 cells/ml cells cultured in one step were differentiated in the hematopoietic stem cell erythroid differentiation denucleation medium for 11 days.
  • the differentiation efficiency of the gene repaired cells in the second stage of differentiation is significantly higher than that of the unrepaired cells, indicating that the former differentiated cells are more mature. This is because the ALAS-2 gene is involved in ferrous iron. Heme synthesis and red blood cell maturation (Zhang, et al. Nucleic Acids Research. 2017; Liu, et al. Nature Communications. 2018), so when the ALAS-2 gene mutation is repaired, the degree of red blood cell differentiation increases. 2) The differentiation efficiency of gene repaired cells in the second stage of differentiation is lower than that of healthy donor-derived cells. This is because the ALAS-2 Int-1-GATA mutation is partially repaired by the gene, and the efficiency is about 40%.
  • Benzidine staining hematopoietic stem cells derived from healthy donors and red blood cells differentiated from hematopoietic stem cells that have undergone gene repair. After Benzidine staining, the proportion of positive cells (shown by the red arrow) is significantly higher than that of hematopoietic stem cells that have not undergone gene repair. Red blood cells. The results of statistical analysis further showed that the percentage of Benzidine-positive cells derived from healthy donors and red blood cells differentiated from hematopoietic stem cells that have undergone gene repair is about 60%, while the red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are only 20%. .
  • red blood cells differentiated from healthy donors and hematopoietic stem cells that have undergone gene repair are mainly mature red blood cells and reticulocytes, while red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are promyelocytic erythrocytes. Mainly, indicating that differentiation is stagnant in the early stage, and mature red blood cells cannot be differentiated.
  • Example 2 Extract cell proteins from the red blood cells differentiated from CD34+HSPC in Example 2.2, and perform Western Blot experiment to detect the protein expression of ALAS-2, GATA-1, and GAPDH genes, as shown in FIG. 12.
  • Example 3 In vitro clone formation of CD34+HSPC derived from bone marrow of XLSA patients with gene repair
  • This experiment involves the detection of colony-formation units (CFU) of CD34+HSPC derived from the bone marrow of gene-edited XLSA patients.
  • CFU colony-formation units
  • Select the 300V 1ms electroporation conditions refer to the Cas9 mRNA, sgRNA-1 and ssODN additions found in Example 2, electroporate Cas9 mRNA, sgRNA-1 and ssODN into CD34+HSPC derived from the bone marrow of the XLSA patient, and 500 cells Resuspend in 1ml H4434 (purchased from Canada STEM CELLS Technologies), IMDM (purchased from Thermo Fisher) and FBS (purchased from Thermo Fisher) in a mixed solution, 14 days later, observe CFU-M, BFU-E, CFU-E under the microscope The number of clones with different morphologies such as CFU-G, CFU-GM, GEMM, etc., and the results are shown in Figure 13.
  • BFU-E, CFU-GM, CFU-E, CFU-MM represent the clonal formation of different blood system lineages such as erythroid, myeloid, and lymphatic system.
  • healthy donors represent healthy donors mobilizing CD34+HSPC derived from peripheral blood, blank control: represents cells that have not undergone gene repair, and gene repair represents cells that have undergone gene repair.
  • the experimental results show that: compared with cells that have not undergone gene repair, the CFU-GM, BFU-E, and CFU-E of the cells that have undergone gene repair are significantly increased.
  • BFU-E and CFU-E represent the erythroid pre-clone and terminal, respectively
  • the differentiated erythroid clones further proved that gene repairing the mutation site of ALAS-2 Int-1-GATA restored the ability of the CD34+HSPC erythroid to become mature red blood cells.
  • the gene repair efficiency is about 40%, the total number of clones formed by the gene repaired cells and the number of different subclones are lower than the number of clones formed by cells from healthy donors, which is in line with experimental expectations.
  • 300V 1ms electroporation conditions were selected, referring to the addition of Cas9 mRNA, sgRNA-1 and ssODN found in Example 2, electroporation of Cas9 mRNA, sgRNA-1 and ssODN into the bone marrow-derived CD34+HSPC of XLSA patients, the transplantation process
  • the NPG immunodeficiency mouse model irradiated by the irradiator purchased from Beijing Vitalstar Biotechnology, Inc.
  • Human CD45 and small blood cells were detected in the peripheral blood 10, 12, and 16 weeks after transplantation.
  • mice CD45 The expression of mouse CD45, and the expression of human CD45 in bone marrow and spleen and mouse CD45 in the bone marrow and spleen 16 weeks after transplantation were detected at the same time.
  • the results are shown in Figure 14.
  • the method of transplantation into mice is: 24 hours before cell transplantation , Irradiated with 1.0Gy rays to clear the bone marrow of the mouse model. Then, 1.0 ⁇ 10 ⁇ 6 cells resuspended with 20 ⁇ L of 0.9% normal saline were injected into the tail vein of the mouse, and then put into a clean animal room Medium feeding.
  • blank control represents cells that have not undergone gene repair, and gene repair represents cells that have undergone gene repair.
  • FIG. 14 and Figure 15 show that after the gene repaired CD34+HSPC, after transplantation into the mouse model, compared with the non-genetically repaired CD34+HSPC, the peripheral blood, bone marrow and spleen of the animal after the genetically modified cell transplantation
  • the increased expression ratio of human hCD45 in humans indicates that the gene repaired CD34+HSPC can be quickly and efficiently implanted into the hematopoietic system of the mouse model, and the differentiation function of the cells in vivo is normal, while the CD34+HSPC that has not undergone gene repair is implanted in the mouse model.
  • the mouse model has abnormal functions in the hematopoietic system, and the implantation efficiency is low.
  • the cells that have undergone gene repair have high expression of CD19 protein, reaching a ratio of about 90%, while the expression of CD19 protein in cells without gene repair is significantly reduced, and the expression ratio is less than 5%, which indicates that cells that have undergone gene repair It can express CD19 protein normally and differentiate into B cells normally, but the B cells of cells that have not undergone gene repair have significantly abnormal differentiation.
  • the above results further prove that the gene repaired cells can efficiently rebuild the hematopoietic system of the mouse model.
  • gene-edited cells can quickly and efficiently rebuild the hematopoietic system of the mouse model.
  • the results of determining whether gene editing occurred in the reconstructed mouse model cells are shown in Figure 18.
  • the genome of the cells before transplantation and the bone marrow 16 weeks after transplantation was extracted, the target fragment was amplified, and the gene repair was analyzed by the next-generation sequencing bioinformatics method. Efficiency (HDR) and Indels efficiency (NHEJ).
  • HDR High-efficiency
  • NHEJ Indels efficiency
  • the genome of the bone marrow of the mice after the second transplantation was extracted 12 weeks, the target fragment was amplified, and the gene repair efficiency (HDR) and the Indels efficiency (NHEJ) were analyzed by second-generation sequencing bioinformatics methods.
  • the results showed that the human-derived cells in the bone marrow 12 weeks after transplantation all had high-efficiency gene editing, and the gene repair efficiency was similar to that of the cells before transplantation, about 40%, as shown in Figure 20.
  • the method of the present invention has the following advantages.
  • the method can gene-edit and efficiently repair hiPSC derived from XLSA patients and CD34+HSPC derived from bone marrow, which meets the needs of clinical treatment of X-chain cyclic iron particles.
  • Immature red blood cell anemia treatment requirements second, high gene repair efficiency, significantly increase the expression of ALAS-2 gene and protein, and significantly increase the synthesis of heme in differentiated red blood cells;
  • gene repaired hematopoietic stem cells can efficiently reconstruct models The hematopoietic system of mice; fourth, the cells after gene editing have no potential off-target phenomenon.
  • the method developed by the present invention may replace traditional hematopoietic stem cell transplantation treatment techniques to cure patients with X-chain ring sideroblast anemia.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Physics & Mathematics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided is a method for efficiently repairing the ALAS-2 gene mutation that causes ring sideroblastic anemia by using gene-editing technology, comprising using gene-editing technology to efficiently and safely genetically modify the specific point mutation of the ALAS-2 gene in the hematopoietic stem cells of patients; restoring ALAS-2 gene expression; and restoring ferroheme synthesis and red blood cell maturation to normal levels, so as to achieve the purpose of treating disease.

Description

一种高效修复环状铁粒幼细胞性贫血基因突变的方法Method for efficiently repairing gene mutation of circular sideroblastic anemia 技术领域Technical field
本发明涉及基因编辑治疗领域,具体地,本发明涉及利用基因编辑技术高效修复导致环状铁粒幼红细胞性贫血的基因突变的方法,其包含利用基因编辑技术,高效安全地基因修饰人的造血干细胞的ALAS-2基因特异的点突变,恢复ALAS-2基因表达,实现治疗疾病的目的。The present invention relates to the field of gene editing therapy. Specifically, the present invention relates to a method for efficiently repairing gene mutations that cause ring sideroblastic anemia by using gene editing technology, which includes using gene editing technology to efficiently and safely genetically modify human hematopoiesis The specific point mutation of the ALAS-2 gene of the stem cells restores the expression of the ALAS-2 gene and achieves the purpose of treating diseases.
背景技术Background technique
遗传性铁粒幼红细胞贫血(Congenital sideroblastic anemia,CSA)是一组铁利用障碍性遗传性疾病。特征为骨髓中出现大量环状铁粒幼红细胞,红细胞无效生成,组织铁储量过多和外周血呈小细胞低色素性贫血。目前分子水平检测已经发现了7种疾病突变型,主要导致铁合成、铁硫复合物、线粒体蛋白合成障碍等病症。其中,X链锁铁粒幼红细胞性贫血是最普遍的疾病类型(Kaneko K,et al.Haematologica.2014)。Hereditary sideroblastic anemia (Congenital sideroblastic anemia, CSA) is a group of inherited diseases of iron utilization disorders. It is characterized by the appearance of a large number of ring sideroblasts in the bone marrow, ineffective production of red blood cells, excessive iron reserves in the tissues, and small cell hypochromic anemia in the peripheral blood. At present, molecular level testing has found 7 disease mutations, which mainly lead to iron synthesis, iron-sulfur complex, and mitochondrial protein synthesis disorders. Among them, X-chain sideroblastic anemia is the most common type of disease (Kaneko K, et al. Haematologica. 2014).
X链锁铁粒幼红细胞贫血(X-linked sideroblastic anemia,XLSA)是一种罕见血液系统遗传病,属于CSA的一种,发病率不足X-linked sideroblastic anemia (XLSA) is a rare genetic disease of the blood system, which is a kind of CSA with insufficient incidence
十万分之一。发病人群主要是男性发病者,女性携带者由于正常的等位基因抑制了病态基因的表达,红细胞的异常较少,因而这种患病家族中女性携带者一般无贫血,但红细胞通常呈现明显的双向性。除少数典型病例于出生后或者婴儿时期出现贫血外,大多数与10-20岁左右出现贫血,偶有至50-60岁并发。XLSA患者除了贫血的症状和体征外,铁过载是常见的并发症,尤其在疾病晚期,可导致患者死亡。患者通常由于不规则的输血和排铁导致铁过载,使得肝脏和脾脏轻度甚至重度肿大,肝功能正常或轻度异常。铁过载最危险的临床表现是心率失常,常出现在疾病晚期。贫血严重的幼儿及少年常出现生长发育迟缓(Wakabayashi,et al.Proc Natl Acad Sci USA.2016)。研究发现,XLSA的发病机理主要是由于编码5-氨基乙酰丙酸合酶2(5-aminolevulinic acid synthase 2,ALAS-2)的基因发生突变所致。ALAS-2 是调控红细胞中催化亚铁血红素合成的关键调控因子。迄今为止,在100多个家族的患者中,发现了至少61种ALAS-2外显子5-11中的突变类型,除此以外,在非编码区尤其是内含子-1区域的点突变,即Int-1-GATA(GATA-1和ALAS-2结合的关键区域),也被报道是主要的突变类型之一(Zhang,et al.Nucliec Acids Res.2017;Campagna,et al.Am J hematol.2014)。One hundred thousandth. The affected population is mainly male patients. Female carriers have fewer red blood cell abnormalities due to normal alleles suppressing the expression of diseased genes. Therefore, female carriers in this diseased family generally have no anemia, but red blood cells are usually obvious. Two-way. Except for a few typical cases that develop anemia after birth or infancy, most of them develop anemia around the age of 10-20, and occasionally occur at the age of 50-60. In addition to the symptoms and signs of anemia in XLSA patients, iron overload is a common complication, especially in the late stage of the disease, which can lead to death. Patients are usually overloaded with iron due to irregular blood transfusion and iron excretion, which makes the liver and spleen slightly or even severely enlarged, and the liver function is normal or mildly abnormal. The most dangerous clinical manifestation of iron overload is arrhythmia, which often appears in the late stages of the disease. Children and teenagers with severe anemia often suffer from growth retardation (Wakabayashi, et al. Proc Natl Acad Sci USA. 2016). Studies have found that the pathogenesis of XLSA is mainly due to mutations in the gene encoding 5-aminolevulinic acid synthase 2 (ALAS-2). ALAS-2 is a key regulator that regulates the synthesis of catalyzed heme in red blood cells. So far, in more than 100 families of patients, at least 61 types of mutations in ALAS-2 exons 5-11 have been found. In addition, point mutations in non-coding regions, especially intron-1 regions , Int-1-GATA (the key region where GATA-1 and ALAS-2 bind), has also been reported to be one of the main types of mutations (Zhang, et al. Nucliec Acids Res. 2017; Campagna, et al. Am J hematol.2014).
目前,虽然存在一些针对XLSA的治疗法,例如注射大剂量的维生素B6、长期输血和排铁治疗、异基因造血干细胞移植等方法。然而,长期的高剂量输血伴随去铁剂排铁治疗导致铁过载,患者的脾脏、肝脏、心脏和肾脏等重要脏器铁大量沉积而引起的器官损伤是XLSA患儿死亡的主要原因之一。异基因造血干细胞移植技术虽然能够根治XLSA,但是由于HLA全相合配型比例低和移植术后的GVHD(Graft-Versus-Host-Disease,移植物抗宿主反应)、免疫排斥导致的死亡,因此目前该治疗技术难以满足患者巨大的被治疗的需求。为了解决异源造血干细胞治疗技术的问题,基于基因修饰自体造血干细胞的转基因疗法和基因编辑疗法应运而生。其中,基因编辑疗法的治疗方案是利用基因编辑工具,如CRISPR/Cas9、锌指核酸酶(Zinc Finer Nulease,ZFN)和转录激活因子样效应物核酸酶(transcription activator-like effector nucleases,TALEN)等,编辑患者的自体的造血干细胞,修复ALAS-2基因突变从而恢复ALAS-2基因的表达,再将经过基因修饰的自体的造血干细胞回输给患者,使患者的亚铁血红素合成及红细胞状态恢复至正常水平,达到治疗疾病的目的。At present, although there are some treatments for XLSA, such as injection of large doses of vitamin B6, long-term blood transfusion and iron excretion treatment, allogeneic hematopoietic stem cell transplantation and other methods. However, long-term high-dose blood transfusions accompanied by iron-removal treatment with iron removal agents lead to iron overload, and organ damage caused by large amounts of iron deposition in important organs such as the spleen, liver, heart, and kidneys is one of the main causes of death in children with XLSA. Although allogeneic hematopoietic stem cell transplantation technology can cure XLSA, it is currently due to the low proportion of HLA homologous matching type, GVHD (Graft-Versus-Host-Disease, Graft-Versus-Host-Disease, Graft-Versus-Host-Disease) and death caused by immune rejection after transplantation. This treatment technology is difficult to meet the huge needs of patients to be treated. In order to solve the problem of heterologous hematopoietic stem cell therapy technology, transgene therapy and gene editing therapy based on genetically modified autologous hematopoietic stem cells have emerged. Among them, the treatment plan of gene editing therapy is to use gene editing tools, such as CRISPR/Cas9, zinc finger nuclease (Zinc Finer Nulease, ZFN), and transcription activator-like effector nuclease (transcription activator-like effector nucleases, TALEN), etc. Edit the patient’s autologous hematopoietic stem cells, repair the ALAS-2 gene mutation to restore the expression of the ALAS-2 gene, and then return the genetically modified autologous hematopoietic stem cells to the patient to make the patient’s heme synthesis and red blood cell status Return to normal level to achieve the purpose of curing diseases.
基因编辑技术是人为的利用编码的核酸酶在DNA序列上特定位点进行插入、敲除以及突变使得基因序列发生改变的基因重组技术。基因编辑工具首先会识别基因组中特定的序列,通过核酸酶产生DNA双链断裂缺口,依赖内源的修复机制非同源末端连接(non-homologous end joining,NHEJ)和同源重组(Homology-directed repair,HDR)两种修复机制修复。前者通过细胞复制和修复会在基因组中引入片段插入和缺失(Insertion and deletions,INDELs),产生突变;后者由于添加了外源的供体核酸作为模板精确修复基因组序列(Dever,et al.Nature.2016)。CRISPR/Cas9作为最新的基因编辑系统,具有操作简单、成本低、可开发空间大等优点,极大地提高了基因编辑可操作性和工作效率(Cong,et al.Science.2013;Jinek,et al.Science.2012)。Gene editing technology is a genetic recombination technology that artificially uses encoded nucleases to insert, knock out, and mutate at specific sites in the DNA sequence to change the gene sequence. Gene editing tools will first identify specific sequences in the genome, generate DNA double-strand break gaps through nucleases, and rely on endogenous repair mechanisms. Non-homologous end joining (NHEJ) and homologous recombination (Homology-directed) repair, HDR) two repair mechanisms to repair. The former introduces insertions and deletions (INDELs) into the genome through cell replication and repair, resulting in mutations; the latter precisely repairs the genome sequence due to the addition of foreign donor nucleic acid as a template (Dever, et al. Nature). .2016). As the latest gene editing system, CRISPR/Cas9 has the advantages of simple operation, low cost, and large development space, which greatly improves the operability and work efficiency of gene editing (Cong, et al. Science. 2013; Jinek, et al. .Science.2012).
然而,虽然针对XLSA的疾病遗传学和发病机理等研究相对成熟,但是 迄今为止,国内外并未报道针对该疾病有效的治疗方案,而利用基因编辑技术开发全新的针对该疾病的细胞治疗技术则为XLSA的疾病治疗带来新的希望。However, although the genetics and pathogenesis of the disease for XLSA are relatively mature, so far, no effective treatment plan for the disease has been reported at home and abroad, and the use of gene editing technology to develop a new cell therapy technology for the disease is Bringing new hope for XLSA disease treatment.
发明概述Summary of the invention
本发明首次利用基因编辑技术,例如CRISPR/Cas9基因编辑技术,开发出了新一代的造血干细胞,成功高效修复了XLSA病人骨髓来源的造血干细胞中ALAS-2基因的点突变,基因修复效率高达约30%-40%,经过基因修复的细胞的ALAS-2基因表达达到健康供者ALAS-2表达的50%左右,从而促进了造血干细胞向成熟红细胞的分化。同时,通过实验证明经过基因修复的造血干细胞能够快速、高效地植入小鼠模型的造血系统,而且植入后的细胞体内分化功能正常,能够实现造血系统的重建。The present invention uses gene editing technology for the first time, such as CRISPR/Cas9 gene editing technology, to develop a new generation of hematopoietic stem cells, and successfully and efficiently repair the point mutation of the ALAS-2 gene in the hematopoietic stem cells derived from the bone marrow of XLSA patients. The gene repair efficiency is as high as approx. 30%-40%, the ALAS-2 gene expression of the gene repaired cells reaches about 50% of the ALAS-2 expression of healthy donors, thereby promoting the differentiation of hematopoietic stem cells into mature red blood cells. At the same time, experiments have proved that the hematopoietic stem cells that have undergone gene repair can be quickly and efficiently implanted into the hematopoietic system of the mouse model, and the differentiation function of the implanted cells is normal in vivo, which can realize the reconstruction of the hematopoietic system.
因此,本申请在一方面,提供了一种通过基因编辑来校正干细胞染色体中5-氨基乙酰丙酸合酶2(ALAS-2)基因突变的方法,其中所述基因编辑包括:Therefore, in one aspect, the present application provides a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations in stem cell chromosomes through gene editing, wherein the gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
一方面,本申请提供了一种通过基因编辑来校正ALAS-2基因突变,从而增加功能性ALAS-2表达的方法,其中所述基因编辑包括:In one aspect, the present application provides a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2, wherein the gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
一方面,本申请提供了一种通过基因编辑来校正干细胞染色体中的ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生的方法,其中所述基因编辑包括:On the one hand, the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing functional ALAS-2 expression, thereby increasing heme production in cells derived from the hematopoietic stem cells, wherein The gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
一方面,本申请提供了一种通过基因编辑来校正干细胞染色体中的ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生,由此促进所述造血干细胞成熟的方法,其中所述基因编辑包括:On the one hand, the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing functional ALAS-2 expression, thereby increasing heme production in cells derived from the hematopoietic stem cells, thereby promoting The method for maturation of hematopoietic stem cells, wherein the gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
一方面,本申请提供了一种通过基因编辑来校正干细胞染色体中的ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生,促进所述造血干细胞成熟,由此来治疗个体的贫血(例如XLSA)的方法,其中所述基因编辑包括:On the one hand, the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of hemoglobin in the cells derived from the hematopoietic stem cells, and promoting the A method for the maturation of hematopoietic stem cells to treat anemia (such as XLSA) in an individual, wherein the gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
一方面,本申请提供了一种通过基因编辑来校正干细胞染色体中的ALAS-2基因突变,增加功能性ALAS-2表达的方法,其中所述基因编辑不在所述造血干细胞基因组造成脱靶或脱靶率低于1%,例如低于0.5%或低于0.1%,其中所述基因编辑包括:On the one hand, the present application provides a method for correcting ALAS-2 gene mutations in stem cell chromosomes through gene editing and increasing functional ALAS-2 expression, wherein said gene editing does not cause off-target or off-target rate in the genome of said hematopoietic stem cells Less than 1%, such as less than 0.5% or less than 0.1%, wherein the gene editing includes:
(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(a) introducing donor DNA containing a single-stranded correction sequence corresponding to the mutation sequence into the hematopoietic stem cell;
(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。(b) The sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction sequence on the donor DNA replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation.
在上述方法的一些实施方案中,所述造血干细胞是CD34 +造血干细胞和 祖细胞(“HSPC”),或人诱导多能干细胞(hiPSC)。 In some embodiments of the above methods, the hematopoietic stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
在上述方法的一些实施方案中,所述造血干细胞从贫血患者,例如铁粒幼红细胞贫血患者,具体地,遗传性铁粒幼红细胞贫血患者,更具体地,XLSA患者得到。In some embodiments of the above methods, the hematopoietic stem cells are obtained from anemia patients, such as sideroblastic anemia patients, specifically, hereditary sideroblastic anemia patients, more specifically, XLSA patients.
在上述方法的一些实施方案中,所述突变位于ALAS-2基因的外显子5-11或内含子-1中。In some embodiments of the above method, the mutation is located in exon 5-11 or intron-1 of the ALAS-2 gene.
在上述方法的一些实施方案中,所述突变位于ALAS-2基因的内含子-1中。In some embodiments of the above methods, the mutation is located in intron-1 of the ALAS-2 gene.
在上述方法的一些实施方案中,所述突变是Int-1-GATA。In some embodiments of the above methods, the mutation is Int-1-GATA.
在上述方法的一些实施方案中,所述序列特异性核酸酶选自下组:RNA引导核酸酶、锌指核酸酶(ZFN)和转录激活因子样效应物核酸酶(TALEN)。In some embodiments of the above methods, the sequence-specific nuclease is selected from the group consisting of RNA guide nuclease, zinc finger nuclease (ZFN), and transcription activator-like effector nuclease (TALEN).
在上述方法的一些实施方案中,所述序列特异性核酸酶是RNA引导核酸酶。In some embodiments of the above methods, the sequence-specific nuclease is an RNA guide nuclease.
在上述方法的一些实施方案中,所述RNA引导核酸酶是Cas。In some embodiments of the above methods, the RNA guide nuclease is Cas.
在上述方法的一些实施方案中,所述RNA引导核酸酶是Cas9。In some embodiments of the above methods, the RNA guide nuclease is Cas9.
在上述方法的一些实施方案中,进一步包括将识别ALAS-2基因的引导RNA(sgRNA)引入到所述CD34 +HSPC中。 In some embodiments of the above method, it further comprises introducing a guide RNA (sgRNA) that recognizes the ALAS-2 gene into the CD34 + HSPC.
在上述方法的一些实施方案中,核酸酶剪切位点距离所述突变位点不超过约20个核苷酸的位点,例如,约15个,13个,12个,11个,约10个,约9个,约8个,约7个,约6个,约5个,约4个,约3个,约2个,约1个核苷酸,或核酸酶剪切位点与所述突变位点重合。In some embodiments of the above methods, the nuclease cleavage site is not more than about 20 nucleotides away from the mutation site, for example, about 15, 13, 12, 11, or about 10 nucleotides. About 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, about 1 nucleotide, or nuclease cleavage site and the The mutation sites overlap.
在上述方法的一些实施方案中,所述sgRNA与所述染色体上的突变位点染色体序列互补或与所述染色体上的突变位点邻近的染色体序列互补。In some embodiments of the above method, the sgRNA is complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome.
在上述方法的一些实施方案中,所述sgRNA中的指导序列为约10个至约25个,约12个至约24个,约14个至约23个,约16个至约22个,约17个至约21个核苷酸长。在上述方法的一些具体实施方案中,所述sgRNA中的指导序列为20个核苷酸长。In some embodiments of the above method, the guide sequences in the sgRNA are about 10 to about 25, about 12 to about 24, about 14 to about 23, about 16 to about 22, about 17 to about 21 nucleotides long. In some specific embodiments of the above method, the guide sequence in the sgRNA is 20 nucleotides long.
在上述方法的一些实施方案中,所述sgRNA是经化学修饰的。在一些实施方案中所述sgRNA是经过核苷酸核糖2’-O-甲基化修饰和/或核苷酸间3’硫代磷酸酯化修饰(也可简称为硫代磷酸化修饰)的。在一些实施方案中所述sgRNA的5’端前一个、二个和/或三个碱基和/或3’端的最后一个核苷酸 核糖碱基的2’-O-甲基化修饰。在一些实施方案中,所述sgRNA包含5’端的前3个核苷酸和3’端后3个核苷酸核糖的2’-O-甲基化修饰和/或核苷酸间的3’硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端前3个和3’端后3个核苷酸中包含核苷酸核糖的2′-O-甲基化修饰且在5’端的前3个和3’端后3个核苷酸间连接中包含硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端的前5个核苷酸及3’端的后5个核苷酸的核糖中包含2’-O-甲基化修饰,并且在5’端的前5个及3’端的后5个核苷酸间连接中包含硫代磷酸酯化修饰。In some embodiments of the above methods, the sgRNA is chemically modified. In some embodiments, the sgRNA is modified by nucleotide ribose 2'-O-methylation and/or internucleotide 3'phosphorothioate modification (also referred to as phosphorothioate modification) . In some embodiments, one, two and/or three bases before the 5'end of the sgRNA and/or the last nucleotide ribose base at the 3'end are 2'-O-methylated modifications. In some embodiments, the sgRNA comprises the first 3 nucleotides at the 5'end and the 3 nucleotides after the 3'end. 2'-O-methylation modification of ribose and/or an internucleotide 3' Phosphorothioate modification. In some embodiments, the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end The connection between the 3 nucleotides after the 3'end contains phosphorothioate modification. In some embodiments, the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end The last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
在上述方法的一些实施方案中,所述sgRNA包含与ALAS-2基因的内含子-1中的序列互补的核酸序列。In some embodiments of the above method, the sgRNA comprises a nucleic acid sequence complementary to a sequence in intron-1 of the ALAS-2 gene.
在上述方法的一些实施方案中,所述sgRNA中包含的与ALAS-2基因的内含子-1中的序列互补的核酸序列选自下组:SEQ ID NO:1-SEQ ID NO:3。In some embodiments of the above method, the nucleic acid sequence that is complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA is selected from the following group: SEQ ID NO: 1-SEQ ID NO: 3.
在上述方法的一些实施方案中,通过电穿孔和转导将所述sgRNA引入到所述造血干细胞中。In some embodiments of the above methods, the sgRNA is introduced into the hematopoietic stem cells by electroporation and transduction.
在上述方法的一些实施方案中,所述供体DNA是环状的。In some embodiments of the above methods, the donor DNA is circular.
在上述方法的一些实施方案中,所述供体DNA是线性的。In some embodiments of the above methods, the donor DNA is linear.
在上述方法的一些实施方案中,所述供体DNA是ssODN。In some embodiments of the above methods, the donor DNA is ssODN.
在上述方法的一些实施方案中,所述供体DNA包含5’端磷酸化修饰,并且其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰。在上述方法的一些实施方案中,所述供体DNA包含5’端磷酸化修饰,其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰,并且5’端的前三个核苷酸和3’端的最后3个核苷酸还包含核糖的2’-O-甲基化修饰。In some embodiments of the above method, the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end. . In some embodiments of the above method, the donor DNA includes a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, And the first three nucleotides at the 5'end and the last three nucleotides at the 3'end also contain 2'-O-methylation modification of ribose.
在上述方法的一些实施方案中,所述校正序列具有与所述突变序列相同的长度。在上述方法的一些实施方案中,所述供体DNA与所述校正序列长度相等。在上述方法的一些实施方案中,所述供体DNA长于所述校正序列。In some embodiments of the above methods, the correction sequence has the same length as the mutation sequence. In some embodiments of the above methods, the donor DNA and the correction sequence are equal in length. In some embodiments of the above methods, the donor DNA is longer than the correction sequence.
在上述方法的一些实施方案中,所述校正序列为约50个至约300个,约60个至约250个,约60个至约240个,约60个至约230个,约60个至约220个,约60个至约210个,约60个至约200个核苷酸长。In some embodiments of the above method, the correction sequence is about 50 to about 300, about 60 to about 250, about 60 to about 240, about 60 to about 230, about 60 to about About 220, about 60 to about 210, about 60 to about 200 nucleotides long.
在上述方法的一些实施方案中,所述校正序列包含与位于所述突变位点的3'端的靶区域基本上互补的5'臂,和与位于所述突变位点的5'端的靶区域基 本上互补的3'臂。在一些具体实施方案中,所述校正序列的5'臂或3'臂分别与所述突变位点的3'端的靶区域或5'端的靶区域具有至少约85%的同源性,至少约90%,至少约95%,至少约96%,至少约97%,至少约98%,至少约99%的同源性。在上述一些具体实施方案中,所述校正序列的5'臂或3'臂分别与所述突变位点的3'端的靶区域或5'端的靶区域具有100%的同源性。In some embodiments of the above method, the correction sequence comprises a 5'arm that is substantially complementary to a target region located at the 3'end of the mutation site, and a 5'arm that is substantially complementary to a target region located at the 5'end of the mutation site. Complementary 3'arms on top. In some specific embodiments, the 5'arm or 3'arm of the correction sequence has at least about 85% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively, at least about 85%. 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% homology. In some of the above specific embodiments, the 5'arm or 3'arm of the correction sequence has 100% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively.
在上述方法的一些实施方案中,所述校正序列的5'臂为约30个至约100个核苷酸长,例如,约35个至约80个,约40个至约70个,约40个至约60个核苷酸长。In some embodiments of the above methods, the 5'arms of the correction sequence are about 30 to about 100 nucleotides long, for example, about 35 to about 80, about 40 to about 70, about 40. From about 60 nucleotides in length.
在上述方法的一些实施方案中,所述校正序列的3'臂为约20个至约100个核苷酸长,例如,约20个至约80个,约20个至约70个,约20个至约60个,约20个至约50个核苷酸长。In some embodiments of the above methods, the 3'arms of the correction sequence are about 20 to about 100 nucleotides long, for example, about 20 to about 80, about 20 to about 70, about 20. From about 20 to about 50 nucleotides in length.
在上述方法的一些实施方案中,所述校正序列的5'臂比所述校正序列的3'臂长。In some embodiments of the above method, the 5'arm of the correction sequence is longer than the 3'arm of the correction sequence.
在上述方法的一些实施方案中,所述校正序列的3'臂比所述校正序列的5'臂长。In some embodiments of the above methods, the 3'arm of the correction sequence is longer than the 5'arm of the correction sequence.
在上述方法的一些实施方案中,所述校正序列的5'臂和所述校正序列的3'臂具有相同的长度。In some embodiments of the above method, the 5'arm of the correction sequence and the 3'arm of the correction sequence have the same length.
在上述方法的一些实施方案中,所述校正序列与除突变位点外的ChrX:55028172-55028268处的靶序列互补。In some embodiments of the above method, the correction sequence is complementary to the target sequence at ChrX:55028172-55028268 except for the mutation site.
在上述方法的一些实施方案中,所述校正序列当包含编码序列时,编码除突变位点以外的与所述突变序列编码的氨基酸序列相同的氨基酸序列。In some embodiments of the above method, when the correction sequence includes a coding sequence, it encodes an amino acid sequence that is the same as the amino acid sequence encoded by the mutation sequence except for the mutation site.
在上述方法的一些实施方案中,所述对应于所述突变的校正序列为SEQ ID NO:4。In some embodiments of the above method, the correction sequence corresponding to the mutation is SEQ ID NO: 4.
在上述方法的一些实施方案中,通过电穿孔将所述供体DNA引入到所述造血干细胞中。In some embodiments of the above methods, the donor DNA is introduced into the hematopoietic stem cells by electroporation.
在上述方法的一些实施方案中,引入所述序列特异性核酸酶包括将编码所述序列特异性核酸酶的mRNA引入到干细胞中。In some embodiments of the above methods, introducing the sequence-specific nuclease includes introducing mRNA encoding the sequence-specific nuclease into stem cells.
在上述方法的一些实施方案中,通过电穿孔将编码所述序列特异性核酸酶的mRNA引入到所述造血干细胞中。In some embodiments of the above methods, the mRNA encoding the sequence-specific nuclease is introduced into the hematopoietic stem cell by electroporation.
在上述方法的一些实施方案中,将编码所述序列特异性核酸酶的mRNA和所述供体DNA同时引入到干细胞中。In some embodiments of the above methods, the mRNA encoding the sequence-specific nuclease and the donor DNA are simultaneously introduced into the stem cell.
在上述方法的一些实施方案中,将sgRNA引入到干细胞中,并且其中将编码所述序列特异性核酸酶的mRNA和所述sgRNA同时引入到干细胞中。In some embodiments of the above methods, sgRNA is introduced into stem cells, and wherein the mRNA encoding the sequence-specific nuclease and the sgRNA are simultaneously introduced into the stem cells.
在上述方法的一些实施方案中,将所述sgRNA和所述供体DNA同时引入到干细胞中。In some embodiments of the above methods, the sgRNA and the donor DNA are simultaneously introduced into the stem cell.
在上述方法的一些实施方案中,将所述sgRNA、编码所述序列特异性核酸酶的mRNA、所述供体DNA分别或同时通过电穿孔或转导的方式引入到干细胞中。In some embodiments of the above methods, the sgRNA, the mRNA encoding the sequence-specific nuclease, and the donor DNA are separately or simultaneously introduced into the stem cells by means of electroporation or transduction.
在上述方法的一些实施方案中,所述sgRNA与所述供体DNA的重量比为约1:12至约12:1,例如,约1:11至约11:1,约1:10至约10:1,约1:9至约9:1,约1:8至约8:1,约1:7至约7:1,约1:6至约6:1。In some embodiments of the above method, the weight ratio of the sgRNA to the donor DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11:1, about 1:10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1.
在上述方法的一些实施方案中,编码所述序列特异性核酸酶的mRNA与所述单链DNA的重量比为约1:12至约12:1,例如,约1:11至约11:1,约1:10至约10:1,约1:9至约9:1,约1:8至约8:1,约1:7至约7:1,约1:6至约6:1。In some embodiments of the above method, the weight ratio of the mRNA encoding the sequence-specific nuclease to the single-stranded DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11:1 , About 1:10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1 .
在上述方法的一些实施方案中,所述造血干细胞从雄性个体得到。In some embodiments of the above methods, the hematopoietic stem cells are obtained from a male individual.
在上述方法的一些实施方案中,所述CD34 +HSPC从雄性个体得到。 In some embodiments of the above methods, the CD34 + HSPC is obtained from a male individual.
在上述方法的一些实施方案中,所述人诱导多能干细胞(hiPSC)从雄性个体得到。In some embodiments of the above methods, the human induced pluripotent stem cells (hiPSC) are obtained from male individuals.
在上述方法的一些实施方案中,所述造血干细胞从雌性个体得到。In some embodiments of the above methods, the hematopoietic stem cells are obtained from a female individual.
在上述方法的一些实施方案中,所述CD34 +HSPC从雌性个体得到。 In some embodiments of the above methods, the CD34 + HSPC is obtained from a female individual.
在上述方法的一些实施方案中,所述人诱导多能干细胞(hiPSC)从雌性个体得到。本申请还涉及通过上述方法获得的经过基因编辑的CD34 +HSPC或人诱导多能干细胞(hiPSC),其中所述CD34 +HSPC或人诱导多能干细胞(hiPSC)来源于贫血患者且经过基因编辑,所述ALAS-2基因的突变得到修正。在一些实施方案中,所述贫血为铁粒幼红细胞贫血,例如遗传性铁粒幼红细胞贫血,具体地,为XLSA。在一些实施方案中,所述ALAS-2基因突变位于ALAS-2基因的外显子5-11或内含子-1中。在一些实施方案中,所述突变是Int-1-GATA。 In some embodiments of the above methods, the human induced pluripotent stem cells (hiPSC) are obtained from a female individual. This application also relates to a gene-edited CD34 + HSPC or human induced pluripotent stem cell (hiPSC) obtained by the above method, wherein the CD34 + HSPC or human induced pluripotent stem cell (hiPSC) is derived from an anemia patient and has been gene-edited, The mutation of the ALAS-2 gene has been corrected. In some embodiments, the anemia is sideroblast anemia, such as hereditary sideroblast anemia, specifically, XLSA. In some embodiments, the ALAS-2 gene mutation is located in exon 5-11 or intron-1 of the ALAS-2 gene. In some embodiments, the mutation is Int-1-GATA.
本申请涉及以下实施方案:This application involves the following implementation schemes:
1.一种通过CRISPS/Cas9基因编辑校正造血干细胞的5-氨基乙酰丙酸合酶2(ALAS-2)基因突变的方法,包括:将包含对应于ALAS-2突变序列的单链校正序列的供体DNA、识别ALAS-2突变序列的sgRNA和编码Cas9蛋白的核 酸序列导入所述造血干细胞,由此所述供体DNA中的校正序列替代所述造血干细胞中的ALAS-2突变序列。1. A method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations of hematopoietic stem cells by CRISPS/Cas9 gene editing, comprising: adding a single-stranded correction sequence corresponding to the ALAS-2 mutation sequence The donor DNA, the sgRNA that recognizes the ALAS-2 mutation sequence, and the nucleic acid sequence encoding the Cas9 protein are introduced into the hematopoietic stem cell, whereby the correction sequence in the donor DNA replaces the ALAS-2 mutation sequence in the hematopoietic stem cell.
2.技术方案1的方法,其中所述造血干细胞是CD34 +HSPC。 2. The method of technical solution 1, wherein the hematopoietic stem cells are CD34 + HSPC.
3.技术方案1或2的方法,其中所述ALAS-2突变序列是ALAS-2基因外显子5-11中的突变序列和/或ALAS-2基因内含子-1中的突变序列。3. The method of technical solution 1 or 2, wherein the ALAS-2 mutant sequence is a mutant sequence in exons 5-11 of the ALAS-2 gene and/or a mutant sequence in intron-1 of the ALAS-2 gene.
4.技术方案1-3任一项的方法,其中所述ALAS-2突变序列位于ALAS-2基因内含子-1中。4. The method of any one of technical solutions 1-3, wherein the ALAS-2 mutant sequence is located in intron-1 of the ALAS-2 gene.
5.技术方案4的方法,其中所述突变为ALAS2内含子-1中的点突变:X:55054635[Chr X(GRCh37/hg19):g.55054635A>G,NM 000032.4:c.-15–2187T>C。5. The method of technical solution 4, wherein the mutation is a point mutation in ALAS2 intron-1: X:55054635[Chr X(GRCh37/hg19):g.55054635A>G,NM000032.4:c.-15– 2187T>C.
6.技术方案4或5的方法,其中Cas9剪切位点距离ALAS-2突变位点不超过约11个核苷酸的位点。6. The method of technical solution 4 or 5, wherein the Cas9 cleavage site is not more than about 11 nucleotides away from the ALAS-2 mutation site.
7.技术方案1-6中任一项的方法,其中所述sgRNA为约17个至约20个核苷酸长。7. The method of any one of technical solutions 1-6, wherein the sgRNA is about 17 to about 20 nucleotides in length.
8.技术方案7的方法,其中所述sgRNA是经化学修饰的。8. The method of technical solution 7, wherein the sgRNA is chemically modified.
9.技术方案8的方法,其中所述sgRNA的修饰包括核苷酸核糖上的2’-O-甲基化修饰或核苷酸间的3’硫代磷酸化修饰或二者。9. The method of technical solution 8, wherein the modification of the sgRNA includes a 2'-O-methylation modification on the nucleotide ribose or an internucleotide 3'phosphorothioate modification or both.
10.技术方案9的方法,其中所述修饰为5’端的前三个核苷酸核糖上的2’-O-甲基化修饰、3’端的最后三个核苷酸核糖上的2’-O-甲基化修饰、5’端的前三个核苷酸的核苷酸间3’硫代磷酸化修饰和3’端的最后三个核苷酸的核苷酸间3’硫代磷酸化修饰。10. The method of technical solution 9, wherein the modification is a 2'-O-methylation modification on the first three nucleotides ribose at the 5'end, and a 2'-O-methylation modification on the last three nucleotides ribose at the 3'end. O-methylation modification, internucleotide 3'phosphorothioate modification of the first three nucleotides at the 5'end and internucleotide 3'phosphorothioate modification of the last three nucleotides at the 3'end .
11.技术方案1-10任一项的方法,其中所述sgRNA的序列选自下组:SEQ ID NO:1、SEQ ID NO:2和SEQ ID NO:3。11. The method of any one of technical solutions 1-10, wherein the sequence of the sgRNA is selected from the following group: SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3.
12.技术方案1-11中任一项的方法,其中所述校正序列为约60个至约200个核苷酸长。12. The method of any one of technical solutions 1-11, wherein the correction sequence is about 60 to about 200 nucleotides long.
13.技术方案12的方法,其中所述校正序列包含与位于所述突变位点的3'的靶区域互补的5'臂,和与位于所述突变位点的5'的靶区域互补的3'臂,其中所述校正序列的5'臂为约40个至约60个核苷酸长,校正序列的3'臂为约20个至约50个核苷酸长。13. The method of technical solution 12, wherein the correction sequence comprises a 5'arm complementary to the target region 3'of the mutation site, and a 3'arm complementary to the target region 5'of the mutation site. 'Arm, wherein the 5'arm of the calibration sequence is about 40 to about 60 nucleotides long, and the 3'arm of the calibration sequence is about 20 to about 50 nucleotides long.
14.技术方案13的方法,其中所述供体DNA是经过化学修饰的。14. The method of technical solution 13, wherein the donor DNA is chemically modified.
15.技术方案14的方法,其中所述化学修饰包括核苷酸核糖上的2’-O- 甲基化修饰或核苷酸间的3’硫代磷酸化修饰或二者。15. The method of claim 14, wherein the chemical modification includes 2'-O-methylation modification on nucleotide ribose or internucleotide 3'phosphorothioate modification or both.
16.技术方案15的方法,其中所述修饰为5’端的前三个核苷酸的核苷酸间3’硫代磷酸化修饰和3’端的最后三个核苷酸的核苷酸间3’硫代磷酸化修饰。16. The method of technical solution 15, wherein the modification is an internucleotide 3'phosphorothioate modification of the first three nucleotides at the 5'end and an internucleotide 3'phosphorothioate modification of the last three nucleotides at the 3'end 'Phosphorothioate modification.
17.技术方案15或16的方法,其中所述修饰还包括5’端磷酸化修饰。17. The method of technical solution 15 or 16, wherein the modification further comprises 5'end phosphorylation modification.
18.技术方案17的方法,其中校正序列与除所述突变位点外的ChrX:55028172-55028268处的靶序列互补。18. The method of technical solution 17, wherein the correction sequence is complementary to the target sequence at ChrX:55028172-55028268 except for the mutation site.
19.技术方案1-18任一项的方法,其中所述供体DNA序列如SEQ ID NO:4所示。19. The method of any one of technical solutions 1-18, wherein the donor DNA sequence is shown in SEQ ID NO: 4.
20.技术方案1-19中任一项的方法,其中通过电穿孔或转导方式将所述sgRNA、供体DNA和编码Cas9蛋白的核酸序列导入到所述造血干细胞中。20. The method of any one of technical solutions 1-19, wherein the sgRNA, the donor DNA and the nucleic acid sequence encoding the Cas9 protein are introduced into the hematopoietic stem cell by electroporation or transduction.
21.技术方案1-20中任一项的方法,其中所述sgRNA与所述供体DNA的重量比为约4:12。21. The method of any one of technical solutions 1-20, wherein the weight ratio of the sgRNA to the donor DNA is about 4:12.
22.技术方案1-21中任一项的方法,其中编码所述Cas9的mRNA与所述供体DNA的重量比为约4:12。22. The method of any one of technical solutions 1-21, wherein the weight ratio of the mRNA encoding the Cas9 to the donor DNA is about 4:12.
23.技术方案1-22中任一项的方法,其中向约1.0*10^6个造血干细胞中导入Cas9 mRNA、sgRNA、供体DNA的重量选自如下任一组:23. The method of any one of technical solutions 1-22, wherein the weight of Cas9 mRNA, sgRNA, and donor DNA introduced into about 1.0*10^6 hematopoietic stem cells is selected from any of the following groups:
1)6μg、4μg、6μg;1) 6μg, 4μg, 6μg;
2)6μg、4μg、8μg;2) 6μg, 4μg, 8μg;
3)6μg、4μg、10μg;3) 6μg, 4μg, 10μg;
4)6μg、4μg、12μg。4) 6μg, 4μg, 12μg.
24.技术方案1-22中任一项的方法,其中Cas9 mRNA:sgRNA:供体DNA的重量比为1:1:1。24. The method of any one of technical solutions 1-22, wherein the weight ratio of Cas9 mRNA: sgRNA: donor DNA is 1:1:1.
25.技术方案20-24中任一项所述的方法,其中所述Cas9 mRNA、sgRNA、供体DNA通过电穿孔导入造血干细胞中,所述电穿孔条件为300V,1ms。25. The method of any one of technical solutions 20-24, wherein the Cas9 mRNA, sgRNA, and donor DNA are introduced into hematopoietic stem cells by electroporation, and the electroporation conditions are 300V, 1ms.
附图说明Description of the drawings
图1针对人X染色体上ALAS-2基因内含子-1的点突变(X:55054635[Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C)附近位置设计的多条sgRNA和供体模板单链DNA的示意图。Figure 1 Point mutations in intron-1 of ALAS-2 gene on human X chromosome (X:55054635[ChrX(GRCh37/hg19):g.55054635A>G,NM 000032.4:c.-15-2187T> C) Schematic diagram of multiple sgRNAs and donor template single-stranded DNA designed at nearby locations.
图2针对人X染色体上ALAS-2基因内含子-1的点突变(X:55054635 [Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C)附近位置设计的多条sgRNA和供体模板单链DNA的序列信息。Figure 2 Point mutations in intron-1 of ALAS-2 gene on human X chromosome (X:55054635 [Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15-2187 T> C) Sequence information of multiple sgRNAs and donor template single-stranded DNA designed at nearby locations.
图3电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的3个sgRNA,分别是sgRNA-1,sgRNA-2和sgRNA-3进入XLSA病人来源的诱导性多潜能干细胞(human induced pluripotent stem cells,hiPSCs),4天后,扩增目的片段和一代测序,插入缺失频率(Indels frequency)通过“Synthego ICE Analysis”在线软件分析产生插入缺失频率的统计分析,n=3个实验重复。Figure 3 Electrotransformation of Cas9 mRNA and the 3 sgRNAs near the point mutation of ALAS-2 intron-1, namely sgRNA-1, sgRNA-2 and sgRNA-3 into human-induced pluripotent stem cells derived from XLSA patients pluripotent stem cells, hiPSCs), 4 days later, the target fragment was amplified and first-generation sequencing, the indel frequency (Indels frequency) was analyzed by the "Synthego ICE Analysis" online software to generate a statistical analysis of the indel frequency, n=3 experimental replicates.
图4电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1,按照不同Cas9和sgRNA的添加量进入XLSA病人来源的hiPSCs,4天后,扩增目的片段和一代测序,插入缺失频率通过“Synthego ICE Analysis”在线软件分析产生,n=3个实验重复。Figure 4 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 into hiPSCs derived from XLSA patients according to the amount of Cas9 and sgRNA added. After 4 days, the target fragment was amplified and first-generation sequencing. The frequency of indels was generated by the "Synthego ICE Analysis" online software analysis, n=3 experimental replicates.
图5电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN,按照不同Cas9、sgRNA和ssODN的添加量进入XLSA病人来源的hiPSCs,4天后,扩增目的片段和NGS,通过生物信息学方法分析NHEJ和HDR的比例。其中,NHEJ:非同源末端连接(non-homologous end joining),代表插入缺失比例,HDR:同源介导修复(Homology-directed repair),代表基因修复的比例,n=3个实验重复。Figure 5 Electrotransform Cas9 mRNA, sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN, enter the hiPSCs derived from XLSA patients according to the addition amount of Cas9, sgRNA and ssODN, and expand after 4 days Increase the target fragment and NGS, and analyze the ratio of NHEJ and HDR through bioinformatics methods. Among them, NHEJ: non-homologous end joining, representing the ratio of indels, HDR: Homology-directed repair, representing the ratio of gene repair, n=3 experimental replicates.
图6电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN,按照不同Cas9、sgRNA和ssODN的添加量进入XLSA病人骨髓来源的CD34+HSPC,4天后,扩增目的片段和NGS,通过生物信息学方法分析NHEJ和HDR的比例。其中,NHEJ:代表插入缺失比例,HDR:代表基因修复的比例,n=3个实验重复。Figure 6 Electrotransformation of Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN, according to different addition amounts of Cas9, sgRNA and ssODN, enter the bone marrow-derived CD34+HSPC of XLSA patients, 4 After days, the target fragment and NGS were amplified, and the ratio of NHEJ and HDR was analyzed by bioinformatics methods. Among them, NHEJ: represents the ratio of indels, HDR: represents the ratio of gene repair, n=3 experimental replicates.
图7电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,4天后,扩增目的片段和NGS,通过生物信息学方法分析NHEJ和HDR的比例。其中,NHEJ代表Indels比例,HDR:代表基因修复的比例,n=3个实验重复。Figure 7: Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients. After 4 days, the target fragment and NGS are amplified, and the target fragment and NGS are amplified. Informatics methods analyze the ratio of NHEJ and HDR. Among them, NHEJ represents the ratio of Indels, HDR: represents the ratio of gene repair, n=3 experimental replicates.
图8电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC2天后进行体外克隆形成实验(CFU检测),14天后统计不同血液系统的克隆数目,BFU-E、CFU-GM、CFU-E、CFU-GEMM代表红系、髓系、淋巴系等血液系统不同谱系的克隆形成。其中,健康供者:代表未经过基因编辑的健康供者,空白对 照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞,n=3个实验重复。Figure 8 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+HSPC derived from the bone marrow of XLSA patients for 2 days, and in vitro clone formation experiment (CFU detection), 14 days later Count the number of clones in different blood systems, BFU-E, CFU-GM, CFU-E, CFU-GEMM represent the formation of clones of different blood system lineages such as erythroid, myeloid, and lymphatic systems. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, gene repair: represent cells that have undergone gene repair, n=3 experimental replicates.
图9电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,进行红细胞分化,分别检测分化后第7天、第13天和第18天人CD71和人CD235a两个膜蛋白表达比例,代表红系分化效率。其中,健康供者:代表未经过基因编辑的健康供者,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。Figure 9: Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. The 7th day after differentiation was detected. The expression ratio of human CD71 and human CD235a membrane proteins on the 13th and 18th days represents the efficiency of erythroid differentiation. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, and gene repair: represent cells that have undergone gene repair.
图10电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,进行红细胞分化,18天后,图A是分化后后细胞的拍照图。图B是Benzidine染色明场示意图。图C是Wright-Giemsa染色明场示意图。标尺:20um。其中,健康供者:代表未经过基因编辑的健康供者,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。Figure 10 Electrotransformation of Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from bone marrow of XLSA patients for red blood cell differentiation. 18 days later, Figure A is the post-differentiation Photograph of the posterior cell. Figure B is a schematic diagram of Benzidine staining bright field. Panel C is a schematic diagram of Wright-Giemsa staining bright field. Ruler: 20um. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, and gene repair: represent cells that have undergone gene repair.
图11电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,进行红细胞分化,18天后,进行Benzidine染色,统计分析Benzidine阳性的比例。其中,健康供者:代表未经过基因编辑的健康供者,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞,n=3个实验重复。Figure 11 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. 18 days later, perform Benzidine staining and statistics Analyze the proportion of Benzidine positive. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, gene repair: represent cells that have undergone gene repair, n=3 experimental replicates.
图12电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,进行红细胞分化,18天后检测,通过荧光定量PCR检测ALAS-2、GATA-1和GAPDH基因mRNA表达。其中,健康供者:代表未经过基因编辑的健康供者,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞,n=3个实验重复。ALAS-2基因和GATA-1与GAPDH和健康供者进行归一化处理。Figure 12 Electrotransform Cas9 mRNA, sgRNA-1 near the point mutation of ALAS-2 intron-1 and donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation, detected after 18 days, and quantified by fluorescence PCR was used to detect the mRNA expression of ALAS-2, GATA-1 and GAPDH genes. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, gene repair: represent cells that have undergone gene repair, n=3 experimental replicates. ALAS-2 gene and GATA-1 were normalized with GAPDH and healthy donors.
图13电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,进行红细胞分化,18天后检测,通过Western Blot实验检测ALAS-2、GATA-1和GAPDH基因蛋白水平表达。其中,健康供者:代表未经过基因编辑的健康供者,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。Figure 13 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 and the donor template ssODN into CD34+HSPC derived from the bone marrow of XLSA patients for red blood cell differentiation. After 18 days, the test was performed by Western Blot experiment. Detect the protein level expression of ALAS-2, GATA-1 and GAPDH genes. Among them, healthy donors: represent healthy donors that have not undergone gene editing, blank control: represent cells that have not undergone gene editing, and gene repair: represent cells that have undergone gene repair.
图14电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+HSPC,电转2天后移植经过基因修复和未经过基因编辑的细胞进入经过辐照仪照射的6周龄的NPG免疫缺陷小鼠模型,10周、12周、16周后,在小鼠外周血检测人CD45阳性细胞的比例,同时移植16周后在小鼠骨髓和脾脏中检测人CD45阳性细胞的比例,其中CD45阳性细胞的比例的计算方式为人CD45阳性细胞%/(人CD45阳性细胞%+小鼠CD45阳性细胞%),人CD45阳性细胞%和小鼠CD45阳性细胞%分别是通过流式分析实验测得的结果。空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。n=6只小鼠。Figure 14 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. The cells from the irradiator enter the 6-week-old NPG immunodeficiency mouse model. After 10 weeks, 12 weeks, and 16 weeks, the proportion of human CD45-positive cells in the peripheral blood of the mouse is detected. At the same time, the proportion of human CD45 positive cells is detected in the mouse after 16 weeks of transplantation. The proportion of human CD45 positive cells detected in mouse bone marrow and spleen. The proportion of CD45 positive cells is calculated as human CD45 positive cells%/(human CD45 positive cells%+mouse CD45 positive cells%), human CD45 positive cells% and small The percentages of mouse CD45 positive cells were measured by flow cytometry experiments. Blank control: represents cells that have not undergone gene editing, and gene repair: represents cells that have undergone gene repair. n=6 mice.
图15电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+的HSPC,电转2天后移植经过基因修复和未经过基因编辑的细胞进入经过辐照仪照射的6周龄的NPG免疫缺陷小鼠模型,16周后分别在小鼠骨髓和脾脏中检测CD3、CD33、CD56、CD19等人细胞膜蛋白占人CD45蛋白的比例。空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。n=6只小鼠。Figure 15 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. The cells entered into a 6-week-old NPG immunodeficiency mouse model irradiated with an irradiator. After 16 weeks, the ratio of human cell membrane proteins such as CD3, CD33, CD56, and CD19 to human CD45 protein was detected in the mouse bone marrow and spleen. Blank control: represents cells that have not undergone gene editing, and gene repair: represents cells that have undergone gene repair. n=6 mice.
图16电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+的HSPC,电转2天后移植经过基因修复和未经过基因编辑的细胞进入经过辐照仪照射的6周龄的NPG免疫缺陷小鼠模型,16周后,流式分析空白对照组和基因修复组中各1只小鼠的骨髓、脾脏和外周血中人CD45阳性细胞的比例。Figure 16 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. The cells entered the 6-week-old NPG immunodeficiency mouse model irradiated by the irradiator. After 16 weeks, flow cytometry analysis of human CD45 in bone marrow, spleen and peripheral blood of 1 mouse in the blank control group and the gene repair group The proportion of positive cells.
图17电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+的HSPC,电转2天后移植经过基因修复和未经过基因编辑的细胞进入经过辐照仪照射的6周龄的NPG免疫缺陷小鼠模型,16周后,流式分析空白对照组和基因修复组中各1只小鼠的骨髓和脾脏中CD3、CD33、CD56、CD19等人细胞膜蛋白占人CD45蛋白的比例。其中,空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因修复的细胞。Figure 17 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. The cells enter the 6-week-old NPG immunodeficiency mouse model that has been irradiated by the irradiator. After 16 weeks, flow cytometry analysis of CD3, CD33, and CD56 in the bone marrow and spleen of 1 mouse in the blank control group and the gene repair group , CD19 and other human cell membrane proteins accounted for the proportion of human CD45 protein. Among them, blank control: represents cells that have not undergone gene editing, and gene repair: represents cells that have undergone gene repair.
图18电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1以及供体模板ssODN进入XLSA病人骨髓来源的CD34+的HSPC,电转2天后移植经过基因修复和未经过基因编辑的细胞进入经过辐照仪照射的6周龄的NPG免疫缺陷小鼠模型,提取移植前细胞和移植后16周骨髓的基因 组,扩增目的片段和NGS,通过生物信息学方法分析NHEJ和HDR的比例。其中,NHEJ:non-homologous end joining,代表Indels比例,HDR:Homology-directed repair,代表基因修复的比例。n=6只小鼠。Figure 18 Electrotransformation of Cas9 mRNA, sgRNA-1 and donor template ssODN near the point mutation of ALAS-2 intron-1 into CD34+ HSPC derived from the bone marrow of XLSA patients. Two days after electrotransformation, the transplantation has undergone gene repair and has not undergone gene editing. Cells from the irradiator enter the 6-week-old NPG immunodeficiency mouse model, extract the genome of the cells before transplantation and the bone marrow 16 weeks after transplantation, amplify the target fragment and NGS, and analyze the NHEJ and HDR by bioinformatics methods. proportion. Among them, NHEJ: non-homologous end joining, represents the ratio of Indels, and HDR: Homology-directed repair, represents the ratio of gene repair. n=6 mice.
图19分离经1次移植16周后NPG免疫缺陷小鼠的骨髓,再移植进入新的经过辐照的NPG免疫缺陷小鼠进行2次移植试验。移植后12周分离骨髓细胞,检测人CD45阳性细胞的比例,其中CD45阳性细胞的比例的计算方式为人CD45阳性细胞%/(人CD45阳性细胞%+小鼠CD45阳性细胞%),人CD45阳性细胞%和小鼠CD45阳性细胞%,分别是通过流式分析实验测得的结果。空白对照:代表移植的是未经过基因编辑的细胞,基因修复:代表移植的是经过基因修复的细胞。Figure 19 Isolation of bone marrow from NPG immunodeficient mice 16 weeks after one transplantation, and transplantation into new irradiated NPG immunodeficient mice for two transplantation experiments. Isolate bone marrow cells at 12 weeks after transplantation and detect the proportion of human CD45-positive cells. The proportion of CD45-positive cells is calculated as human CD45-positive cells%/(human CD45-positive cells%+mouse CD45-positive cells%), human CD45-positive cells % And mouse CD45 positive cells% are the results measured by flow cytometry experiments. Blank control: represents cells that have not undergone gene editing, and gene repair: represents cells that have undergone gene repair.
图20分离经1次移植NPG免疫缺陷小鼠16周后的骨髓,再移植进入新的经过辐照的NPG免疫缺陷小鼠进行2次移植试验。提取移植后12周分离骨髓细胞检测基因修复效率,分析NHEJ和HDR的比例。其中,NHEJ:为非同源末端连接,代表Indels比例,HDR:为同源重组修复,代表基因修复的比例。Figure 20 Isolation of bone marrow 16 weeks after one transplantation of NPG immunodeficiency mice, and then transplantation into new irradiated NPG immunodeficiency mice for two transplantation experiments. The bone marrow cells were isolated 12 weeks after transplantation to test the gene repair efficiency and analyze the ratio of NHEJ and HDR. Among them, NHEJ: non-homologous end connection, representing the ratio of Indels, HDR: homologous recombination repair, representing the ratio of gene repair.
图21电转Cas9 mRNA和针对ALAS-2内含子-1的点突变附近的sgRNA-1进入XLSA病人来源的hiPSCs,电转2天后提取基因组,扩增通过序列相似性预测分析以及无偏全基因组分析方法Digenome-Seq方法预测出的32个潜在脱靶位点的目的片段并进行NGS测序分析,通过生物信息学方法分析每个脱靶位点的突变频率。其中空白对照:代表未经过基因编辑的细胞,基因修复:代表经过基因编辑的细胞,POT:潜在脱靶位点(potential off-target),On-target(靶标位点)表示基因编辑效率。Figure 21 Electrotransform Cas9 mRNA and sgRNA-1 near the point mutation of ALAS-2 intron-1 into hiPSCs derived from XLSA patients, extract the genome 2 days after electrotransformation, and amplify through sequence similarity prediction analysis and unbiased whole-genome analysis Methods The target fragments of 32 potential off-target sites predicted by the Digenome-Seq method were analyzed by NGS sequencing, and the mutation frequency of each off-target site was analyzed by bioinformatics methods. The blank control: represents cells that have not undergone gene editing, gene repair: represents cells that have undergone gene editing, POT: potential off-target, and On-target represents gene editing efficiency.
发明详述Detailed description of the invention
本申请提供了一种通过基因编辑校正5-氨基乙酰丙酸合酶2(ALAS-2)基因突变的方法,该方法能够高效修复XLSA患者来源的hiPSC和CD34+HSPC中的ALAS-2基因突变,显著提高ALAS-2基因和蛋白表达,从而显著提高分化的红细胞中亚铁血红素的合成,促进红细胞成熟和携带氧的能力,改善贫血患者的症状,从而克服传统治疗方法的缺陷,满足临床治疗的要求。This application provides a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations through gene editing, which can efficiently repair the ALAS-2 gene mutations in hiPSC and CD34+HSPC derived from XLSA patients , Significantly increase the expression of ALAS-2 gene and protein, thereby significantly increasing the synthesis of heme in differentiated red blood cells, promoting the maturation of red blood cells and the ability to carry oxygen, improving the symptoms of anemia patients, so as to overcome the shortcomings of traditional treatment methods and meet the clinical needs Treatment requirements.
I.定义I. Definition
本申请所述的“基因编辑”指对基因组进行定点修饰以实现在基因水平 上定点删除、插入和/或替换具体核苷酸和核苷酸片段的技术。目前人们熟知的基因编辑技术包括人工核酸酶介导的锌指核酸酶(ZFN)技术、转录激活子样效应物核酸酶(TALEN)技术和RNA引导的CRISPR/Cas核酸酶(CRISPR/Cas RGNs)技术。它们都能特异性地识别靶位点,对其单链或双链进行精准切割后,由细胞内源性的修复机制来完成对靶标基因的敲除和替换。其中,CRISPR/Cas技术是一项新兴的基因编辑技术,它利用与目标序列互补的sg RNA引导Cas酶定点切割DNA。The "gene editing" mentioned in this application refers to the technique of site-specific modification of the genome to achieve site-specific deletion, insertion, and/or replacement of specific nucleotides and nucleotide fragments at the gene level. At present, well-known gene editing technologies include artificial nuclease-mediated zinc finger nuclease (ZFN) technology, transcription activator-like effector nuclease (TALEN) technology, and RNA-guided CRISPR/Cas nuclease (CRISPR/Cas RGNs) technology. They can specifically recognize the target site, and after precise cutting of its single or double strand, the cell's endogenous repair mechanism completes the knockout and replacement of the target gene. Among them, CRISPR/Cas technology is an emerging gene editing technology, which uses sgRNA complementary to the target sequence to guide Cas enzyme to cut DNA at a specific point.
“突变序列”为与正常的天然序列相比,核苷酸序列发生改变的基因序列。替换突变序列,从而实现对突变序列的校正的核苷酸序列称为“校正序列”。“供体DNA”是包含“校正序列”的DNA。包含校正序列的供体DNA分子通过电穿孔或转导等方式被导入细胞后,与突变序列可发生同源重组,从而校正序列可以替换突变序列,实现基因编辑。A "mutated sequence" is a gene sequence whose nucleotide sequence has been changed compared to a normal natural sequence. The nucleotide sequence that replaces the mutated sequence to achieve correction of the mutated sequence is called a "correction sequence". "Donor DNA" is DNA containing a "correction sequence". After the donor DNA molecule containing the correction sequence is introduced into the cell by means of electroporation or transduction, homologous recombination can occur with the mutant sequence, so that the correction sequence can replace the mutant sequence to realize gene editing.
“干细胞”是指具有旺盛的增殖潜力、多向分化的能力和自我更新能力的细胞群体。“造血干细胞”是指具有旺盛的增殖潜力、向血细胞多向分化的能力和自我更新能力的细胞群体。造血干细胞不仅能分化、补充各种血细胞,还能通过自我更新保持干细胞的特性和数量。造血干细胞的分化程度和增殖能力不一,有异质性。多能造血干细胞最原始,先分化为定向多能造血干细胞,例如能生成粒系、红系、单核系和巨核-血小板系的髓系造血干细胞及能发生B淋巴细胞和T淋巴细胞的淋巴干细胞。这两类干细胞既保持着造血干细胞基本特点,又略有分化,分别负责“骨髓成分”和淋巴细胞的发生,故称定向多能造血干细胞。它们进一步分化成为造血祖细胞,此细胞虽然也是原始的血细胞,但是它已丧失造血干细胞的许多基本特点,如已失去多向性分化能力,只能朝向一系或密切相关的二系细胞分化;失去了反复自我更新能力,而要依靠造血干细胞的增殖分化来补充数量;增殖潜力有限,只能分裂数次。根据造血祖细胞所能分化生成的血细胞系多少,又分为单能造血祖细胞(只分化为一个血细胞系)和寡能造血祖细胞(可分化为2~3个血细胞系)。本申请所述“造血干细胞”指可通过分化或通过定向诱导分化形成粒系、红系、单核系、巨核-血小板系和/或淋巴系细胞的细胞群体,是多能造血干细胞、定向多能造血干细胞、造血祖细胞的总称。造血干细胞可来源于骨髓(骨髓造血干细胞)、外周血(外周造血干细胞)、脐带血(脐带血造血干细胞),也可来源于胎盘干细胞或hiPSC等。本申请所述的“CD34阳性造血干细胞/祖 细胞”,简写CD34阳性HSPC(hematopoietic stem/progenitor cell,HSPC)或CD34+HSPC,指表面表达CD34标志物的、有造血功能的造血干细胞和祖细胞的细胞群体。可以使用例如流式细胞术和荧光标记的抗CD34抗体来检测和计数CD34阳性造血干细胞/祖细胞(HSPC)。"Stem cell" refers to a cell population with vigorous proliferation potential, multi-differentiation ability and self-renewal ability. "Hematopoietic stem cells" refer to cell populations with vigorous proliferation potential, multidirectional differentiation into blood cells, and self-renewal capabilities. Hematopoietic stem cells can not only differentiate and supplement various blood cells, but also maintain the characteristics and quantity of stem cells through self-renewal. The degree of differentiation and proliferation ability of hematopoietic stem cells are different and heterogeneous. Pluripotent hematopoietic stem cells are the most primitive, and first differentiate into directed pluripotent hematopoietic stem cells, such as myeloid hematopoietic stem cells that can produce granulocytes, erythroid, mononuclear and megakaryocyte-platelet lines, and lymphoids that can produce B lymphocytes and T lymphocytes stem cell. These two types of stem cells not only maintain the basic characteristics of hematopoietic stem cells, but are also slightly differentiated. They are responsible for the occurrence of "bone marrow components" and lymphocytes, so they are called directed pluripotent hematopoietic stem cells. They further differentiate into hematopoietic progenitor cells. Although this cell is also a primitive blood cell, it has lost many of the basic characteristics of hematopoietic stem cells. Lost the ability of repeated self-renewal, and rely on the proliferation and differentiation of hematopoietic stem cells to supplement the number; the proliferation potential is limited and can only divide several times. According to the number of blood cell lines that hematopoietic progenitor cells can differentiate, they are divided into unipotent hematopoietic progenitor cells (differentiated into only one blood cell line) and oligopotent hematopoietic progenitor cells (differentiated into 2 to 3 blood cell lines). The “hematopoietic stem cells” mentioned in the present application refer to cell populations that can form granulocytes, erythroid, monocytes, megakaryocytes-platelet cells and/or lymphoid cells through differentiation or directed differentiation. They are pluripotent hematopoietic stem cells and are multipotent. Generic term for hematopoietic stem cells and hematopoietic progenitor cells. Hematopoietic stem cells can be derived from bone marrow (bone marrow hematopoietic stem cells), peripheral blood (peripheral hematopoietic stem cells), umbilical cord blood (umbilical cord blood hematopoietic stem cells), and can also be derived from placental stem cells or hiPSC. The "CD34-positive hematopoietic stem cell/progenitor cell" mentioned in this application, abbreviated as CD34-positive HSPC (hematopoietic stem/progenitor cell, HSPC) or CD34+HSPC, refers to hematopoietic stem cells and progenitor cells that express CD34 markers on the surface and have hematopoietic function Cell population. For example, flow cytometry and fluorescently labeled anti-CD34 antibodies can be used to detect and count CD34-positive hematopoietic stem/progenitor cells (HSPC).
如本申请使用的,“CRISPR/Cas”是一种基因编辑技术,包括但不限于各种自然存在或人工设计的CRISPR/Cas系统,如CRISPR/Cas9系统。自然存在的CRISPR/Cas系统(Naturally occurring CRISPR/Cas system)是细菌和古细菌在长期演化过程中形成的一种适应性免疫防御,可用来对抗入侵的病毒及外源DNA。例如简单的CRISPR/Cas9系统包含Cas9酶、crRNA(CRISPR-derived RNA)和tracrRNA(trans-activating crRNA)三个组分。其中crRNA(CRISPR-derived RNA)包含指导序列以及与tracrRNA部分互补的序列。tracrRNA即反式作用crRNA(trans-activating RNA),包含较长的恒定碱基序列,提供由CRISPR核酸酶(例如Cas9酶)结合的“茎环”结构。crRNA通过碱基配对与tracrRNA(trans-activating RNA)结合形成tracrRNA/crRNA复合物,通过此复合物中crRNA与目的序列形成互补,并通过tracrRNA中的“茎环”结构,可将Cas9核酸酶引导至目的序列的靶位点剪切双链DNA。而通过人工设计,可将tracrRNA和crRNA结合,改造形成具有引导作用的sgRNA(single guide RNA),使所述sgRNA足以引导Cas9对DNA的定点切割。作为一种RNA引导的dsDNA结合蛋白,Cas9核酸酶能够共定位RNA、DNA和蛋白,拥有巨大的改造潜力。CRISPR/Cas系统可使用一类,二类或三类Cas蛋白。本发明的一些实施方式中,所述方法使用Cas9。其他适用的CRISPR/Cas系统包括但不限于WO2013176772,WO2014065596,WO2014018423,US8,697,359、PCT/CN2018/112068、PCT/CN2018/112027中所描述的系统和方法。As used in this application, "CRISPR/Cas" is a gene editing technology, including but not limited to various naturally occurring or artificially designed CRISPR/Cas systems, such as the CRISPR/Cas9 system. The naturally occurring CRISPR/Cas system (Naturally occurring CRISPR/Cas system) is an adaptive immune defense formed during the long-term evolution of bacteria and archaea, which can be used to fight invading viruses and foreign DNA. For example, a simple CRISPR/Cas9 system includes three components: Cas9 enzyme, crRNA (CRISPR-derived RNA) and tracrRNA (trans-activating crRNA). Among them, crRNA (CRISPR-derived RNA) contains a guide sequence and a sequence partially complementary to tracrRNA. tracrRNA is trans-activating RNA (trans-activating RNA), which contains a long constant base sequence and provides a "stem-loop" structure bound by CRISPR nuclease (such as Cas9 enzyme). crRNA combines with tracrRNA (trans-activating RNA) through base pairing to form a tracrRNA/crRNA complex, through which crRNA and the target sequence are complementary, and through the "stem-loop" structure in tracrRNA, Cas9 nuclease can be guided Cut the double-stranded DNA to the target site of the target sequence. Through artificial design, tracrRNA and crRNA can be combined to transform into a guiding sgRNA (single guide RNA), so that the sgRNA is sufficient to guide Cas9's targeted cleavage of DNA. As an RNA-guided dsDNA binding protein, Cas9 nuclease can co-localize RNA, DNA and protein, and has great potential for transformation. The CRISPR/Cas system can use type 1, type 2 or type 3 Cas proteins. In some embodiments of the invention, the method uses Cas9. Other applicable CRISPR/Cas systems include but are not limited to the systems and methods described in WO2013176772, WO2014065596, WO2014018423, US8,697,359, PCT/CN2018/112068, PCT/CN2018/112027.
细胞“分化”是指同一来源的细胞逐渐产生形态结构、功能特征各不相同的细胞类群的过程。Cell "differentiation" refers to the process in which cells from the same source gradually produce cell groups with different morphological structures and functional characteristics.
从造血干细胞到红细胞的“分化”包括造血干细胞阶段、红系祖细胞阶段、红系前体细胞(原红细胞至晚红细胞)的增殖与分化阶段、网织红细胞的增殖及成熟过程,以及网织红细胞向外周血释放成熟为红细胞的阶段。造血干细胞阶段:目前已知,造血干细胞主要存在于骨髓、脾、肝等造血组织内。也有少量循环于外周血中。红系祖细胞阶段:在红系祖细胞(progenitor  cell)阶段,细胞是处于造血干细胞与红系前体细胞之间的细胞群。造血干细胞在骨髓造血微环境的影响下分化为红系祖细胞。造血微环境包括微血管系统、神经系统和造血间质等部分。通过体液因子、细胞因子对造血干细胞的分化起特殊的作用和影响。红系前体细胞阶段:包括原始红细胞、早幼红细胞、中幼红细胞、晚幼红细胞及网织红细胞阶段而达到成熟红细胞。The "differentiation" from hematopoietic stem cells to erythrocytes includes hematopoietic stem cell stage, erythroid progenitor cell stage, erythroid precursor cell (primary red blood cell to late red blood cell) proliferation and differentiation stage, reticulocyte proliferation and maturation process, and reticulum Red blood cells are released from peripheral blood to mature into red blood cells. Hematopoietic stem cell stage: It is currently known that hematopoietic stem cells mainly exist in bone marrow, spleen, liver and other hematopoietic tissues. There is also a small amount of circulation in the peripheral blood. Erythroid progenitor cell stage: In the progenitor cell stage, cells are a cell group between hematopoietic stem cells and erythroid precursor cells. Hematopoietic stem cells differentiate into erythroid progenitor cells under the influence of bone marrow hematopoietic microenvironment. The hematopoietic microenvironment includes the microvascular system, nervous system and hematopoietic interstitium. Humoral factors and cytokines have a special effect and influence on the differentiation of hematopoietic stem cells. Erythroid precursor cell stage: including primitive red blood cells, early young red blood cells, middle young red blood cells, late young red blood cells and reticulocytes to reach mature red blood cells.
“非同源性末端接合”也简称为NHEJ(Non-homologous end joining),是真核生物细胞在不依赖DNA同源性的情况下,而为了避免DNA或染色体断裂的滞留和因此造成的DNA降解的影响,强行将两个DNA断端彼此连接在一起的一种DNA双链断裂修复机制,在基因编辑过程中,NHEJ可能产生插入和缺失(Indels,Insertions and deletions),导致基因突变。"Non-homologous end joining" is also abbreviated as NHEJ (Non-homologous end joining), which refers to eukaryotic cells that do not rely on DNA homology to avoid the retention of DNA or chromosome breaks and the resulting DNA The effect of degradation is a DNA double-strand break repair mechanism that forcibly connects two DNA ends to each other. In the process of gene editing, NHEJ may produce insertions and deletions (Indels, Insertions and deletions), leading to gene mutations.
“同源修复”也简称为HDR(Homology-directed repair),也可称为同源介导的双链DNA修复。是细胞内一种修复DNA双链损伤的机制。只有当细胞核内存在与损伤DNA同源的DNA片段时,HDR才能发生。"Homology-directed repair" is also referred to as HDR (Homology-directed repair), and can also be referred to as homology-mediated double-stranded DNA repair. It is a mechanism for repairing DNA double-strand damage in cells. HDR can only occur when there are DNA fragments homologous to the damaged DNA in the nucleus.
“贫血”是指人体外周血红细胞容量减少,低于正常范围下限的一种临床症状。“铁粒幼细胞性贫血”是铁利用障碍性疾病,特征为骨髓中出现大量环状铁粒幼红细胞,红细胞无效生成,组织铁储量过多和外周血呈小细胞低色素性贫血。铁粒幼细胞性贫血主要分为获得性和遗传性铁粒幼细胞性贫血,其中遗传性铁粒幼细胞性贫血多为青少年、男性及有家族史。铁利用不良,血红素合成障碍和红细胞无效生成是本病发病的主要环节。铁利用不良和血红素合成障碍的结果是形成低色素性贫血,铁在红细胞及各组织内大量堆积,使红细胞形态及功能受损,致红细胞过早破坏。铁大量沉积于各组织内,形成血色病,影响各组织器官功能。"Anemia" refers to a clinical symptom in which the volume of human peripheral blood red blood cells decreases below the lower limit of the normal range. "Sideroblastic anemia" is a disorder of iron utilization. It is characterized by the appearance of a large number of ring sideroblasts in the bone marrow, ineffective production of red blood cells, excessive tissue iron reserves, and small cell hypochromic anemia in the peripheral blood. Sideroblastic anemia is mainly divided into acquired and hereditary sideroblastic anemia. Among them, hereditary sideroblastic anemia is mostly adolescents, males and have family history. Poor iron utilization, impaired heme synthesis and ineffective production of red blood cells are the main links in the pathogenesis of this disease. The result of poor iron utilization and heme synthesis disorder is the formation of hypochromic anemia. A large amount of iron accumulates in red blood cells and various tissues, which damages the morphology and function of red blood cells and causes premature destruction of red blood cells. A large amount of iron deposits in various tissues, forming hemochromatosis, affecting the functions of various tissues and organs.
II.基因编辑校正ALAS-2基因突变II. Gene editing to correct ALAS-2 gene mutations
本申请涉及通过基因修复ALAS-2基因中特定突变,例如位于ALAS-2基因的外显子5-11或内含子-1中的突变,例如Int-1-GATA点突变,提高来源于铁粒幼红细胞贫血患者,例如遗传性铁粒幼红细胞贫血患者,具体地,XLSA患者的CD34 +HSPC或人诱导多能干细胞(hiPSC)中ALAS-2基因和蛋白表达,从而治疗包括遗传性铁粒幼红细胞贫血,例如XLSA的铁粒幼红细胞贫血。 This application relates to genetic repair of specific mutations in the ALAS-2 gene, such as mutations in exons 5-11 or intron-1 of the ALAS-2 gene, such as Int-1-GATA point mutations, to improve the source of iron Myeloblastic anemia patients, such as hereditary sideroblast anemia patients, specifically, CD34 + HSPC or human induced pluripotent stem cells (hiPSC) in XLSA patients with ALAS-2 gene and protein expression, so that the treatment includes hereditary sideroblasts Immature red blood cell anemia, such as sideroblast anemia of XLSA.
在一些实施方案中,本发明涉及一种通过基因编辑来校正造血干细胞染色体中5-氨基乙酰丙酸合酶2(ALAS-2)基因突变的方法,其中所述基因编辑 包括:(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。本发明还涉及一种通过基因编辑来校正ALAS-2基因突变,从而增加功能性ALAS-2表达的方法。本发明还涉及一种通过基因编辑来校正ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生的方法。本发明还涉及一种通过基因编辑来校正ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生,由此促进所述造血干细胞成熟的方法。本发明还涉及一种通过基因编辑来校正ALAS-2基因突变,增加功能性ALAS-2表达,从而增加源自所述造血干细胞的细胞中血红素产生,促进所述造血干细胞成熟,由此来治疗个体的包括遗传性铁粒幼红细胞贫血,例如XLSA的铁粒幼红细胞贫血的方法。In some embodiments, the present invention relates to a method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations in the chromosomes of hematopoietic stem cells through gene editing, wherein the gene editing includes: (a) A donor DNA containing a single-stranded correction sequence corresponding to the mutant sequence is introduced into the hematopoietic stem cell; (b) a sequence-specific nuclease that cleaves the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the donor The correction sequence on the somatic DNA replaces the mutation sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation. The invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2. The present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing and increasing the expression of functional ALAS-2, thereby increasing the production of heme in cells derived from the hematopoietic stem cells. The present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of heme in the cells derived from the hematopoietic stem cells, thereby promoting the maturation of the hematopoietic stem cells . The present invention also relates to a method for correcting ALAS-2 gene mutations through gene editing, increasing the expression of functional ALAS-2, thereby increasing the production of heme in the cells derived from the hematopoietic stem cells, and promoting the maturation of the hematopoietic stem cells, thereby Methods of treating individuals include hereditary sideroblast anemia, such as XLSA sideroblast anemia.
在上述方法的一些具体实施方案中,所述造血干细胞是从铁粒幼红细胞贫血,包括遗传性铁粒幼红细胞贫血,例如XLSA的患者获得的。在一些实施方案中,所述患者为雄性个体或雌性个体,所获得的的干细胞为CD34 +造血干细胞和祖细胞(“HSPC”),或人诱导多能干细胞(hiPSC)。 In some specific embodiments of the above methods, the hematopoietic stem cells are obtained from patients with sideroblast anemia, including hereditary sideroblast anemia, such as XLSA. In some embodiments, the patient is a male individual or a female individual, and the obtained stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
在上述方法的一些具体实施方案中,CD34阳性造血干细胞/祖细胞从包含造血来源细胞的生物体(个体)分离获得。“分离”是指从其原始环境取出。例如,如果细胞与在其天然状态通常伴随它的一些或所有组分分开时,则它是分离的。造血干细胞/祖细胞可以从成人的未分级分离或分级分离的骨髓获得或分离,包括股骨,髋骨,肋骨,胸骨和其它骨骼。In some specific embodiments of the above method, CD34-positive hematopoietic stem/progenitor cells are isolated from an organism (individual) containing cells of hematopoietic origin. "Separate" means to remove from its original environment. For example, a cell is isolated if it is separated from some or all of the components that normally accompany it in its natural state. Hematopoietic stem cells/progenitor cells can be obtained or isolated from unfractionated or fractionated bone marrow of adults, including femurs, hip bones, ribs, sternum and other bones.
造血干细胞和祖细胞可以利用针和注射器从髋骨取出直接获得或分离,或者从血液中获得,通常是在造血干细胞动员剂诸如G-CSF(粒细胞集落刺激因子)预处理后从血液中获得。造血干细胞和祖细胞的其他来源包括脐带血、胎盘血和经过动员的个体的外周血。Hematopoietic stem cells and progenitor cells can be directly obtained or separated from the hip bone using a needle and syringe, or obtained from the blood, usually obtained from the blood after pretreatment with a hematopoietic stem cell mobilizer such as G-CSF (granulocyte colony stimulating factor) . Other sources of hematopoietic stem and progenitor cells include cord blood, placental blood, and peripheral blood of mobilized individuals.
从个体(例如骨髓或外周血)分离获得细胞群体后可对其进行进一步纯化获得CD34阳性造血干细胞/祖细胞。例如可以通过免疫去除分离的细胞群体中的成熟谱系定向细胞,例如通过用结合一组“谱系”抗原(例如CD2,CD3,CD11b,CD14,CD15,CD16,CD19,CD56,CD123和CD235a)的抗体标记固体基质,之后用结合CD34阳性抗原的抗体分离原始造血干细胞和祖 细胞。用于从多种细胞来源纯化造血干细胞和祖细胞的试剂盒是商业可获得的,并且在具体实施方案中,这些试剂盒可以与本发明的方法一起使用。After the cell population is isolated from an individual (such as bone marrow or peripheral blood), it can be further purified to obtain CD34-positive hematopoietic stem cells/progenitor cells. For example, mature lineage-directed cells in an isolated cell population can be removed by immunization, for example, by using antibodies that bind to a set of "lineage" antigens (eg CD2, CD3, CD11b, CD14, CD15, CD16, CD19, CD56, CD123, and CD235a) The solid matrix is labeled, and then the original hematopoietic stem cells and progenitor cells are separated with antibodies that bind to CD34 positive antigen. Kits for purifying hematopoietic stem and progenitor cells from a variety of cell sources are commercially available, and in specific embodiments, these kits can be used with the methods of the present invention.
“CD34阳性造血干细胞/祖细胞”可代表富含CD34阳性细胞的细胞群中至少50%、60%、70%、80%、90%、95%、96%、97%、98%、99%或100%的CD34阳性造血干细胞/祖细胞(HSPC)。"CD34 positive hematopoietic stem cells/progenitor cells" can represent at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% of the cell population rich in CD34 positive cells Or 100% CD34-positive hematopoietic stem/progenitor cells (HSPC).
在上述方法的一些实施方案中,所述基因编辑包括将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中。在一些实施方案中,所述的单链校正序列替换ALAS-2基因中的突变序列。In some embodiments of the above method, the gene editing includes introducing a donor DNA containing a single-stranded correction sequence corresponding to the mutant sequence into the hematopoietic stem cell. In some embodiments, the single-stranded correction sequence replaces the mutant sequence in the ALAS-2 gene.
在一些实施方案中,所述的供体DNA与所述的单链校正序列核苷酸组成和长度相同。在一些实施方案中,所述供体DNA较所述单链校正序列长,例如所述的单链校正序列的一端或二端添加有一个或多个核苷酸。在一些实施方案中,所述添加的核苷酸组成的序列是核酸酶特异性识别位点。在一些实施方案中,所述供体DNA中校正序列的两端还可以进一步包含针对所述核酸酶特异性识别位点的保护碱基。在一些实施方案中所述供体DNA还进一步包含一个或多个LNA核苷。在一些实施方案中,所述供体DNA是单链的。在一些实施方案中,所述供体DNA是环状的。在一些实施方案中,所述的供体DNA以质粒或病毒载体的形式提供。在一些实施方案中,所述供体DNA是ssODN(单链供体寡核苷酸,single-stranded donor oligonucleotides)。在一些实施方案中,所述供体DNA为SEQ ID NO:4所示。In some embodiments, the donor DNA has the same nucleotide composition and length as the single-stranded correction sequence. In some embodiments, the donor DNA is longer than the single-stranded correction sequence, for example, one or more nucleotides are added to one or both ends of the single-stranded correction sequence. In some embodiments, the sequence composed of the added nucleotides is a nuclease specific recognition site. In some embodiments, both ends of the calibration sequence in the donor DNA may further include protective bases for the specific recognition site of the nuclease. In some embodiments, the donor DNA further comprises one or more LNA nucleosides. In some embodiments, the donor DNA is single-stranded. In some embodiments, the donor DNA is circular. In some embodiments, the donor DNA is provided in the form of a plasmid or viral vector. In some embodiments, the donor DNA is ssODN (single-stranded donor oligonucleotides). In some embodiments, the donor DNA is shown in SEQ ID NO: 4.
在一些实施方案中,所述供体DNA是经化学修饰的,例如核苷酸核糖上的2’-O-甲基化修饰、核苷酸间的3’硫代磷酸酯化修饰以及5’端磷酸化修饰。在一些具体实施方案中,所述化学修饰为所述供体DNA5’端前3个核苷酸和3’端后3个核苷酸核糖的2’-O-甲基化修饰和/或核苷酸间的3’硫代磷酸酯化修饰。在一些具体实施方案中,所述化学修饰为所述供体DNA的5’端前一个、二个和/或三个碱基和/或3’端的最后一个核苷酸核糖的2’-O-甲基化修饰。在一些实施方案中,所述供体DNA在5’端前3个和3’端后3个核苷酸中包含核苷酸核糖的2′-O-甲基化修饰且在5’端的前3个和3’端后3个核苷酸间连接中包含硫代磷酸酯化修饰。在一些实施方案中,所述供体DNA在5’端的前5个核苷酸及3’端的后5个核苷酸的核糖中包含2’-O-甲基化修饰,并且在5’端的前5个及3’端的后5个核苷酸间连接中包含硫代磷酸酯化修饰。在一些特定的实施方案中,所述供体DNA包含5’端磷酸化修饰,并 且其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰。在一些特定的实施方案中,所述供体DNA包含5’端磷酸化修饰,其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰,并且5’端的前三个核苷酸和3’端的最后3个核苷酸还包含核糖的2’-O-甲基化修饰。In some embodiments, the donor DNA is chemically modified, such as 2'-O-methylation modification on nucleotide ribose, 3'phosphorothioate modification between nucleotides, and 5' End phosphorylation modification. In some embodiments, the chemical modification is a 2'-O-methylation modification and/or a ribose 3 nucleotides before the 5'end and 3 nucleotides after the 3'end of the donor DNA. 3'phosphorothioate modification between glycidyl acids. In some specific embodiments, the chemical modification is one, two and/or three bases before the 5'end of the donor DNA and/or the 2'-O of the last nucleotide ribose at the 3'end. -Methylation modification. In some embodiments, the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose in the 3 nucleotides before the 5'end and the 3 nucleotides after the 3'end, and the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose before the 5'end. The three internucleotide linkages after the 3 and 3'ends contain phosphorothioate modification. In some embodiments, the donor DNA contains a 2'-O-methylation modification in the ribose of the first 5 nucleotides at the 5'end and the last 5 nucleotides at the 3'end, and the ribose at the 5'end The first 5 and the last 5 internucleotide linkages at the 3'end contain phosphorothioate modification. In some specific embodiments, the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end. In some specific embodiments, the donor DNA contains a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, and The first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end also contain the 2'-O-methylation modification of ribose.
在本申请所述的一些方法中,基因编辑包括将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中。在一些实施方案中,序列特异性核酸酶包括RNA引导核酸酶、锌指核酸酶(ZFN)和转录激活因子样效应物核酸酶(TALEN)。所述序列特异性核酸酶可以是例如RNA引导核酸酶,即Cas核酸酶,具体地可以是Cas9。在上述方法的一个具体实施方案中,所述核酸酶剪切位点距离所述突变位点不超过约11个核苷酸的位点。在一些实施方案中,将编码Cas9的mRNA,例如含有ARCA帽的mRNA导入(例如通过电穿孔或其他基因转导的方式)干细胞。在一些实施方案中,编码所述Cas核酸酶(例如Cas9)的核苷酸通过病毒载体(例如慢病毒载体)导入所述造血干细胞。在一些实施方案中,所述sgRNA与Cas9编码核酸存在于同一载体中。在一些实施方案中,所述sgRNA与Cas9编码核酸存在于不同载体中。In some of the methods described in this application, gene editing includes introducing a sequence-specific nuclease that cleaves the ALAS-2 gene into the hematopoietic stem cells. In some embodiments, sequence-specific nucleases include RNA guide nuclease, zinc finger nuclease (ZFN), and transcription activator-like effector nuclease (TALEN). The sequence-specific nuclease may be, for example, an RNA guide nuclease, that is, a Cas nuclease, and specifically may be Cas9. In a specific embodiment of the above method, the nuclease cleavage site is no more than about 11 nucleotides away from the mutation site. In some embodiments, mRNA encoding Cas9, such as mRNA containing an ARCA cap, is introduced into stem cells (e.g., by electroporation or other means of gene transduction). In some embodiments, the nucleotide encoding the Cas nuclease (e.g., Cas9) is introduced into the hematopoietic stem cell via a viral vector (e.g., a lentiviral vector). In some embodiments, the sgRNA and the Cas9-encoding nucleic acid are present in the same vector. In some embodiments, the sgRNA and the Cas9-encoding nucleic acid are in different vectors.
在本申请方法的一些实施方案中,进一步包括将识别ALAS-2基因的sgRNA引入到所述造血干细胞,例如CD34 +HSPC中。 In some embodiments of the method of the present application, it further includes introducing sgRNA that recognizes the ALAS-2 gene into the hematopoietic stem cells, such as CD34 + HSPC.
一般而言,sgRNA中的“指导序列”是与靶多核苷酸序列具有足够的互补性以与靶序列杂交并指导CRISPR复合物与靶序列的序列特异性结合的任何多核苷酸序列。在一些实施方案中,当使用适当的比对算法最佳比对时,指导序列及其相应靶序列间的互补程度为约或大于约80%、85%、90%、95%、96%、97%、98%、99%或更多。最佳比对可使用用于比对序列的任何适当的算法确定,其非限制性实例包括Smith-Waterman算法、Needleman-Wimsch算法、基于Burrows-Wheeler Transform的算法(例如Burrows Wheeler Aligner)、ClustalW、Clustai X、BLAT、Novoalign(Novocraft Technologies,ELAND((Illumina,San Diego,CA)、SOAP(可在soap.genomics.org.cn获得)和Maq(可在maq.sourceforge.net获得)。在一些实施方案中,指导序列长度可以为约或大于约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、55、60、65、70、75或更多个核苷酸。在一些实施方案中,指导序列长度少于约75、70、65、60、55、50、45、40、35、30、25、20、15、12或更少的核苷酸。指导序列指导CRISPR 复合物与靶序列的序列特异性结合的能力可通过任何适当的测定方法评估。例如,可向具有相应靶序列的宿主细胞提供足以形成CRISPR复合物的CRISPR系统的组件(包括待测试的指导序列),如可通过使用编码CRISPR序列组件的载体转染,随后评估靶序列内的优先切割(如通过如本文所述的Surveyor测定)来进行。同样地,靶多核苷酸序列的切割可在测试管中通过提供靶序列、CRISPR复合物(包含待测试的指导序列和不同于指导序列的对照指导序列)的组件,并比较测试和对照指导序列在靶序列的结合或切割率,以此进行评估。也可以使用本领域技术人员知道的其它测定方法进行上述测定和评估。Generally speaking, the "guide sequence" in sgRNA is any polynucleotide sequence that has sufficient complementarity with the target polynucleotide sequence to hybridize with the target sequence and direct the sequence-specific binding of the CRISPR complex to the target sequence. In some embodiments, when an appropriate alignment algorithm is used for optimal alignment, the degree of complementarity between the guide sequence and its corresponding target sequence is about or greater than about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more. The optimal alignment can be determined using any appropriate algorithm for aligning sequences. Non-limiting examples include Smith-Waterman algorithm, Needleman-Wimsch algorithm, Burrows-Wheeler Transform-based algorithms (such as Burrows Wheeler Aligner), ClustalW, Clustai 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 some implementations In the scheme, the guide sequence length can be about or greater than about 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, 55, 60, 65, 70, 75 or more nucleotides. In some embodiments, the guide sequence length is less than about 75, 70, 65, 60, 55 , 50, 45, 40, 35, 30, 25, 20, 15, 12 or fewer nucleotides. The ability of the guide sequence to direct the sequence-specific binding of the CRISPR complex to the target sequence can be assessed by any appropriate assay method For example, a host cell with the corresponding target sequence can be provided with the components of the CRISPR system (including the guide sequence to be tested) sufficient to form a CRISPR complex, for example, by transfection with a vector encoding the CRISPR sequence component, and then evaluate the target sequence. The preferential cleavage (such as by the Surveyor assay as described herein) is carried out. Similarly, the cleavage of the target polynucleotide sequence can be performed in the test tube by providing the target sequence, the CRISPR complex (containing the guide sequence to be tested and the other The evaluation is carried out by comparing the binding or cleavage rate of the test and the control guide sequence in the target sequence. Other methods known to those skilled in the art can also be used for the above determination and evaluation.
在上述方法的一些实施方案中,所述sgRNA可以是经修饰的,例如,可以是经过化学修饰的,具体地,所述sgRNA是经过核苷酸核糖2’-O-甲基化修饰和/或核苷酸间3’硫代磷酸酯修饰的。“化学修饰的sgRNA”指针对sgRNA进行特殊的化学修饰,例如对其5’和3’末端3个核苷酸的核糖的2’-O-甲基化修饰和/或核苷酸间的3’硫代磷酸酯修饰。例如,所述化学修饰为所述sgRNA的5’端前一个、二个和/或三个碱基和/或3’端的最后一个核苷酸核糖的2’-O-甲基化修饰。在一些实施方案中,所述sgRNA包含5’端的前3个核苷酸和3’端后3个核苷酸核糖的2’-O-甲基修饰和/或核苷酸间的3’硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端前3个和3’端后3个核苷酸中包含核苷酸核糖的2′-O-甲基化修饰且在5’端的前3个和3’端后3个核苷酸间连接中包含硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端的前5个核苷酸及3’端的后5个核苷酸的核糖中包含2’-O-甲基化修饰,并且在5’端的前5个及3’端的后5个核苷酸间连接中包含硫代磷酸酯化修饰。In some embodiments of the above method, the sgRNA may be modified, for example, it may be chemically modified, specifically, the sgRNA is modified by nucleotide ribose 2'-O-methylation and/ Or internucleotide 3'phosphorothioate modified. "Chemically modified sgRNA" refers to the special chemical modification of sgRNA, such as the 2'-O-methylation modification of ribose with 3 nucleotides at the 5'and 3'ends and/or the 3'between nucleotides. 'Phosphorothioate modification. For example, the chemical modification is a 2'-O-methylation modification of one, two and/or three bases before the 5'end of the sgRNA and/or the last nucleotide ribose at the 3'end. In some embodiments, the sgRNA comprises 2'-O-methyl modification of ribose at the first 3 nucleotides at the 5'end and 3 nucleotides at the rear of the 3'end and/or an internucleotide 3'sulfur Phosphorylation modification. In some embodiments, the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end The connection between the 3 nucleotides after the 3'end contains phosphorothioate modification. In some embodiments, the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end The last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
经过化学修饰的sgRNA至少具有以下两个优点。第一、由于sgRNA是单链形式的RNA,其半衰期非常短,进入到细胞后,会迅速降解(最长不超过12小时),而Cas9蛋白结合sgRNA发挥基因编辑作用则至少需要48小时。因此,采用经过化学修饰的sgRNA,进入细胞后,稳定表达,与Cas9蛋白结合后,能高效基因编辑基因组,产生Indels。第二、未经修饰的sgRNA穿透细胞膜能力差,无法有效进入细胞或组织发挥相应功能。而经过了化学修饰的sgRNA穿透细胞膜的能力通常是增强的。在本发明中可以采用本领域中常用的化学修饰方法,只要能够提高sgRNA稳定性(延长半衰期)和提升进入细胞膜能力,均可以使用。除了实施例中使用的具体的化学修饰之外,还包括采 用其它的修饰方法,例如,Deleavey GF1,Damha MJ.Designing chemically modified oligonucleotides for targeted gene silencing.Chem Biol.2012 Aug 24;19(8):937-54,以及Hendel et al.Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells.Nat Biotechnol.2015 Sep;33(9):985-989文献中报道的化学修饰方法。The chemically modified sgRNA has at least the following two advantages. First, because sgRNA is a single-stranded form of RNA, its half-life is very short. After entering the cell, it will be rapidly degraded (up to 12 hours), while it takes at least 48 hours for Cas9 protein to bind sgRNA to perform gene editing. Therefore, using chemically modified sgRNA, after entering the cell, it is stably expressed, and after being combined with the Cas9 protein, the genome can be efficiently gene-edited to produce Indels. Second, unmodified sgRNA has poor ability to penetrate cell membranes and cannot effectively enter cells or tissues to perform corresponding functions. The ability of chemically modified sgRNA to penetrate cell membranes is usually enhanced. In the present invention, chemical modification methods commonly used in the art can be used, as long as they can improve the stability of sgRNA (extend the half-life) and enhance the ability to enter the cell membrane. In addition to the specific chemical modification used in the examples, it also includes the use of other modification methods, for example, Deleavey GF1, Damha MJ. Designing chemically modified oligonucleotides for targeted gene silencing. Chem Biol. 2012 Aug 24; 19(8): 937-54, and Hendel et al. Chemically modified guide RNAs enhancement CRISPR-Cas genome editing in human primary cells. Nat Biotechnol. 2015 Sep; 33(9):985-989 The chemical modification methods reported in the literature.
在上述方法的一个具体实施方案中,所述sgRNA可与所述染色体上的突变位点染色体序列互补或与所述染色体上的突变位点邻近的染色体序列互补。具体地,所述sgRNA可包含与ALAS-2基因的内含子-1中的序列互补的核酸序列。在上述方法的一些实施方案中,所述sgRNA包含的与ALAS-2基因的内含子-1中的序列互补的核酸序列可为约17个至约20个核苷酸长。具体地,所述sgRNA选自下组:SEQ ID NO:1、SEQ ID NO:2、SEQ ID NO:3。In a specific embodiment of the above method, the sgRNA may be complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome. Specifically, the sgRNA may include a nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene. In some embodiments of the above method, the nucleic acid sequence that is complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA may be about 17 to about 20 nucleotides long. Specifically, the sgRNA is selected from the following group: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3.
III.ALAS-2基因修复效率的提高及脱靶率的降低III. The improvement of ALAS-2 gene repair efficiency and the reduction of off-target rate
本申请利用CRISPR/Cas9基因编辑技术,开发出了高效修复ALAS-2基因突变的方法,基因修复效率高达约30%-40%,经过基因修复的细胞的ALAS-2基因表达达到健康供者ALAS-2表达的50%左右,从而可以显著缓解铁幼粒红细胞贫血(例如XLSA)患者的临床表现。此外,如实施例5所示,使用所述方法可在靶标位点(On-target)产生极高的突变频率,远超出空白对照组的突变频率,且接近100%,而在潜在脱靶位点则无法测得基因编辑组与空白对照组的显著差异。因此,根据现有检测技术的检测结果,所述方法不会在所述造血干细胞基因组造成脱靶。但考虑到检测技术检测能力的限制及背景信号的影响,因此所述脱靶率低于1%,例如低于0.5%或低于0.1%。这种极低的脱靶率,可以提升所述方法用于造血干细胞基因修复的安全性。This application uses CRISPR/Cas9 gene editing technology to develop a method for efficiently repairing ALAS-2 gene mutations. The gene repair efficiency is as high as about 30%-40%. The ALAS-2 gene expression of the cells after the gene repair reaches the healthy donor ALAS -2 expression is about 50%, which can significantly alleviate the clinical manifestations of patients with sideroblast anemia (such as XLSA). In addition, as shown in Example 5, using the method can produce extremely high mutation frequency at the target site (On-target), far exceeding the mutation frequency of the blank control group, and close to 100%, while at potential off-target sites Then the significant difference between the gene editing group and the blank control group cannot be measured. Therefore, according to the detection results of the existing detection technology, the method will not cause off-target in the hematopoietic stem cell genome. However, considering the limitation of the detection capability of the detection technology and the influence of the background signal, the off-target rate is less than 1%, for example, less than 0.5% or less than 0.1%. This extremely low off-target rate can improve the safety of the method for gene repair of hematopoietic stem cells.
在一些实施方案中,本申请提供了一种通过基因编辑来校正ALAS-2基因突变,从而增加功能性ALAS-2表达的方法,其中所述基因编辑包括:(a)将包含对应于所述突变序列的单链校正序列的供体DNA导入所述造血干细胞中;(b)将剪切ALAS-2基因的序列特异性核酸酶导入所述造血干细胞中,其中所述供体DNA上的校正序列替代所述造血干细胞的染色体上的突变序列,由此校正所述突变。在一些实施方案中,所述造血干细胞是CD34 +造血干细胞和祖细胞(“HSPC”),或人诱导多能干细胞(hiPSC)。 In some embodiments, the present application provides a method for correcting ALAS-2 gene mutations through gene editing, thereby increasing the expression of functional ALAS-2, wherein the gene editing includes: (a) comprising: The donor DNA of the single-stranded correction sequence of the mutant sequence is introduced into the hematopoietic stem cell; (b) the sequence-specific nuclease that cuts the ALAS-2 gene is introduced into the hematopoietic stem cell, wherein the correction on the donor DNA The sequence replaces the mutant sequence on the chromosome of the hematopoietic stem cell, thereby correcting the mutation. In some embodiments, the hematopoietic stem cells are CD34 + hematopoietic stem and progenitor cells ("HSPC"), or human induced pluripotent stem cells (hiPSC).
在本申请方法的一些实施方案中,所述序列特异性核酸酶为RNA引导核 酸酶,具体地为Cas9。本发明人发现,核酸酶剪切位点距离所述突变位点越近越有助于提高修复效率。在一些实施方案中,核酸酶剪切位点距离所述突变位点不超过约11个核苷酸,例如,约10个,约9个,约8个,约7个,约6个,约5个,约4个,约3个,约2个,约1个核苷酸,或核酸酶剪切位点与所述突变位点重合。在本申请方法的一些实施方案中,所述sgRNA与所述染色体上的突变位点染色体序列互补或与所述染色体上的突变位点邻近的染色体序列互补。在一些实施方案中,所述sgRNA包含与ALAS-2基因的内含子-1中的序列互补的核酸序列。在本申请方法的一些实施方案中,所述sgRNA包含的与ALAS-2基因的内含子-1中的序列互补的核酸序列为17-20个核苷酸长。在一些实施方案中,所述sgRNA选自下组:SEQ ID NO:1-SEQ ID NO:3,优选的为SEQ ID NO:1。In some embodiments of the method of the present application, the sequence-specific nuclease is an RNA-guided nuclease, specifically Cas9. The inventors found that the closer the nuclease cleavage site is to the mutation site, the more helpful it is to improve the repair efficiency. In some embodiments, the nuclease cleavage site is no more than about 11 nucleotides away from the mutation site, for example, about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, about 1 nucleotide, or nuclease cleavage site overlaps the mutation site. In some embodiments of the method of the present application, the sgRNA is complementary to the chromosomal sequence of the mutation site on the chromosome or complementary to the chromosomal sequence adjacent to the mutation site on the chromosome. In some embodiments, the sgRNA comprises a nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene. In some embodiments of the method of the present application, the nucleic acid sequence complementary to the sequence in intron-1 of the ALAS-2 gene contained in the sgRNA is 17-20 nucleotides long. In some embodiments, the sgRNA is selected from the following group: SEQ ID NO: 1-SEQ ID NO: 3, preferably SEQ ID NO: 1.
在本申请方法的一些实施方案中,所述sgRNA是经化学修饰的。具体地,所述sgRNA是经过核苷酸核糖的2’-O-甲基化修饰和/或核苷酸间3’硫代磷酸酯化修饰的,例如,所述化学修饰为所述sgRNA的5’端前一个、二个和/或三个碱基和/或3’端的最后一个核苷酸核糖碱基的2’-O-甲基化修饰。在一些实施方案中,所述sgRNA包含5’端的前3个核苷酸和3’端后3个核苷酸核糖的2’-O-甲基化修饰和/或核苷酸间的3’硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端前3个和3’端后3个核苷酸中包含核苷酸核糖的2′-O-甲基化修饰且在5’端的前3个和3’端后3个核苷酸间连接中包含硫代磷酸酯化修饰。在一些实施方案中,所述sgRNA在5’端的前5个核苷酸及3’端的后5个核苷酸的核糖中包含2’-O-甲基化修饰,并且在5’端的前5个及3’端的后5个核苷酸间连接中包含硫代磷酸酯化修饰。In some embodiments of the methods of the application, the sgRNA is chemically modified. Specifically, the sgRNA is modified by 2'-O-methylation of nucleotide ribose and/or internucleotide 3'phosphorothioate modification, for example, the chemical modification is that of the sgRNA 2'-O-methylation modification of one, two and/or three bases before the 5'end and/or the last nucleotide ribobase at the 3'end. In some embodiments, the sgRNA comprises the first 3 nucleotides at the 5'end and the 3 nucleotides after the 3'end. 2'-O-methylation modification of ribose and/or an internucleotide 3' Phosphorothioate modification. In some embodiments, the sgRNA comprises the 2'-O-methylation modification of the nucleotide ribose in the first 3 nucleotides of the 5'end and the 3 nucleotides after the 3'end and the first 3 nucleotides of the 5'end The connection between the 3 nucleotides after the 3'end contains phosphorothioate modification. In some embodiments, the sgRNA includes a 2'-O-methylation modification in the first 5 nucleotides of the 5'end and the last 5 nucleotides of the 3'end of the ribose, and the first 5 nucleotides of the 5'end The last 5 internucleotide linkages at the 3'end and the 3'end include phosphorothioate modification.
在本申请方法的一些实施方案中,所述供体序列长于校正序列。在一些实施方案中,所述供体序列与校正序列长度相等,为约60个至约200个核苷酸长,例如60个至约180个,60个至约160个,60个至约140个,60个至约120个,60个至约100个,60个至约80个核苷酸长。在一些实施方案中,所述校正序列包含与位于所述突变位点的3'端的靶区域基本上互补的5'臂,和与位于所述突变位点的5'端的靶区域基本上互补的3'臂。所述基本上互补指所述校正序列的5'臂或3'臂分别与所述突变位点的3'端的靶区域或5'端的靶区域具有较高的同源性,例如至少约90%,至少约95%,至少约96%,至少约97%,至少约98%,至少约99%的同源性。最优选所述校正序列的5'臂或3'臂分别与 所述突变位点的3'端的靶区域或5'端的靶区域具有100%的同源性。所述校正序列的5'臂比所述校正序列的3'臂长,所述校正序列的3'臂比所述校正序列的5'臂长,或者所述校正序列的5'臂和所述校正序列的3'臂具有相同的长度。在一些实施方案中,所述供体DNA的序列为SEQ ID NO:4所示。In some embodiments of the method of the application, the donor sequence is longer than the correction sequence. In some embodiments, the donor sequence is the same length as the correction sequence, and is about 60 to about 200 nucleotides in length, for example, 60 to about 180, 60 to about 160, 60 to about 140. One, 60 to about 120, 60 to about 100, 60 to about 80 nucleotides long. In some embodiments, the correction sequence comprises a 5'arm that is substantially complementary to a target region located at the 3'end of the mutation site, and a 5'arm that is substantially complementary to a target region located at the 5'end of the mutation site. 3'arm. The substantially complementary means that the 5'arm or the 3'arm of the correction sequence has high homology with the target region at the 3'end or the target region at the 5'end of the mutation site, for example, at least about 90%. , At least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% homology. Most preferably, the 5'arm or 3'arm of the correction sequence has 100% homology with the target region at the 3'end or the target region at the 5'end of the mutation site, respectively. The 5'arm of the correction sequence is longer than the 3'arm of the correction sequence, the 3'arm of the correction sequence is longer than the 5'arm of the correction sequence, or the 5'arm of the correction sequence and the The 3'arms of the calibration sequence have the same length. In some embodiments, the sequence of the donor DNA is shown in SEQ ID NO: 4.
在一些实施方案中,所述供体DNA是经化学修饰的,例如核苷酸核糖上的2’-O-甲基化修饰、核苷酸间的3’硫代磷酸酯化修饰以及5’端磷酸化修饰。在一些具体实施方案中,所述化学修饰为所述供体DNA5’端前3个核苷酸和3’端后3个核苷酸核糖的2’-O-甲基化修饰和/或核苷酸间的3’硫代磷酸酯化修饰。在一些具体实施方案中,所述化学修饰为所述供体DNA的5’端前一个、二个和/或三个碱基和/或3’端的最后一个核苷酸核糖的2’-O-甲基化修饰。在一些实施方案中,所述供体DNA在5’端前3个和3’端后3个核苷酸中包含核苷酸核糖的2′-O-甲基化修饰且在5’端的前3个和3’端后3个核苷酸间连接中包含硫代磷酸酯化修饰。在一些实施方案中,所述供体DNA在5’端的前5个核苷酸及3’端的后5个核苷酸的核糖中包含2’-O-甲基化修饰,并且在5’端的前5个及3’端的后5个核苷酸间连接中包含硫代磷酸酯化修饰。在一些特定的实施方案中,所述供体DNA包含5’端磷酸化修饰,并且其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰。在一些特定的实施方案中,所述供体DNA包含5’端磷酸化修饰,其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰,并且5’端的前三个核苷酸和3’端的最后3个核苷酸还包含核糖的2’-O-甲基化修饰。In some embodiments, the donor DNA is chemically modified, such as 2'-O-methylation modification on nucleotide ribose, 3'phosphorothioate modification between nucleotides, and 5' End phosphorylation modification. In some embodiments, the chemical modification is a 2'-O-methylation modification and/or a ribose 3 nucleotides before the 5'end and 3 nucleotides after the 3'end of the donor DNA. 3'phosphorothioate modification between glycidyl acids. In some specific embodiments, the chemical modification is one, two and/or three bases before the 5'end of the donor DNA and/or the 2'-O of the last nucleotide ribose at the 3'end. -Methylation modification. In some embodiments, the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose in the 3 nucleotides before the 5'end and the 3 nucleotides after the 3'end, and the donor DNA contains the 2'-O-methylation modification of the nucleotide ribose before the 5'end. The three internucleotide linkages after the 3 and 3'ends contain phosphorothioate modification. In some embodiments, the donor DNA contains a 2'-O-methylation modification in the ribose of the first 5 nucleotides at the 5'end and the last 5 nucleotides at the 3'end, and the ribose at the 5'end The first 5 and the last 5 internucleotide linkages at the 3'end contain phosphorothioate modification. In some specific embodiments, the donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end. In some specific embodiments, the donor DNA contains a phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides of the 5'end and the last 3 nucleotides of the 3'end, and The first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end also contain the 2'-O-methylation modification of ribose.
在本申请方法的一些实施方案中,通过电穿孔(或称电转导)将所述供体DNA,sgRNA和/或编码Cas9的mRNA顺序引入或同时引入到所述造血干细胞中。In some embodiments of the method of the present application, the donor DNA, sgRNA and/or mRNA encoding Cas9 are introduced sequentially or simultaneously into the hematopoietic stem cells by electroporation (or electrotransduction).
在本申请方法的一些实施方案中,所述sgRNA与所述供体DNA,例如ssODN的重量比为约1:12至约12:1,例如,约1:11至约11:1,约1:10至约10:1,约1:9至约9:1,约1:8至约8:1,约1:7至约7:1,约1:6至约6:1。在本申请方法的一些实施方案中,编码所述序列特异性核酸酶的mRNA与所述单链DNA的重量比为约1:12至约12:1,例如,约1:11至约11:1,约1:10至约10:1,约1:9至约9:1,约1:8至约8:1,约1:7至约7:1,约1:6至约6:1。在一些具体实施方案中,所述Cas9 mRNA:sgRNA-1:ssODN是6μg:4μg:6μg、6μg:4μg:8μg、6μg:4μg:10μg、6μg:4μg:12μg。In some embodiments of the method of the present application, the weight ratio of the sgRNA to the donor DNA, such as ssODN, is about 1:12 to about 12:1, for example, about 1:11 to about 11:1, about 1. :10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6:1. In some embodiments of the method of the present application, the weight ratio of the mRNA encoding the sequence-specific nuclease to the single-stranded DNA is about 1:12 to about 12:1, for example, about 1:11 to about 11: 1. About 1:10 to about 10:1, about 1:9 to about 9:1, about 1:8 to about 8:1, about 1:7 to about 7:1, about 1:6 to about 6: 1. In some specific embodiments, the Cas9 mRNA: sgRNA-1: ssODN is 6 μg: 4 μg: 6 μg, 6 μg: 4 μg: 8 μg, 6 μg: 4 μg: 10 μg, 6 μg: 4 μg: 12 μg.
在本申请方法的一个具体实施方案中,通过电穿孔(或称电转导)将所述供体DNA,sgRNA和/或编码Cas9的mRNA顺序引入或同时引入到所述造血干细胞中。所述电转导条件例如250-360V,0.5-1ms;250-300V,0.5-1ms;250V,1ms;250V,2ms;300V,0.5ms;300V,1ms;360V,0.5ms;或360V,1ms。In a specific embodiment of the method of the present application, the donor DNA, sgRNA and/or mRNA encoding Cas9 are introduced sequentially or simultaneously into the hematopoietic stem cells by electroporation (or electrotransduction). The electrical transduction conditions are, for example, 250-360V, 0.5-1ms; 250-300V, 0.5-1ms; 250V, 1ms; 250V, 2ms; 300V, 0.5ms; 300V, 1ms; 360V, 0.5ms; or 360V, 1ms.
IV.基因修复的造血干细胞的植入和分化IV. Implantation and differentiation of gene repaired hematopoietic stem cells
通过本申请上述方法基因修复的造血干细胞,即ALAS-2基因突变(例如该基因的外显子5-11或内含子-1中基因突变,具体例如Int-1-GATA突变)得到修正或修复的造血干细胞,可以回输给铁粒幼红细胞贫血(例如遗传性铁粒幼红细胞贫血,具体例如XLSA)的患者。并且,将通过本申请上述方法基因修复的造血干细胞回输给患者后,所述造血干细胞可在所述患者骨髓中长期定植,并成功重建所述患者造血系统。此外,通过本申请上述方法基因修复的造血干细胞与未经基因修复的造血干细胞在非编辑位点的突变频率无显著差异,因此因脱靶而对患者造成的安全隐患较低。在一些实施方案中,ALAS-2基因突变修复的CD34 +HSPC来源于待治疗个体的外周血(经过或不经过骨髓造血干细胞动员)或从其骨髓中获得。在一些实施方案中,在将ALAS-2基因突变得到修复的所述CD34 +HSPC回输给所述个体之前,使用造血干细胞红系扩增和分化培养基对所述CD34+HSPC群体进行红系扩增和分化,其中,所述造血干细胞红系扩增和分化培养基包括基础培养基,以及生长因子的组合物,其中所述生长因子的组合物包括干细胞生长因子(SCF);白介素3(IL-3)和促红细胞生成素(EPO)。在一些实施方案中,还包括使用红系分化脱核培养基进行造血干细胞红系分化脱核,所述红系分化脱核培养基包含基础培养基、生长因子、以及孕酮受体和糖皮质激素受体的拮抗剂和/或抑制剂。在一些实施方案中,所述红系分化脱核培养基中的生长因子包括促红细胞生成素(EPO),所述孕酮受体和糖皮质激素受体的拮抗剂和/或抑制剂为选自下述化合物(I)~(IV)中的任一种或两种及以上: The hematopoietic stem cells gene-repaired by the above-mentioned method of this application, that is, the ALAS-2 gene mutation (for example, the gene mutation in exons 5-11 or intron-1 of the gene, specifically the Int-1-GATA mutation) is corrected or The repaired hematopoietic stem cells can be returned to patients with sideroblast anemia (for example, hereditary sideroblast anemia, specifically XLSA). In addition, after the hematopoietic stem cells genetically repaired by the above method of the present application are returned to the patient, the hematopoietic stem cells can be colonized in the bone marrow of the patient for a long time, and the hematopoietic system of the patient can be successfully reconstructed. In addition, there is no significant difference in the mutation frequency of non-edited sites between hematopoietic stem cells gene-repaired by the above-mentioned method of this application and hematopoietic stem cells that have not been gene-repaired, so the potential safety hazard to patients due to off-target is low. In some embodiments, the ALAS-2 gene mutation repaired CD34 + HSPC is derived from the peripheral blood of the individual to be treated (with or without bone marrow hematopoietic stem cell mobilization) or obtained from the bone marrow of the individual. In some embodiments, before the CD34+ HSPC whose ALAS-2 gene mutation has been repaired is returned to the individual, the CD34+HSPC population is subjected to erythroidization using hematopoietic stem cell erythroid expansion and differentiation medium. Expansion and differentiation, wherein the hematopoietic stem cell erythroid expansion and differentiation medium includes a basal medium and a composition of growth factors, wherein the composition of growth factors includes stem cell growth factor (SCF); interleukin 3 ( IL-3) and erythropoietin (EPO). In some embodiments, it further includes using erythroid differentiation and denucleation medium for erythroid differentiation and denucleation of hematopoietic stem cells, the erythroid differentiation and denucleation medium comprising a basal medium, growth factors, and progesterone receptors and glucocorticoids Antagonists and/or inhibitors of hormone receptors. In some embodiments, the growth factor in the erythroid differentiation and denucleation medium includes erythropoietin (EPO), and the antagonist and/or inhibitor of the progesterone receptor and glucocorticoid receptor is selected From any one or two or more of the following compounds (I) to (IV):
Figure PCTCN2020112227-appb-000001
Figure PCTCN2020112227-appb-000001
在一些实施方案中,所述造血干细胞红系扩增和分化培养基包含基础培养基和生长因子添加剂,其中所述基础培养基可选自任何无血清基础培养基,例如STEMSPAN TM SFEM II(STEM CELLS TECHNOLOGY Inc.),IMDM(Iscove's Modified Dulbecco's Medium),可选择地补充有ITS(Thermofisher)、L-gulutamin(Thermofisher)、维生素C和/或牛血清白蛋白;其中生长因子添加剂选自IL-3、SCF和EPO中的一个或多个的组合。 In some embodiments, the hematopoietic stem cell erythroid expansion and differentiation medium comprises a basal medium and growth factor additives, wherein the basal medium can be selected from any serum-free basal medium, such as STEMSPAN TM SFEM II (STEM CELLS TECHNOLOGY Inc.), IMDM (Iscove's Modified Dulbecco's Medium), optionally supplemented with ITS (Thermofisher), L-gulutamin (Thermofisher), vitamin C and/or bovine serum albumin; wherein the growth factor additive is selected from IL-3 A combination of one or more of, SCF and EPO.
在上述造血干细胞红系扩增和分化培养基中可以使用任何常用的基础培养基,例如STEMSPAN TM SFEM II(购自STEM CELL TECHONOLOGIES);例如购自Thermo Fisher的IMDM、DF12、Knockout DMEM、RPMI 1640、Alpha  MEM、DMEM等。此外,可以根据需要向这些基础培养基中进一步添加其他成分,例如可以添加ITS(即主要包括胰岛素、人转铁蛋白以及硒元素)、L-谷氨酰胺、维生素C以及牛血清白蛋白。例如可以在IMDM培养基中外加ITS、外加2mM L-谷氨酰胺、外加10-50μg/ml维生素C以及0.5-5质量%的BSA(牛血清白蛋白)。此外,上述DF12可以外加同样浓度的ITS,L-谷氨酰胺,维生素C和牛血清白蛋白。Knockout DMEM可以外加同样浓度的ITS,L-谷氨酰胺,维生素C和牛血清白蛋白,RPMI 1640可以外加同样浓度的ITS,L-谷氨酰胺,维生素C和牛血清白蛋白,Alpha MEM可以外加同样浓度的ITS,L-谷氨酰胺,维生素C和牛血清白蛋白,DMEM也可以外加同样浓度的ITS,L-谷氨酰胺,维生素C和牛血清白蛋白。在此,各种基础培养基中外加的ITS的浓度可以是:胰岛素浓度是0.1mg/ml、人转铁蛋白是0.0055mg/ml、硒元素6.7×10 -6mg/ml。此外,外加的ITS各成分的浓度也可以根据实际需要来调整。ITS可以从Thermofisher购买,并根据需要调节成合适的最终使用浓度。 Any commonly used basic medium can be used in the above hematopoietic stem cell erythroid expansion and differentiation medium, such as STEMSPAN TM SFEM II (purchased from STEM CELL TECHONOLOGIES); for example, IMDM, DF12, Knockout DMEM, RPMI 1640 from Thermo Fisher , Alpha MEM, DMEM, etc. In addition, other components can be further added to these basic media as needed, for example, ITS (that is, mainly including insulin, human transferrin, and selenium), L-glutamine, vitamin C, and bovine serum albumin can be added. For example, ITS, 2mM L-glutamine, 10-50μg/ml vitamin C and 0.5-5 mass% BSA (bovine serum albumin) can be added to the IMDM medium. In addition, the above-mentioned DF12 can be added with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin. Knockout DMEM can be supplemented with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin, RPMI 1640 can be supplemented with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin, and Alpha MEM can be supplemented with the same concentration ITS, L-glutamine, vitamin C and bovine serum albumin, DMEM can also be added with the same concentration of ITS, L-glutamine, vitamin C and bovine serum albumin. Here, the concentration of additional ITS in various basal media can be: insulin concentration is 0.1 mg/ml, human transferrin is 0.0055 mg/ml, selenium element is 6.7×10 -6 mg/ml. In addition, the concentration of each component of ITS added can also be adjusted according to actual needs. ITS can be purchased from Thermofisher and adjusted to the appropriate final use concentration as required.
可以将上述通过基因编辑修复的造血干细胞直接或培养一天或多天后回输给所述铁粒幼红细胞贫血(例如XLSA)患者进行治疗。在一些实施方案中,所述造血干细胞在施用于个体之前培养一天或多天(例如2天、3天、4天、5天、6天、7天、8天)。在一些实施方案中,在将所述造血干细胞回输给患者个体之前,将所述细胞在冷冻条件下储存至少24小时。在一些实施方案中,在冷冻条件下进行储存之前,将所述细胞培养一天或多天(例如2天、3天、4天、5天、6天、7天、8天)。The above-mentioned hematopoietic stem cells repaired by gene editing can be directly or cultured for one or more days and then returned to the sideroblast anemia (for example, XLSA) patient for treatment. In some embodiments, the hematopoietic stem cells are cultured for one or more days (e.g., 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days) before being administered to the individual. In some embodiments, the hematopoietic stem cells are stored in a frozen condition for at least 24 hours before the hematopoietic stem cells are returned to the individual patient. In some embodiments, the cells are cultured for one or more days (e.g., 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days) before being stored under freezing conditions.
在一些实施方案中,所述治疗包括向所述个体施用(比如静脉注射,包括单次静脉注射)≥2x10 6、≥5x10 6、≥1x10 7、≥2x10 7个细胞/kg体重上述ALAS-2基因突变修复的造血干细胞。 In some embodiments, the treatment comprises administering to the individual (such as intravenous injection, comprising a single intravenous injection) ≥2x10 6, ≥5x10 6, ≥1x10 7, ≥2x10 7 cells / kg body weight above ALAS-2 Hematopoietic stem cells repaired by genetic mutations.
随着造血干细胞的增殖和分化,可以检测到亚铁血红素的产生。可以用Benzidine染色:即联苯胺染色,在过氧化氢存在条件下,使联苯胺能够与血红蛋白中的亚铁血红素结合并反应,产生棕色或蓝色沉淀,检测亚铁血红素的合成,以评估基因治疗效果。也可以通过本领域常规方法检测ALAS-2基因和/或蛋白的表达来评估基因治疗的效果。With the proliferation and differentiation of hematopoietic stem cells, the production of heme can be detected. It can be stained with Benzidine: benzidine staining. In the presence of hydrogen peroxide, benzidine can bind and react with the heme in hemoglobin to produce a brown or blue precipitate to detect the synthesis of heme. Assess the effect of gene therapy. The effect of gene therapy can also be evaluated by detecting the expression of ALAS-2 gene and/or protein by conventional methods in the art.
为了进一步评估ALAS-2基因突变修复后的造血干细胞的表型,也可以将分化后的细胞,进行例如Benzidine染色和Wright-Giemsa染色进行评估。健康供者来源的造血干细胞以及经过基因修复的造血干细胞分化后的红细胞 主要是成熟红细胞和网织红细胞,而未经过基因修复的造血干细胞分化出的红细胞以早幼红细胞为主,表明分化停滞在早期阶段,无法分化出成熟的红细胞。通过本领域已知的检测方法,例如Benzidine染色和Wright-Giemsa染色方法可以判断造血干细胞分化情况,从而得以判断造血干细胞ALAS-2基因突变是否得到修正。In order to further evaluate the phenotype of hematopoietic stem cells after ALAS-2 gene mutation repair, the differentiated cells can also be evaluated by, for example, Benzidine staining and Wright-Giemsa staining. The red blood cells differentiated from healthy donors and hematopoietic stem cells that have undergone gene repair are mainly mature red blood cells and reticulocytes, while the red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are mainly promyelocytic red blood cells, indicating that differentiation is stagnant. In the early stage, mature red blood cells cannot be differentiated. The detection methods known in the art, such as Benzidine staining and Wright-Giemsa staining methods, can determine the differentiation of hematopoietic stem cells, so as to determine whether the ALAS-2 gene mutation of hematopoietic stem cells has been corrected.
实施例Example
实施例1:高效基因修复XLSA病人来源的hiPSC中ALAS-2内含子-1点Example 1: Efficient gene repair of ALAS-2 intron-1 point in hiPSC derived from XLSA patients 突变mutation
本实施例涉及利用CRISPR/Cas9系统基因编辑XLSA病人来源的人诱导性多潜能干细胞(Human induced pluripotent stem cells,hiPSC),高效修复ALAS-2内含子-1点突变,位置在(X:55054635[Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C),由于该位点是GATA-1与ALAS-2基因的结合处,因此将该点突变命名为Int-1-GATA。This embodiment relates to the use of CRISPR/Cas9 system to edit XLSA patient-derived human induced pluripotent stem cells (Human induced pluripotent stem cells, hiPSC) to efficiently repair the ALAS-2 intron-1 point mutation, the location is (X: 55054635 [Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C), because this site is the junction of GATA-1 and ALAS-2 genes, so The point mutation is named Int-1-GATA.
为了修复疾病突变,首先利用“CRISPR RGEN TOOLS”软件设计针对Int-1-GATA突变位点附近基因组的sgRNA并合成经过化学修饰的3个sgRNA,其中所述sgRNA中包含的与目的序列互补的序列编码信息分别如下:sgRNA-1:aactctggcaactttacctg(SEQ ID NO:1),sgRNA-2:caactttacctgtggtctgc(SEQ ID NO:2),sgRNA-3:gggctgagcctgcagaccac(SEQ ID NO:3),同时设计用于该突变的供体DNA,该供体DNA序列信息如下:tcccacgccctggtctcagcttggggagtggtcagaccccaatggcgataaactctggcaactttacctgtggtctgcaggctcagccccaagtgct(SEQ ID NO:4),全长97nt,如图1和2所示。Cas9 mRNA编码信息如下:gacaagaagtacagcatcggcctggacatcggcaccaactctgtgggctgggccgtgatcaccgacgagtacaaggtgcccagcaagaaattcaaggtgctgggcaacaccgaccggcacagcatcaagaagaacctgatcggagccctgctgttcgacagcggcgaaacagccgaggccacccggctgaagagaaccgccagaagaagatacaccagacggaagaaccggatctgctatctgcaagagatcttcagcaacgagatggccaaggtggacgacagcttcttccacagactggaagagtccttcctggtggaagaggataagaagcacgagcggcaccccatcttcggcaacatcgtggacgaggtggcctaccacgagaagtaccccaccatctaccacctgagaaagaaactggtggacagcaccgacaaggccgacctgcggctgatctatctggccctggcccacatgatcaagttccggggccacttcctgatcgagggcgacctgaaccccgacaacagcgacgtggacaagctgttcatccagctggtgcagacctacaaccagctgttcgaggaaaaccc catcaacgccagcggcgtggacgccaaggccatcctgtctgccagactgagcaagagcagacggctggaaaatctgatcgcccagctgcccggcgagaagaagaatggcctgttcggcaacctgattgccctgagcctgggcctgacccccaacttcaagagcaacttcgacctggccgaggatgccaaactgcagctgagcaaggacacctacgacgacgacctggacaacctgctggcccagatcggcgaccagtacgccgacctgtttctggccgccaagaacctgtccgacgccatcctgctgagcgacatcctgagagtgaacaccgagatcaccaaggcccccctgagcgcctctatgatcaagagatacgacgagcaccaccaggacctgaccctgctgaaagctctcgtgcggcagcagctgcctgagaagtacaaagagattttcttcgaccagagcaagaacggctacgccggctacattgacggcggagccagccaggaagagttctacaagttcatcaagcccatcctggaaaagatggacggcaccgaggaactgctcgtgaagctgaacagagaggacctgctgcggaagcagcggaccttcgacaacggcagcatcccccaccagatccacctgggagagctgcacgccattctgcggcggcaggaagatttttacccattcctgaaggacaaccgggaaaagatcgagaagatcctgaccttccgcatcccctactacgtgggccctctggccaggggaaacagcagattcgcctggatgaccagaaagagcgaggaaaccatcaccccctggaacttcgaggaagtggtggacaagggcgcttccgcccagagcttcatcgagcggatgaccaacttcgataagaacctgcccaacgagaaggtgctgcccaagcacagcctgctgtacgagtacttcaccgtgtataacgagctgaccaaagtgaaatacgtgaccgagggaatgagaaagcccgccttcctgagcggcgagcagaaaaaggccatcgtggacctgctgttcaagaccaaccggaaagtgaccgtgaagcagctgaaagaggactacttcaagaaaatcgagtgcttcgactccgtggaaatctccggcgtggaagatcggttcaacgcctccctgggcacataccacgatctgctgaaaattatcaaggacaaggacttcctggacaatgaggaaaacgaggacattctggaagatatcgtgctgaccctgacactgtttgaggacagagagatgatcgaggaacggctgaaaacctatgcccacctgttcgacgacaaagtgatgaagcagctgaagcggcggagatacaccggctggggcaggctgagccggaagctgatcaacggcatccgggacaagcagtccggcaagacaatcctggatttcctgaagtccgacggcttcgccaacagaaacttcatgcagctgatccacgacgacagcctgacctttaaagaggacatccagaaagcccaggtgtccggccagggcgatagcctgcacgagcacattgccaatctggccggcagccccgccattaagaagggcatcctgcagacagtgaaggtggtggacgagctcgtgaaagtgatgggccggcacaagcccgagaacatcgtgatcgaaatggccagagagaaccagaccacccagaagggacagaagaacagccgcgagagaatgaagcggatcgaagagggcatcaaagagctgggcagccagatcctgaaagaacaccccgtggaaaacacccagctgcagaacgagaagctgtacctgtactacctgcagaatgggcgggatatgtacgtggaccaggaactggacatcaaccggctgtccgactacgatgtggaccatatcgtgcctcagagctttctgaaggacgactccatcgacaacaaggtgctgaccagaagcgacaagaaccggggcaagagcgacaacgtgccctccgaagaggtcgtgaagaagatgaagaactactggcggcagctgctgaacgccaagctgattacccagagaaagttcgacaatctgaccaaggccgagagaggcggcctgagcgaactggataaggccggcttcatcaagagacagctggtggaaacccggcagatcacaaagcacgtggcacagatcctggactcccggatgaacactaagtacgacgagaatgacaagctgatccgggaagtgaaagtgatcaccctgaagtccaagctggtgtccgatttccggaaggatttccagttttacaaa gtgcgcgagatcaacaactaccaccacgcccacgacgcctacctgaacgccgtcgtgggaaccgccctgatcaaaaagtaccctaagctggaaagcgagttcgtgtacggcgactacaaggtgtacgacgtgcggaagatgatcgccaagagcgagcaggaaatcggcaaggctaccgccaagtacttcttctacagcaacatcatgaactttttcaagaccgagattaccctggccaacggcgagatccggaagcggcctctgatcgagacaaacggcgaaaccggggagatcgtgtgggataagggccgggattttgccaccgtgcggaaagtgctgagcatgccccaagtgaatatcgtgaaaaagaccgaggtgcagacaggcggcttcagcaaagagtctatcctgcccaagaggaacagcgataagctgatcgccagaaagaaggactgggaccctaagaagtacggcggcttcgacagccccaccgtggcctattctgtgctggtggtggccaaagtggaaaagggcaagtccaagaaactgaagagtgtgaaagagctgctggggatcaccatcatggaaagaagcagcttcgagaagaatcccatcgactttctggaagccaagggctacaaagaagtgaaaaaggacctgatcatcaagctgcctaagtactccctgttcgagctggaaaacggccggaagagaatgctggcctctgccggcgaactgcagaagggaaacgaactggccctgccctccaaatatgtgaacttcctgtacctggccagccactatgagaagctgaagggctcccccgaggataatgagcagaaacagctgtttgtggaacagcacaagcactacctggacgagatcatcgagcagatcagcgagttctccaagagagtgatcctggccgacgctaatctggacaaagtgctgtccgcctacaacaagcaccgggataagcccatcagagagcaggccgagaatatcatccacctgtttaccctgaccaatctgggagcccctgccgccttcaagtactttgacaccaccatcgaccggaagaggtacaccagcaccaaagaggtgctggacgccaccctgatccaccagagcatcaccggcctgtacgagacacggatcgacctgtctcagctgggaggcgac。(SEQ ID NO:5)In order to repair disease mutations, first use "CRISPR RGEN TOOLS" software to design sgRNA targeting the genome near the Int-1-GATA mutation site and synthesize 3 chemically modified sgRNAs, where the sgRNA contains a sequence complementary to the target sequence The coding information is as follows: sgRNA-1: aactctggcaactttacctg (SEQ ID NO: 1), sgRNA-2: caactttacctgtggtctgc (SEQ ID NO: 2), sgRNA-3: gggctgagcctgcagaccac (SEQ ID NO: 3), which is also designed for this mutation The donor DNA sequence information is as follows: tcccacgccctggtctcagcttggggagtggtcagaccccaatggcgataaactctggcaactttacctgtggtctgcaggctcagccccaagtgct (SEQ ID NO: 4), the total length is 97 nt, as shown in Figures 1 and 2. Cas9 mRNA encoding information is as follows: gacaagaagtacagcatcggcctggacatcggcaccaactctgtgggctgggccgtgatcaccgacgagtacaaggtgcccagcaagaaattcaaggtgctgggcaacaccgaccggcacagcatcaagaagaacctgatcggagccctgctgttcgacagcggcgaaacagccgaggccacccggctgaagagaaccgccagaagaagatacaccagacggaagaaccggatctgctatctgcaagagatcttcagcaacgagatggccaaggtggacgacagcttcttccacagactggaagagtccttcctggtggaagaggataagaagcacgagcggcaccccatcttcggcaacatcgtggacgaggtggcctaccacgagaagtaccccaccatctaccacctgagaaagaaactggtggacagcaccgacaaggccgacctgcggctgatctatctggccctggcccacatgatcaagttccggggccacttcctgatcgagggcgacctgaaccccgacaacagcgacgtggacaagctgttcatccagctggtgcagacctacaaccagctgttcgaggaaaaccc catcaacgccagcggcgtggacgccaaggccatcctgtctgccagactgagcaagagcagacggctggaaaatctgatcgcccagctgcccggcgagaagaagaatggcctgttcggcaacctgattgccctgagcctgggcctgacccccaacttcaagagcaacttcgacctggccgaggatgccaaactgcagctgagcaaggacacctacgacgacgacctggacaacctgctggcccagatcggcgaccagtacgccgacctgtttctggccgccaagaacctgtccgacgccatcctgctgagcgacatcctgagagtgaacaccgagatcaccaaggcccccctgagcgcctctatgatcaagagatacgacgagcacca ccaggacctgaccctgctgaaagctctcgtgcggcagcagctgcctgagaagtacaaagagattttcttcgaccagagcaagaacggctacgccggctacattgacggcggagccagccaggaagagttctacaagttcatcaagcccatcctggaaaagatggacggcaccgaggaactgctcgtgaagctgaacagagaggacctgctgcggaagcagcggaccttcgacaacggcagcatcccccaccagatccacctgggagagctgcacgccattctgcggcggcaggaagatttttacccattcctgaaggacaaccgggaaaagatcgagaagatcctgaccttccgcatcccctactacgtgggccctctggccaggggaaacagcagattcgcctggatgaccagaaagagcgaggaaaccatcaccccctggaacttcgaggaagtggtggacaagggcgcttccgcccagagcttcatcgagcggatgaccaacttcgataagaacctgcccaacgagaaggtgctgcccaagcacagcctgctgtacgagtacttcaccgtgtataacgagctgaccaaagtgaaatacgtgaccgagggaatgagaaagcccgccttcctgagcggcgagcagaaaaaggccatcgtggacctgctgttcaagaccaaccggaaagtgaccgtgaagcagctgaaagaggactacttcaagaaaatcgagtgcttcgactccgtggaaatctccggcgtggaagatcggttcaacgcctccctgggcacataccacgatctgctgaaaattatcaaggacaaggacttcctggacaatgaggaaaacgaggacattctggaagatatcgtgctgaccctgacactgtttgaggacagagagatgatcgaggaacggctgaaaacctatgcccacctgttcgacgacaaagtgatgaagcagctgaagcggcggagatacaccggctggggcaggctg agccggaagctgatcaacggcatccgggacaagcagtccggcaagacaatcctggatttcctgaagtccgacggcttcgccaacagaaacttcatgcagctgatccacgacgacagcctgacctttaaagaggacatccagaaagcccaggtgtccggccagggcgatagcctgcacgagcacattgccaatctggccggcagccccgccattaagaagggcatcctgcagacagtgaaggtggtggacgagctcgtgaaagtgatgggccggcacaagcccgagaacatcgtgatcgaaatggccagagagaaccagaccacccagaagggacagaagaacagccgcgagagaatgaagcggatcgaagagggcatcaaagagctgggcagccagatcctgaaagaacaccccgtggaaaacacccagctgcagaacgagaagctgtacctgtactacctgcagaatgggcgggatatgtacgtggaccaggaactggacatcaaccggctgtccgactacgatgtggaccatatcgtgcctcagagctttctgaaggacgactccatcgacaacaaggtgctgaccagaagcgacaagaaccggggcaagagcgacaacgtgccctccgaagaggtcgtgaagaagatgaagaactactggcggcagctgctgaacgccaagctgattacccagagaaagttcgacaatctgaccaaggccgagagaggcggcctgagcgaactggataaggccggcttcatcaagagacagctggtggaaacccggcagatcacaaagcacgtggcacagatcctggactcccggatgaacactaagtacgacgagaatgacaagctgatccgggaagtgaaagtgatcaccctgaagtccaagctggtgtccgatttccggaaggatttccagttttacaaa gtgcgcgagatcaacaactaccaccacgcccacgacgcctacctgaacgccgtcgtgggaacc gccctgatcaaaaagtaccctaagctggaaagcgagttcgtgtacggcgactacaaggtgtacgacgtgcggaagatgatcgccaagagcgagcaggaaatcggcaaggctaccgccaagtacttcttctacagcaacatcatgaactttttcaagaccgagattaccctggccaacggcgagatccggaagcggcctctgatcgagacaaacggcgaaaccggggagatcgtgtgggataagggccgggattttgccaccgtgcggaaagtgctgagcatgccccaagtgaatatcgtgaaaaagaccgaggtgcagacaggcggcttcagcaaagagtctatcctgcccaagaggaacagcgataagctgatcgccagaaagaaggactgggaccctaagaagtacggcggcttcgacagccccaccgtggcctattctgtgctggtggtggccaaagtggaaaagggcaagtccaagaaactgaagagtgtgaaagagctgctggggatcaccatcatggaaagaagcagcttcgagaagaatcccatcgactttctggaagccaagggctacaaagaagtgaaaaaggacctgatcatcaagctgcctaagtactccctgttcgagctggaaaacggccggaagagaatgctggcctctgccggcgaactgcagaagggaaacgaactggccctgccctccaaatatgtgaacttcctgtacctggccagccactatgagaagctgaagggctcccccgaggataatgagcagaaacagctgtttgtggaacagcacaagcactacctggacgagatcatcgagcagatcagcgagttctccaagagagtgatcctggccgacgctaatctggacaaagtgctgtccgcctacaacaagcaccgggataagcccatcagagagcaggccgagaatatcatccacctgtttaccctgaccaatctgggagcccctgccgccttcaagtactttgaca ccaccatcgaccggaagaggtacaccagcaccaaagaggtgctggacgccaccctgatccaccagagcatcaccggcctgtacgagacacggatcgacctgtctcagctgggaggcgac. (SEQ ID NO: 5)
sgRNA的化学合成是指对于sgRNA的5’端的前三个核苷酸以及3’端的最后三个核苷酸的核糖进行了2’-O-甲基化物修饰和核苷酸间的3’硫代磷酸酯化修饰。如下述化学式所示,左侧是化学修饰后的sgRNA,右侧是未经过修饰的sgRNA。所述供体DNA包含5’端磷酸化修饰,并且其5’端的前三个核苷酸和3’端的最后3个核苷酸间包含硫代磷酸酯修饰。Cas9 mRNA和sgRNA均购自美国Trilink Biotechnologies公司。The chemical synthesis of sgRNA refers to the modification of the ribose of the first three nucleotides of the 5'end and the last three nucleotides of the 3'end of the sgRNA with 2'-O-methylation and the 3'sulfur between the nucleotides. Phosphorylation modification. As shown in the following chemical formula, the left side is the chemically modified sgRNA, and the right side is the unmodified sgRNA. The donor DNA contains phosphorylation modification at the 5'end, and phosphorothioate modification is included between the first three nucleotides at the 5'end and the last 3 nucleotides at the 3'end. Both Cas9 mRNA and sgRNA were purchased from Trilink Biotechnologies, USA.
Figure PCTCN2020112227-appb-000002
Figure PCTCN2020112227-appb-000002
为了精确基因修复XLSA病人来源的hiPSC中Int-1-GATA突变,我们首先进行sgRNA切割效率的评估,即的比例。In order to accurately repair the Int-1-GATA mutation in hiPSC derived from XLSA patients, we first evaluated the sgRNA cleavage efficiency, that is, the ratio.
我们扩增培养了XLSA病人来源的hiPSC(中国医学科学院天津血液病研究所提供)。在BTX ECM830电转仪选取了“300V 1ms”的电转条件,将经过合成的Cas9 mRNA和经过化学修饰合成的sgRNA-1、sgRNA-2和sgRNA-3分别电转进入XLSA病人来源的hiPSC中,电转4天后,提取该hiPSC的基因组,选取sgRNA切割位点左右各约450bp,总长度为905bp的片段进行扩增,进行Sanger测序。We expanded and cultivated hiPSC derived from XLSA patients (provided by Tianjin Institute of Hematology, Chinese Academy of Medical Sciences). In the BTX ECM830 electroporation instrument, the electroporation condition of "300V 1ms" was selected, and the synthesized Cas9 mRNA and the chemically modified synthetic sgRNA-1, sgRNA-2 and sgRNA-3 were respectively electroporated into the hiPSC derived from the XLSA patient, and electroporated 4 After days, the genome of the hiPSC was extracted, and fragments of about 450 bp around the sgRNA cleavage site and a total length of 905 bp were selected for amplification, and Sanger sequencing was performed.
正向引物:ctgagcatatcatggccaaa(SEQ ID NO:6)Forward primer: ctgagcatatcatggccaaa (SEQ ID NO: 6)
反向引物:catatggcaacctccttcatc(SEQ ID NO:7)Reverse primer: catatggcaacctccttcatc (SEQ ID NO: 7)
针对测序结果利用“Synthego ICE Analysis”在线软件分析产生Indels效率的统计分析。其中,“Synthego ICE Analysis”在线软件是在线分析Indels效率的软件,以一代测序结果为基础,分析Indels引起的双峰突变的效率,可以参考如下网址:Use "Synthego ICE Analysis" online software to analyze the sequencing results to generate statistical analysis of Indels efficiency. Among them, the "Synthego ICE Analysis" online software is a software for online analysis of Indels efficiency. Based on the first-generation sequencing results, to analyze the efficiency of double-peak mutations caused by Indels, you can refer to the following website:
https://www.synthego.com/products/bioinformatics/crispr-analysis.https://www.synthego.com/products/bioinformatics/crispr-analysis.
结果表明,本实施例中合成的3条sgRNA中,sgRNA-1、sgRNA-2和sgRNA-3的Indels效率分别约为35%、10%和40%,如图3所示。研究表明:第一、高效的Indels效率是基因修复的前提,因此我们选择sgRNA-1和sgRNA-3作为优选的sgRNA;第二、sgRNA的切割位点距离基因修复的位点越近,基 因修复的效率越高(Xiquan Liang,et al.Journal of Biotechnology.2016;Mark A.et al.Science Translational Medicine.2017)。评估切割位点和基因修复位点之间的距离。如图1所示,箭头指针位置即为切割位点,sgRNA-1的切割位点距离基因修复位点为5nt。因此,我们最终确定sgRNA-1为最合适的sgRNA进行后续评估。The results show that among the three sgRNAs synthesized in this example, the Indels efficiency of sgRNA-1, sgRNA-2, and sgRNA-3 are about 35%, 10%, and 40%, respectively, as shown in FIG. 3. Studies have shown that: first, efficient Indels efficiency is the prerequisite for gene repair, so we choose sgRNA-1 and sgRNA-3 as the preferred sgRNA; second, the closer the sgRNA cutting site is to the gene repair site, the closer the gene repair is. The higher the efficiency (Xiquan Liang, et al. Journal of Biotechnology. 2016; Mark A. et al. Science Translational Medicine. 2017). Evaluate the distance between the cutting site and the gene repair site. As shown in Figure 1, the position of the arrow pointer is the cutting site, and the cutting site of sgRNA-1 is 5 nt away from the gene repair site. Therefore, we finally determined sgRNA-1 as the most suitable sgRNA for subsequent evaluation.
针对sgRNA-1,我们对电转相同量的hiPSC(1.0*10^6细胞)进行了Cas9和sgRNA的添加量优化,即分别以以下比例电转Cas9mRNA:sgRNA,1μg:1μg,2μg:2μg,3μg:3μg,4μg:4μg,6μg:6μg进入到hiPSC中,电转4天后,提取该hiPSC的基因组,选取sgRNA切割位点左右各约450bp,总长度为905bp的片段进行扩增,进行Sanger测序。通过“Synthego ICE Analysis”在线软件分析Indels效率。结果表明,随着Cas9 mRNA和sgRNA添加量增加,Indels效率提升,4ug:4ug的效率最高,达到约50%的基因编辑效率,如图4所示。For sgRNA-1, we optimized the amount of Cas9 and sgRNA to electrotransform the same amount of hiPSC (1.0*10^6 cells), that is, electrotransform Cas9mRNA: sgRNA, 1μg:1μg, 2μg:2μg, 3μg, respectively: 3μg, 4μg: 4μg, 6μg: 6μg enter the hiPSC, 4 days after electroporation, extract the genome of the hiPSC, select about 450bp around the sgRNA cleavage site, and a total length of 905bp fragments for amplification and Sanger sequencing. Analyze the efficiency of Indels through the "Synthego ICE Analysis" online software. The results showed that with the increase of Cas9 mRNA and sgRNA, the efficiency of Indels increased, and the efficiency of 4ug:4ug was the highest, reaching about 50% gene editing efficiency, as shown in Figure 4.
为了精确修复hiPSC中Int-1-GATA的点突变,我们按照不同的Cas9:sgRNA:ssODN的添加量,同时将Cas9 mRNA,sgRNA和ssODN电转进入1.0*10^6hiPSC中,4天后,提取该hiPSC的基因组,通过二代测序生物信息学方法分析基因修复效率(HDR)以及Indels效率(NHEJ),如图5所示。Cas9 mRNA:sgRNA:ssODN的测试量分别是1μg:1μg:1μg,2μg:2μg:2μg,3μg:3μg:3μg,4μg:4μg:4μg,6μg:6μg:6μg,结果表明,随着添加量增加,基因修复效率HDR提高,其中基因修复效率HDR最高的是6μg:6μg:6μg,约为25%,由此证明我们成功高效修复了XLSA病人来源的hiPSC中的点突变,在Int-1-GATA位点成功实现了从C到T的修复。Cas9 mRNA:sgRNA:ssODN是6μg:6μg:6μg诱导了较高的Indels效率(%NHEJ,如图5所示)。In order to precisely repair the Int-1-GATA point mutation in hiPSC, we according to the different addition amount of Cas9:sgRNA:ssODN, and at the same time electrotransform Cas9 mRNA, sgRNA and ssODN into 1.0*10^6hiPSC, 4 days later, extract the hiPSC The genome is analyzed by the second-generation sequencing bioinformatics method for gene repair efficiency (HDR) and Indels efficiency (NHEJ), as shown in Figure 5. The test volume of Cas9 mRNA:sgRNA:ssODN is 1μg:1μg:1μg, 2μg:2μg:2μg, 3μg:3μg:3μg, 4μg:4μg:4μg, 6μg:6μg:6μg. The results show that as the amount of addition increases, The gene repair efficiency HDR is improved. The highest gene repair efficiency HDR is 6μg:6μg:6μg, which is about 25%. This proves that we have successfully repaired the point mutations in hiPSC derived from XLSA patients at the Int-1-GATA position. Point successfully realized the repair from C to T. Cas9 mRNA:sgRNA:ssODN is 6μg:6μg:6μg, which induces a higher Indels efficiency (%NHEJ, as shown in Figure 5).
实施例2高效基因修复XLSA病人骨髓来源的CD34+HSPC中ALAS-2Example 2 Efficient gene repair of ALAS-2 in CD34+HSPC derived from bone marrow of XLSA patients 内含子-1点突变Intron-1 point mutation
在实施例1中,我们实现了高效修复XLSA来源的hiPSC的Int-1-GATA点突变,参考实施例1中添加的Cas9 mRNA、sgRNA-1和ssODN的量,在本实验中我们尝试基因修复XLSA病人骨髓来源的CD34+HSPC。In Example 1, we achieved an efficient repair of the Int-1-GATA point mutation of hiPSC derived from XLSA. Refer to the amount of Cas9 mRNA, sgRNA-1 and ssODN added in Example 1. In this experiment, we tried gene repair CD34+HSPC from bone marrow of XLSA patients.
选取300v 1ms的电转条件,分别电转不同Cas9 mRNA,sgRNA-1和ssODN的量进入XLSA病人骨髓来源的CD34+HSPC,其中每1.0*10^6细胞中添加的Cas9 mRNA:sgRNA-1:ssODN是6μg:4μg:6μg和6μg:4μg:10μg,4天后, 提取该HSPC的基因组,通过二代测序生物信息学方法分析基因修复效率(HDR)以及Indels效率(NHEJ),如图6所示。结果表明,在不同Cas9 mRNA、sgRNA-1和ssODN的添加量条件下,我们成功基因修复了XLSA的In-1-GATA突变。此外,在相同Cas9 mRNA和sgRNA-1的添加量条件下,随着供体模板ssODN的量增加,基因修复效率提高,6μg:4μg:10μg条件下基因修复效率达到约30%,提示我们供体模板的添加量会影响基因修复的效率。Select 300v 1ms electroporation conditions, and electrotransform different amounts of Cas9 mRNA, sgRNA-1 and ssODN into CD34+HSPC derived from the bone marrow of XLSA patients, where the Cas9 mRNA:sgRNA-1:ssODN added in every 1.0*10^6 cells is 6 μg: 4 μg: 6 μg and 6 μg: 4 μg: 10 μg, 4 days later, the genome of the HSPC was extracted, and gene repair efficiency (HDR) and Indels efficiency (NHEJ) were analyzed by next-generation sequencing bioinformatics method, as shown in FIG. 6. The results showed that under the conditions of different amounts of Cas9 mRNA, sgRNA-1 and ssODN, we successfully genetically repaired the In-1-GATA mutation of XLSA. In addition, under the same conditions of the addition of Cas9 mRNA and sgRNA-1, as the amount of the donor template ssODN increases, the gene repair efficiency increases, and the gene repair efficiency reaches about 30% under the conditions of 6μg:4μg:10μg, suggesting that our donors The amount of template added will affect the efficiency of gene repair.
为了验证该结论,我们选取300v 1ms的电转条件,分别电转不同Cas9 mRNA,sgRNA-1和ssODN的量进入XLSA病人骨髓来源的CD34+HSPC,其中Cas9 mRNA:sgRNA-1:ssODN是6μg:4μg:12μg,4天后,提取该HSPC的基因组,通过二代测序生物信息学方法分析基因修复效率(HDR)以及Indels效率(NHEJ),如图7所示,基因修复的效率达到约40%,由此表明在XLSA病人来源的CD34+HSPC上,我们成功高效基因修复了疾病突变,达到了现有技术中报道的最高基因修复效率(Mark,et al.Science Translational Medicine.2017;Park,et al.Nucleic Acids Research.2019)。In order to verify this conclusion, we selected 300v 1ms electroporation conditions, and electrotransformed different amounts of Cas9 mRNA, sgRNA-1 and ssODN into the bone marrow-derived CD34+HSPC of XLSA patients. Cas9 mRNA: sgRNA-1: ssODN is 6μg:4μg: 12μg, 4 days later, the HSPC genome was extracted, and the gene repair efficiency (HDR) and Indels efficiency (NHEJ) were analyzed by the next-generation sequencing bioinformatics method. As shown in Figure 7, the gene repair efficiency reached about 40%. It shows that on CD34+HSPC derived from XLSA patients, we successfully repaired disease mutations with high efficiency and reached the highest gene repair efficiency reported in the prior art (Mark, et al. Science Translational Medicine. 2017; Park, et al. Nucleic) Acids Research. 2019).
实施例3红系分化基因修复后的XLSA病人骨髓来源的CD34+HSPCExample 3 CD34+HSPC derived from bone marrow of XLSA patients after erythroid differentiation gene repair 以评估细胞表型改变To assess cell phenotypic changes
3.1红细胞分化3.1 Red blood cell differentiation
参考实施例2中发现的Cas9 mRNA、sgRNA-1和ssODN的添加量(6μg:4μg:12μg),选取300v 1ms的电转条件,分别电转Cas9 mRNA,sgRNA-1和ssODN进入XLSA病人骨髓来源的HSPC,利用下述“两步法”分化方案进行红细胞分化实验。此外,我们红系分化了健康供者动员外周血来源的CD34+HSPC作为阳性对照。Refer to the addition amount of Cas9 mRNA, sgRNA-1 and ssODN found in Example 2 (6μg:4μg:12μg), select 300v 1ms electroporation conditions, and electrotransform Cas9 mRNA, sgRNA-1 and ssODN into HSPC derived from the bone marrow of XLSA patients, respectively , Use the following "two-step method" differentiation protocol for red blood cell differentiation experiments. In addition, our erythroid differentiation of healthy donors mobilized CD34+HSPC derived from peripheral blood as a positive control.
其中两步法分化为利用HSPC红系扩增和分化培养基进行分化,然后利用HSPC红系分化脱核培养基进行分化。The two-step method of differentiation is to use HSPC erythroid amplification and differentiation medium for differentiation, and then use HSPC erythroid differentiation and denucleation medium for differentiation.
造血干细胞红系扩增和分化培养基为基础培养基为StemSpan TM SFEM II,生长因子为50-200ng/ml SCF,10-100ng/ml IL-3,1-10U EPO/ml,培养条件:利用造血干细胞红系扩增和分化培养基培养造血干细胞1.0×10^5细胞/ml,扩增7天。 The erythroid expansion and differentiation medium of hematopoietic stem cells is StemSpan TM SFEM II, the growth factor is 50-200ng/ml SCF, 10-100ng/ml IL-3, 1-10U EPO/ml, culture conditions: use Hematopoietic stem cell erythroid expansion and differentiation medium culture hematopoietic stem cells 1.0×10^5 cells/ml for 7 days.
造血干细胞红系分化脱核培养基为基础培养基为STEMSPAN TM SFEM II,生长因子为1-10U EPO,100-1000μg/ml人转铁蛋白,化学小分子为0.5-10 μm mifepristone,将利用上一步骤培养的1.0×10^6细胞/ml细胞在造血干细胞红系分化脱核培养基分化11天。 The erythroid differentiation and denucleation medium for hematopoietic stem cells is STEMSPAN TM SFEM II, the growth factor is 1-10 U EPO, 100-1000 μg/ml human transferrin, and the chemical small molecule is 0.5-10 μm mifepristone. The 1.0×10^6 cells/ml cells cultured in one step were differentiated in the hematopoietic stem cell erythroid differentiation denucleation medium for 11 days.
我们检测了红系分化后第7天、13天和18天,CD71和CD235a表达比例,如图8所示。结果显示:分化第7天,经过基因修复的细胞CD71和CD235a表达比例最高,达到31.71%;分化第13天,经过基因修复的细胞和健康供者的细胞均高表达CD71和CD235a,比例分别是68.90%和67.55%,显著高于未经过基因修复的细胞,比例是30.96%;分化第18天,经过基因修复的细胞CD71和CD235a的比例未54.48%,显著高于经过基因修复的细胞,比例为25.64%,但是低于健康供者来源的细胞,比例为90.13%。以上结果得出如下结论:1)经过基因修复的细胞在分化第二阶段的分化效率显著高于未经过修复的细胞,表明前者分化的细胞更加成熟,这是因为ALAS-2基因参与了亚铁血红素合成以及红细胞成熟(Zhang,et al.Nucleic Acids Research.2017;Liu,et al.Nature Communications.2018),因此当ALAS-2基因突变被修复后,红细胞分化程度提高。2)经过基因修复的细胞在分化第二阶段的分化效率低于健康供者来源的细胞,这是由于ALAS-2 Int-1-GATA突变被部分基因修复,效率约为40%。We tested the expression ratio of CD71 and CD235a on the 7, 13 and 18 days after erythroid differentiation, as shown in Figure 8. The results showed that on the 7th day of differentiation, the expression ratio of CD71 and CD235a in the cells after gene repair was the highest, reaching 31.71%; on the 13th day of differentiation, the cells after the gene repair and the cells of healthy donors both expressed high CD71 and CD235a, the ratios were respectively 68.90% and 67.55%, which are significantly higher than those of cells that have not undergone gene repair, the proportion is 30.96%; on the 18th day of differentiation, the proportion of CD71 and CD235a cells that have undergone gene repair is less than 54.48%, which is significantly higher than that of cells that have undergone gene repair. It is 25.64%, but is lower than the cells from healthy donors, the ratio is 90.13%. The above results draw the following conclusions: 1) The differentiation efficiency of the gene repaired cells in the second stage of differentiation is significantly higher than that of the unrepaired cells, indicating that the former differentiated cells are more mature. This is because the ALAS-2 gene is involved in ferrous iron. Heme synthesis and red blood cell maturation (Zhang, et al. Nucleic Acids Research. 2017; Liu, et al. Nature Communications. 2018), so when the ALAS-2 gene mutation is repaired, the degree of red blood cell differentiation increases. 2) The differentiation efficiency of gene repaired cells in the second stage of differentiation is lower than that of healthy donor-derived cells. This is because the ALAS-2 Int-1-GATA mutation is partially repaired by the gene, and the efficiency is about 40%.
3.2红细胞分化后,Benzidine染色和Wright-Giemsa染色3.2 After red blood cell differentiation, Benzidine staining and Wright-Giemsa staining
为了进一步评估红系分化基因修复后的造血干细胞的表型,我们将实施例2.1中的分化后的细胞,进行了Benzidine染色和Wright-Giemsa染色,如图9和10所示。实验结果如下:In order to further evaluate the phenotype of hematopoietic stem cells after erythroid differentiation gene repair, we performed Benzidine staining and Wright-Giemsa staining on the differentiated cells in Example 2.1, as shown in Figures 9 and 10. The experimental results are as follows:
1)明场拍照结果表明:健康供者来源的造血干细胞以及经过基因修复的造血干细胞分化后的红细胞呈现明显的红色,表明亚铁血红素合成显著增加,而未经过基因修复的造血干细胞分化出的红细胞呈白色,表明亚铁血红素合成显著缺乏。1) The results of bright-field photography show that the red blood cells differentiated from healthy donors and gene-repaired hematopoietic stem cells show a distinct red color, indicating that the synthesis of heme is significantly increased, while hematopoietic stem cells that have not undergone gene repair are differentiated The red blood cells are white, indicating a significant lack of heme synthesis.
2)Benzidine染色:健康供者来源的造血干细胞以及经过基因修复的造血干细胞分化后的红细胞,Benzidine染色后,阳性细胞比例(红色箭头所示)显著高于未经过基因修复的造血干细胞分化出的红细胞。统计分析结果进一步显示,健康供者来源的造血干细胞以及经过基因修复的造血干细胞分化后的红细胞,Benzidine阳性细胞比例约为60%,而未经过基因修复的造血干细胞分化出的红细胞仅为20%。2) Benzidine staining: hematopoietic stem cells derived from healthy donors and red blood cells differentiated from hematopoietic stem cells that have undergone gene repair. After Benzidine staining, the proportion of positive cells (shown by the red arrow) is significantly higher than that of hematopoietic stem cells that have not undergone gene repair. Red blood cells. The results of statistical analysis further showed that the percentage of Benzidine-positive cells derived from healthy donors and red blood cells differentiated from hematopoietic stem cells that have undergone gene repair is about 60%, while the red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are only 20%. .
3)Wright-Giemsa染色:健康供者来源的造血干细胞以及经过基因修复的造血干细胞分化后的红细胞主要是成熟红细胞和网织红细胞,而未经过基因修复的造血干细胞分化出的红细胞以早幼红细胞为主,表明分化停滞在早期阶段,无法分化出成熟的红细胞。3) Wright-Giemsa staining: red blood cells differentiated from healthy donors and hematopoietic stem cells that have undergone gene repair are mainly mature red blood cells and reticulocytes, while red blood cells differentiated from hematopoietic stem cells that have not undergone gene repair are promyelocytic erythrocytes. Mainly, indicating that differentiation is stagnant in the early stage, and mature red blood cells cannot be differentiated.
3.3检测CD34+HSPC分化来的红细胞中ALAS-2、GATA-1和GAPDH  mRNA和蛋白表达 3.3 Detection of ALAS-2, GATA-1 and GAPDH mRNA and protein expression in red blood cells differentiated from CD34+HSPC
1)将实施例2.2中从CD34+HSPC分化来的红细胞提取细胞的mRNA,反转录成cDNA,通过荧光定量PCR检测ALAS-2、GATA-1、GAPDH基因的mRNA表达,与GAPDH和健康供者的结果进行归一化处理。如图11所示。1) Extract cell mRNA from the red blood cells differentiated from CD34+HSPC in Example 2.2, reverse transcribed into cDNA, and detect the mRNA expression of ALAS-2, GATA-1, GAPDH genes by fluorescent quantitative PCR, and compare them with GAPDH and healthy donors. The result of the person is normalized. As shown in Figure 11.
实验结果表明:经过基因修复的CD34+HSPC分化后的红细胞中ALAS-2基因表达显著高于未经过基因修饰的细胞,而且经过基因修复的细胞的ALAS-2基因表达达到健康供者ALAS-2表达的50%,这与基因修复的效率接近。此外,虽然我们基因修复了ALAS-2中GATA-1的结合位点的基因突变,但是GATA-1基因在三种细胞中表达无显著差异,这与之前的报道一致(Zhang,et al.Nucleic Acids Research.2017)。The experimental results showed that the ALAS-2 gene expression in red blood cells differentiated from gene repaired CD34+HSPC was significantly higher than that of cells without genetic modification, and the ALAS-2 gene expression of the gene repaired cells reached the healthy donor ALAS-2 50% of the expression, which is close to the efficiency of gene repair. In addition, although we genetically repaired the genetic mutation of the GATA-1 binding site in ALAS-2, there was no significant difference in the expression of the GATA-1 gene in the three cells, which is consistent with previous reports (Zhang, et al. Nucleic Acids Research. 2017).
2)将实施例2.2中从CD34+HSPC分化来的红细胞提取细胞的蛋白,进行Western Blot实验,检测ALAS-2、GATA-1、GAPDH基因的蛋白表达,如图12所示。2) Extract cell proteins from the red blood cells differentiated from CD34+HSPC in Example 2.2, and perform Western Blot experiment to detect the protein expression of ALAS-2, GATA-1, and GAPDH genes, as shown in FIG. 12.
实验结果表明:经过基因修复的CD34+HSPC分化后的红细胞中ALAS-2基因表达显著高于未经过基因修饰的细胞。此外,虽然我们基因修复了ALAS-2中GATA-1的结合位点的基因突变,但是GATA-1蛋白在三种细胞中表达无显著差异,这与之前的报道一致(Zhang,et al.Nucleic Acids Research.2017)。The experimental results showed that the ALAS-2 gene expression in red blood cells differentiated from CD34+HSPC after gene repair was significantly higher than that of cells without genetic modification. In addition, although we genetically repaired the genetic mutation of the GATA-1 binding site in ALAS-2, there was no significant difference in the expression of GATA-1 protein in the three cells, which is consistent with previous reports (Zhang, et al. Nucleic Acids Research. 2017).
实施例3:基因修复XLSA病人骨髓来源的CD34+HSPC的体外克隆形成Example 3: In vitro clone formation of CD34+HSPC derived from bone marrow of XLSA patients with gene repair
本实验涉及基因编辑XLSA病人骨髓来源的CD34+HSPC的克隆形成单位(CFU,colony-formation units)检测。This experiment involves the detection of colony-formation units (CFU) of CD34+HSPC derived from the bone marrow of gene-edited XLSA patients.
选取300V 1ms的电转条件,参考实施例2中发现的Cas9 mRNA、sgRNA-1和ssODN的添加量,分别电转Cas9 mRNA,sgRNA-1和ssODN进入XLSA病人骨髓来源的CD34+HSPC,将500个细胞重悬入1ml H4434(购自加拿大STEM CELLS TECHNOLOGIES)和IMDM(购自Thermo Fisher)以及FBS(购自 Thermo Fisher)的混合液中,14天后显微镜下观察CFU-M、BFU-E、CFU-E、CFU-G、CFU-GM、GEMM等不同形态的克隆形成数目,结果如图13所示。其中,BFU-E、CFU-GM、CFU-E、CFU-MM代表红系、髓系、淋巴系等血液系统不同谱系的克隆形成。其中,健康供者代表健康供者动员外周血来源的CD34+HSPC,空白对照:代表未经过基因修复的细胞,基因修复代表经过基因修复的细胞。Select the 300V 1ms electroporation conditions, refer to the Cas9 mRNA, sgRNA-1 and ssODN additions found in Example 2, electroporate Cas9 mRNA, sgRNA-1 and ssODN into CD34+HSPC derived from the bone marrow of the XLSA patient, and 500 cells Resuspend in 1ml H4434 (purchased from Canada STEM CELLS Technologies), IMDM (purchased from Thermo Fisher) and FBS (purchased from Thermo Fisher) in a mixed solution, 14 days later, observe CFU-M, BFU-E, CFU-E under the microscope The number of clones with different morphologies such as CFU-G, CFU-GM, GEMM, etc., and the results are shown in Figure 13. Among them, BFU-E, CFU-GM, CFU-E, CFU-MM represent the clonal formation of different blood system lineages such as erythroid, myeloid, and lymphatic system. Among them, healthy donors represent healthy donors mobilizing CD34+HSPC derived from peripheral blood, blank control: represents cells that have not undergone gene repair, and gene repair represents cells that have undergone gene repair.
实验结果表明:与未经过基因修复的细胞相比,经过基因修复的细胞CFU-GM、BFU-E、CFU-E显著增高,BFU-E和CFU-E分别代表红系的前克隆和终末分化的红系克隆,由此进一步证明基因修复ALAS-2 Int-1-GATA的突变位点恢复了CD34+HSPC红系成为成熟红细胞的能力。此外,由于基因修复效率约为40%,因此经过基因修复的细胞的形成的克隆总数以及不同的亚克隆数目低于健康供者来源的细胞形成的克隆数,符合实验预期。The experimental results show that: compared with cells that have not undergone gene repair, the CFU-GM, BFU-E, and CFU-E of the cells that have undergone gene repair are significantly increased. BFU-E and CFU-E represent the erythroid pre-clone and terminal, respectively The differentiated erythroid clones further proved that gene repairing the mutation site of ALAS-2 Int-1-GATA restored the ability of the CD34+HSPC erythroid to become mature red blood cells. In addition, since the gene repair efficiency is about 40%, the total number of clones formed by the gene repaired cells and the number of different subclones are lower than the number of clones formed by cells from healthy donors, which is in line with experimental expectations.
实施例4基因修复XLSA病人骨髓来源的CD34+HSPC重建小鼠模型的Example 4 Gene repair of CD34+HSPC derived from bone marrow of XLSA patients to reconstruct a mouse model 造血系统Hematopoietic system
本实施选取300V 1ms的电转条件,参考实施例2中发现的Cas9 mRNA、sgRNA-1和ssODN的添加量,电转Cas9 mRNA,sgRNA-1和ssODN进入XLSA病人骨髓来源的CD34+HSPC,移植进入经过辐照仪照射的NPG免疫缺陷小鼠模型(购自北京维通达生物技术有限公司(Beijing Vitalstar Biotechnology,Inc.)。在移植10周、12周、16周后的外周血中检测人CD45和小鼠CD45的表达情况,同时检测移植16周后骨髓、脾脏的人CD45和小鼠CD45的表达情况,其结果如图14所示。其中移植到小鼠中的方法为:在细胞移植24小时前,进行1.0Gy射线照射,清除小鼠模型的骨髓。随后将用20μL 0.9%的生理盐水重悬的1.0×10^6的细胞注射到小鼠的尾静脉中,随后放入洁净级别的动物房中饲养。其中,空白对照:代表未经过基因修复的细胞,基因修复代表经过基因修复的细胞。In this implementation, 300V 1ms electroporation conditions were selected, referring to the addition of Cas9 mRNA, sgRNA-1 and ssODN found in Example 2, electroporation of Cas9 mRNA, sgRNA-1 and ssODN into the bone marrow-derived CD34+HSPC of XLSA patients, the transplantation process The NPG immunodeficiency mouse model irradiated by the irradiator (purchased from Beijing Vitalstar Biotechnology, Inc.). Human CD45 and small blood cells were detected in the peripheral blood 10, 12, and 16 weeks after transplantation. The expression of mouse CD45, and the expression of human CD45 in bone marrow and spleen and mouse CD45 in the bone marrow and spleen 16 weeks after transplantation were detected at the same time. The results are shown in Figure 14. The method of transplantation into mice is: 24 hours before cell transplantation , Irradiated with 1.0Gy rays to clear the bone marrow of the mouse model. Then, 1.0×10^6 cells resuspended with 20μL of 0.9% normal saline were injected into the tail vein of the mouse, and then put into a clean animal room Medium feeding. Among them, blank control: represents cells that have not undergone gene repair, and gene repair represents cells that have undergone gene repair.
图14和图15的结果表明经过基因修复的CD34+HSPC,移植进入小鼠模型后,与未经过基因修复的CD34+HSPC相比,经过基因修饰的细胞移植的动物的外周血、骨髓和脾脏中人源的hCD45表达比例提高,表明经过基因修复的CD34+HSPC能够快速、高效地植入小鼠模型的造血系统,细胞体内分化功能正常,而未经过基因修复的CD34+HSPC在植入小鼠模型造血系统方 面出现功能异常,植入效率低。The results of Figure 14 and Figure 15 show that after the gene repaired CD34+HSPC, after transplantation into the mouse model, compared with the non-genetically repaired CD34+HSPC, the peripheral blood, bone marrow and spleen of the animal after the genetically modified cell transplantation The increased expression ratio of human hCD45 in humans indicates that the gene repaired CD34+HSPC can be quickly and efficiently implanted into the hematopoietic system of the mouse model, and the differentiation function of the cells in vivo is normal, while the CD34+HSPC that has not undergone gene repair is implanted in the mouse model. The mouse model has abnormal functions in the hematopoietic system, and the implantation efficiency is low.
同时,在移植基因修复的细胞的小鼠中,16周后检测了骨髓和脾脏中人源的CD3、CD33、CD19、CD56等细胞膜蛋白的表达如图16和17所示。结果表明,与未经过基因修复的细胞相比,经过基因修复的细胞能正常表达相同比例的CD3、CD56和CD33蛋白,表明两种细胞均可以分化为T细胞、NK细胞和髓系细胞等血液系统的细胞,然而,经过基因修复的细胞高表达CD19蛋白,达到约90%比例,而未经基因修复的细胞CD19蛋白表达显著降低,表达比例低于5%,由此表明经过基因修复的细胞能正常表达CD19蛋白,正常分化为B细胞,而未经过基因修复的细胞B细胞分化显著异常。以上结果进一步证明,经过基因修复的细胞能高效重建小鼠模型的造血系统。At the same time, in the mice transplanted with gene repaired cells, the expression of human-derived CD3, CD33, CD19, CD56 and other cell membrane proteins in the bone marrow and spleen was detected after 16 weeks, as shown in Figures 16 and 17. The results show that compared with cells that have not undergone gene repair, cells that have undergone gene repair can normally express the same proportions of CD3, CD56, and CD33 proteins, indicating that both cells can differentiate into T cells, NK cells, and myeloid cells. However, the cells that have undergone gene repair have high expression of CD19 protein, reaching a ratio of about 90%, while the expression of CD19 protein in cells without gene repair is significantly reduced, and the expression ratio is less than 5%, which indicates that cells that have undergone gene repair It can express CD19 protein normally and differentiate into B cells normally, but the B cells of cells that have not undergone gene repair have significantly abnormal differentiation. The above results further prove that the gene repaired cells can efficiently rebuild the hematopoietic system of the mouse model.
此外,虽然基因编辑的细胞能够快速、高效重建小鼠模型的造血系统。针对重建小鼠模型的细胞是否发生了基因编辑的判断结果如图18所示,提取移植前细胞、移植16周后骨髓的基因组,扩增目的片段,通过二代测序生物信息学方法分析基因修复效率(HDR)以及Indels效率(NHEJ)。结果表明移植16周后骨髓中的人源细胞均发生了高效的基因编辑,基因修复效率与移植前细胞相似,在30~40%之间。In addition, although gene-edited cells can quickly and efficiently rebuild the hematopoietic system of the mouse model. The results of determining whether gene editing occurred in the reconstructed mouse model cells are shown in Figure 18. The genome of the cells before transplantation and the bone marrow 16 weeks after transplantation was extracted, the target fragment was amplified, and the gene repair was analyzed by the next-generation sequencing bioinformatics method. Efficiency (HDR) and Indels efficiency (NHEJ). The results showed that the human-derived cells in the bone marrow had undergone high-efficiency gene editing 16 weeks after transplantation, and the gene repair efficiency was similar to that of the cells before transplantation, ranging from 30 to 40%.
为了进一步评估基因修复ALAS-2的疾病点突变对于CD34+HSPC干性的影响,进行了二次移植试验。上述实验进行到16周时,采集小鼠骨髓细胞,每只小鼠的骨髓移植到2只新的经过辐照后的NPG小鼠体内,称为2次移植。2次移植12周采集小鼠的骨髓进行流式分析人CD45和小鼠CD45的表达情况,其中,空白对照代表未经过基因修复的细胞,基因修复代表经过基因修复的细胞。图19结果表明,与未经过编辑的细胞相比,经过基因修复小鼠的人源重建水平显著提高,进一步证明基因修复ALAS-2后的CD34+HSPC具有长期的干性和分化能力。In order to further evaluate the effect of gene repair ALAS-2 disease point mutations on the stemness of CD34+HSPC, a second transplantation test was carried out. At 16 weeks of the above experiment, mouse bone marrow cells were collected, and the bone marrow of each mouse was transplanted into two new irradiated NPG mice, which was called 2 transplantation. The bone marrow of mice was collected for 12 weeks after two transplants for flow cytometric analysis of the expression of human CD45 and mouse CD45. The blank control represents cells that have not undergone gene repair, and gene repair represents cells that have undergone gene repair. The results in Figure 19 show that compared with unedited cells, the level of human reconstitution in genetically repaired mice is significantly improved, which further proves that CD34+HSPC after genetic repair of ALAS-2 has long-term stemness and differentiation capabilities.
此外提取二次移植小鼠12周后骨髓的基因组,扩增目的片段,通过二代测序生物信息学方法分析基因修复效率(HDR)以及Indels效率(NHEJ)。结果表明移植12周后骨髓中的人源细胞均具有高效的基因编辑,基因修复效率与移植前细胞相似,在40%左右,如图20所示。In addition, the genome of the bone marrow of the mice after the second transplantation was extracted 12 weeks, the target fragment was amplified, and the gene repair efficiency (HDR) and the Indels efficiency (NHEJ) were analyzed by second-generation sequencing bioinformatics methods. The results showed that the human-derived cells in the bone marrow 12 weeks after transplantation all had high-efficiency gene editing, and the gene repair efficiency was similar to that of the cells before transplantation, about 40%, as shown in Figure 20.
实施例5潜在脱靶效应分析Example 5 Analysis of potential off-target effects
为了评估基因修复ALAS-2后的潜在脱靶效应,通过同时采用序列相似 性预测分析以及无偏全基因组分析方法Digenome-Seq方法(Daesik Kim,et al.Nature Methods.2014),寻找sgRNA最可能引起脱靶的潜在脱靶位点(potential off-target)共32个,POT代表潜在脱靶位点。由于病人来源的CD34+HSPC数量限制,因此选用基因编辑的人潜能干细胞(hiPSCs)为目的细胞进行验证,对潜在脱靶位点的情况进行定点检测。如图21所示,其中,空白对照代表未经过基因编辑的细胞,基因修复代表经过基因编辑的细胞。结果表明,与空白对照组的细胞相比,经过基因编辑的细胞在32个潜在脱靶位点的突变频率无显著差异,进一步表明无潜在脱靶现象发生。In order to evaluate the potential off-target effects after gene repair ALAS-2, the Digenome-Seq method (Daesik Kim, et al. Nature Methods. 2014) was used to find the most likely cause of sgRNA by using both sequence similarity prediction analysis and unbiased whole-genome analysis methods. There are 32 potential off-targets, and POT stands for potential off-target. Due to the limited number of CD34+HSPCs from patients, gene-edited human potential stem cells (hiPSCs) were selected as the target cells for verification, and spot detection of potential off-target sites was carried out. As shown in Figure 21, the blank control represents cells that have not undergone gene editing, and gene repair represents cells that have undergone gene editing. The results showed that compared with the cells in the blank control group, there was no significant difference in the mutation frequency of the gene-edited cells at 32 potential off-target sites, further indicating that no potential off-target phenomenon occurred.
工业实用性Industrial applicability
根据本发明,本发明的方法有以下几个优点,第一、本方法能够基因编辑并高效修复XLSA病人来源的hiPSC以及骨髓来源的CD34+HSPC,满足了临床治疗X-链锁环状铁粒幼红细胞贫血的治疗要求;第二、基因修复效率高,显著提高ALAS-2基因和蛋白表达,显著提高分化的红细胞中亚铁血红素的合成;第三、基因修复的造血干细胞能够高效重建模型小鼠的造血系统;第四、基因编辑后的细胞无潜在的脱靶现象发生。基于此,本发明开发的方法将有可能代替传统的造血干细胞移植治疗技术来治愈X-链锁环状铁粒幼红细胞贫血的患者。According to the present invention, the method of the present invention has the following advantages. First, the method can gene-edit and efficiently repair hiPSC derived from XLSA patients and CD34+HSPC derived from bone marrow, which meets the needs of clinical treatment of X-chain cyclic iron particles. Immature red blood cell anemia treatment requirements; second, high gene repair efficiency, significantly increase the expression of ALAS-2 gene and protein, and significantly increase the synthesis of heme in differentiated red blood cells; third, gene repaired hematopoietic stem cells can efficiently reconstruct models The hematopoietic system of mice; fourth, the cells after gene editing have no potential off-target phenomenon. Based on this, the method developed by the present invention may replace traditional hematopoietic stem cell transplantation treatment techniques to cure patients with X-chain ring sideroblast anemia.

Claims (25)

  1. 一种通过CRISPS/Cas9基因编辑校正造血干细胞的5-氨基乙酰丙酸合酶2(ALAS-2)基因突变的方法,包括:将包含对应于ALAS-2突变序列的单链校正序列的供体DNA、识别ALAS-2突变序列的sgRNA和编码Cas9蛋白的核酸序列导入所述造血干细胞,由此所述供体DNA中的校正序列替代所述造血干细胞中的ALAS-2突变序列。A method for correcting 5-aminolevulinic acid synthase 2 (ALAS-2) gene mutations of hematopoietic stem cells through CRISPS/Cas9 gene editing, including: adding a donor containing a single-stranded correction sequence corresponding to the ALAS-2 mutation sequence DNA, sgRNA that recognizes the ALAS-2 mutation sequence, and the nucleic acid sequence encoding the Cas9 protein are introduced into the hematopoietic stem cell, whereby the correction sequence in the donor DNA replaces the ALAS-2 mutation sequence in the hematopoietic stem cell.
  2. 权利要求1的方法,其中所述造血干细胞是CD34 +HSPC。 The method of claim 1, wherein said hematopoietic stem cells are CD34 + HSPC.
  3. 权利要求1或2的方法,其中所述ALAS-2突变序列是ALAS-2基因外显子5-11中的突变序列和/或ALAS-2基因内含子-1中的突变序列。The method of claim 1 or 2, wherein the ALAS-2 mutant sequence is a mutant sequence in exons 5-11 of the ALAS-2 gene and/or a mutant sequence in intron-1 of the ALAS-2 gene.
  4. 权利要求1-3任一项的方法,其中所述ALAS-2突变序列位于ALAS-2基因内含子-1中。The method of any one of claims 1-3, wherein the ALAS-2 mutant sequence is located in intron-1 of the ALAS-2 gene.
  5. 权利要求4的方法,其中所述突变为ALAS2内含子-1中的点突变:X:55054635[Chr X(GRCh37/hg19):g.55054635 A>G,NM 000032.4:c.-15–2187 T>C。The method of claim 4, wherein the mutation is a point mutation in ALAS2 intron-1: X:55054635[Chr X(GRCh37/hg19):g.55054635A>G,NM 000032.4:c.-15-2187 T>C.
  6. 权利要求4或5的方法,其中Cas9剪切位点距离ALAS-2突变位点不超过约11个核苷酸的位点。The method of claim 4 or 5, wherein the Cas9 cleavage site is no more than about 11 nucleotides away from the ALAS-2 mutation site.
  7. 权利要求1-6中任一项的方法,其中所述sgRNA为约17个至约20个核苷酸长。The method of any one of claims 1-6, wherein the sgRNA is about 17 to about 20 nucleotides in length.
  8. 权利要求7的方法,其中所述sgRNA是经化学修饰的。The method of claim 7, wherein the sgRNA is chemically modified.
  9. 权利要求8的方法,其中所述sgRNA的修饰包括核苷酸核糖上的2’-O-甲基化修饰或核苷酸间的3’硫代磷酸化修饰或二者。The method of claim 8, wherein the modification of the sgRNA includes 2'-O-methylation modification on the nucleotide ribose or internucleotide 3'phosphorothioate modification or both.
  10. 权利要求9的方法,其中所述修饰为5’端的前三个核苷酸核糖上的2’-O-甲基化修饰、3’端的最后三个核苷酸核糖上的2’-O-甲基化修饰、5’端的前三个核苷酸的核苷酸间3’硫代磷酸化修饰和3’端的最后三个核苷酸的核苷酸间3’硫代磷酸化修饰。The method of claim 9, wherein the modification is a 2'-O-methylation modification on the first three nucleotides ribose at the 5'end, and 2'-O- on the last three nucleotides ribose at the 3'end. Methylation modification, the internucleotide 3'phosphorothioate modification of the first three nucleotides at the 5'end, and the internucleotide 3'phosphorothioate modification of the last three nucleotides at the 3'end.
  11. 权利要求1-10任一项的方法,其中所述sgRNA的序列选自下组:SEQ ID NO:1、SEQ ID NO:2和SEQ ID NO:3。The method of any one of claims 1-10, wherein the sequence of the sgRNA is selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3.
  12. 权利要求1-11中任一项的方法,其中所述校正序列为约60个至约200个核苷酸长。The method of any one of claims 1-11, wherein the correction sequence is about 60 to about 200 nucleotides long.
  13. 权利要求12的方法,其中所述校正序列包含与位于所述突变位点的 3'的靶区域互补的5'臂,和与位于所述突变位点的5'的靶区域互补的3'臂,其中所述校正序列的5'臂为约40个至约60个核苷酸长,校正序列的3'臂为约20个至约50个核苷酸长。The method of claim 12, wherein the correction sequence comprises a 5'arm complementary to the target region 3'of the mutation site, and a 3'arm complementary to the target region 5'of the mutation site , Wherein the 5'arm of the calibration sequence is about 40 to about 60 nucleotides long, and the 3'arm of the calibration sequence is about 20 to about 50 nucleotides long.
  14. 权利要求13的方法,其中所述供体DNA是经过化学修饰的。The method of claim 13, wherein the donor DNA is chemically modified.
  15. 权利要求14的方法,其中所述化学修饰包括核苷酸核糖上的2’-O-甲基化修饰或核苷酸间的3’硫代磷酸化修饰或二者。The method of claim 14, wherein the chemical modification comprises a 2'-O-methylation modification on the nucleotide ribose or an internucleotide 3'phosphorothioate modification or both.
  16. 权利要求15的方法,其中所述修饰为5’端的前三个核苷酸的核苷酸间3’硫代磷酸化修饰和3’端的最后三个核苷酸的核苷酸间3’硫代磷酸化修饰。The method of claim 15, wherein the modification is an internucleotide 3'phosphorothioate modification of the first three nucleotides at the 5'end and an internucleotide 3'phosphorylation modification of the last three nucleotides at the 3'end. Generation phosphorylation modification.
  17. 权利要求15或16的方法,其中所述修饰还包括5’端磷酸化修饰。The method of claim 15 or 16, wherein the modification further comprises a 5'end phosphorylation modification.
  18. 权利要求17的方法,其中校正序列与除所述突变位点外的ChrX:55028172-55028268处的靶序列互补。The method of claim 17, wherein the correction sequence is complementary to the target sequence at ChrX:55028172-55028268 except for the mutation site.
  19. 权利要求1-18任一项的方法,其中所述供体DNA序列如SEQ ID NO:4所示。The method of any one of claims 1-18, wherein the donor DNA sequence is shown in SEQ ID NO: 4.
  20. 权利要求1-19中任一项的方法,其中通过电穿孔或转导方式将所述sgRNA、供体DNA和编码Cas9蛋白的核酸序列导入到所述造血干细胞中。The method of any one of claims 1-19, wherein the sgRNA, the donor DNA and the nucleic acid sequence encoding the Cas9 protein are introduced into the hematopoietic stem cell by electroporation or transduction.
  21. 权利要求1-20中任一项的方法,其中所述sgRNA与所述供体DNA的重量比为约4:12。The method of any one of claims 1-20, wherein the weight ratio of the sgRNA to the donor DNA is about 4:12.
  22. 权利要求1-21中任一项的方法,其中编码所述Cas9的mRNA与所述供体DNA的重量比为约4:12。The method of any one of claims 1-21, wherein the weight ratio of the mRNA encoding the Cas9 to the donor DNA is about 4:12.
  23. 权利要求1-22中任一项的方法,其中向约1.0*10^6个造血干细胞中导入Cas9 mRNA、sgRNA、供体DNA的重量选自如下任一组:The method of any one of claims 1-22, wherein the weight of Cas9 mRNA, sgRNA, and donor DNA introduced into about 1.0*10^6 hematopoietic stem cells is selected from any of the following groups:
    1)6μg、4μg、6μg;1) 6μg, 4μg, 6μg;
    2)6μg、4μg、8μg;2) 6μg, 4μg, 8μg;
    3)6μg、4μg、10μg;3) 6μg, 4μg, 10μg;
    4)6μg、4μg、12μg。4) 6μg, 4μg, 12μg.
  24. 权利要求1-22中任一项的方法,其中Cas9 mRNA:sgRNA:供体DNA的重量比为1:1:1。The method of any one of claims 1-22, wherein the weight ratio of Cas9 mRNA: sgRNA: donor DNA is 1:1:1.
  25. 权利要求20-24中任一项所述的方法,其中所述Cas9 mRNA、sgRNA、供体DNA通过电穿孔导入造血干细胞中,所述电穿孔条件为300V,1ms。The method of any one of claims 20-24, wherein the Cas9 mRNA, sgRNA, and donor DNA are introduced into hematopoietic stem cells by electroporation, and the electroporation conditions are 300V, 1ms.
PCT/CN2020/112227 2019-08-29 2020-08-28 Method for efficiently repairing gene mutation of ring sideroblastic anemia WO2021037234A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080056107.1A CN114222816A (en) 2019-08-29 2020-08-28 Method for efficiently repairing gene mutation of cricoid sideroblasts

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2019/103235 2019-08-29
CN2019103235 2019-08-29

Publications (1)

Publication Number Publication Date
WO2021037234A1 true WO2021037234A1 (en) 2021-03-04

Family

ID=74684653

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/112227 WO2021037234A1 (en) 2019-08-29 2020-08-28 Method for efficiently repairing gene mutation of ring sideroblastic anemia

Country Status (2)

Country Link
CN (1) CN114222816A (en)
WO (1) WO2021037234A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002010454A2 (en) * 2000-07-28 2002-02-07 Genaissance Pharmaceuticals, Inc. Haplotypes of the alas2 gene
WO2017079591A2 (en) * 2015-11-04 2017-05-11 Acceleron Pharma Inc. Methods for increasing red blood cell levels and treating ineffective erythropoiesis
CN107949641A (en) * 2015-06-17 2018-04-20 Uab研究基金会 CRISPR/CAS9 compounds for genome editor
CN109735497A (en) * 2017-10-27 2019-05-10 博雅辑因(北京)生物科技有限公司 A method of improving fetal hemoglobin expression

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002010454A2 (en) * 2000-07-28 2002-02-07 Genaissance Pharmaceuticals, Inc. Haplotypes of the alas2 gene
CN107949641A (en) * 2015-06-17 2018-04-20 Uab研究基金会 CRISPR/CAS9 compounds for genome editor
WO2017079591A2 (en) * 2015-11-04 2017-05-11 Acceleron Pharma Inc. Methods for increasing red blood cell levels and treating ineffective erythropoiesis
CN109735497A (en) * 2017-10-27 2019-05-10 博雅辑因(北京)生物科技有限公司 A method of improving fetal hemoglobin expression
CN109735574A (en) * 2017-10-27 2019-05-10 博雅辑因(北京)生物科技有限公司 A method of improving fetal hemoglobin expression

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KANEKO KIRIKO, KUBOTA YOSHIKO, NOMURA KAZUMI, HAYASHIMOTO HARUKA, CHIDA TAISEI, YOSHINO NAOTO, WAYAMA MARINA, OGASAWARA KATSUTOSHI: "Establishment of a Cell Model of X-linked Sideroblastic Anemia Using Genome Editing", EXPERIMENTAL HEMATOLOGY, vol. 65, 1 September 2018 (2018-09-01), pages 57 - 68e2, XP055787462, ISSN: 0301-472X, DOI: 10.1016/j.exphem.2018.06.002 *
YINGCHI ZHANG, JINGLIAO ZHANG, WENBIN AN, YANG WAN, SHIHUI MA, JIE YIN, XICHUAN LI, JIE GAO, WEIPING YUAN, YE GUO, JAMES DOUGLAS E: "Intron 1 GATA Site Enhances ALAS2 Expression Indispensably During Erythroid Differentiation", NUCLEIC ACIDS RESEARCH, vol. 45, no. 2, 7 October 2016 (2016-10-07), pages 657 - 671, XP055787458, ISSN: 0305-1048, DOI: 10.1093/nar/gkw901 *

Also Published As

Publication number Publication date
CN114222816A (en) 2022-03-22

Similar Documents

Publication Publication Date Title
CN111278976B (en) Method for improving fetal hemoglobin expression
WO2020168300A1 (en) Modified natural killer (nk) cells for immunotherapy
JP2018518182A (en) CRISPR / CAS9 complex for genome editing
JP2018518181A (en) CRISPR / Cas9 complex for introducing functional polypeptides into cells of the blood cell lineage
CA3091136A1 (en) Method for producing low-antigenic cell
Ozakpinar et al. Ovarian stem cells: From basic to clinical applications
EP3964579A1 (en) Method for predicting effectiveness of treatment of hemoglobinopathy
JP2022547105A (en) Evaluation method of gene editing therapy based on off-target evaluation
WO2021037234A1 (en) Method for efficiently repairing gene mutation of ring sideroblastic anemia
CN112442516A (en) Method for efficiently repairing gene mutation of cricoid sideroblasts
CA3199435A1 (en) Methods of inducing antibody-dependent cellular cytotoxicity (adcc) using modified natural killer (nk) cells
TW202100752A (en) Method for predicting the effectiveness of the treatment of hemoglobinopathies
WO2024059824A2 (en) Immune cells with combination gene perturbations
Yang et al. In situ correction of various β-thalassemia mutations in human hematopoietic stem cells
WO2023220364A2 (en) Improved methods and compositions for transgene delivery and/or reconstituting microglia
IL302315A (en) Safe harbor loci

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: 20857799

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20857799

Country of ref document: EP

Kind code of ref document: A1