CN114934066A - Gene editing system for osteolithiasis and application thereof - Google Patents
Gene editing system for osteolithiasis and application thereof Download PDFInfo
- Publication number
- CN114934066A CN114934066A CN202210455336.1A CN202210455336A CN114934066A CN 114934066 A CN114934066 A CN 114934066A CN 202210455336 A CN202210455336 A CN 202210455336A CN 114934066 A CN114934066 A CN 114934066A
- Authority
- CN
- China
- Prior art keywords
- editing system
- gene editing
- nucleotide sequence
- seq
- vector
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Granted
Links
- 238000010362 genome editing Methods 0.000 title claims abstract description 28
- 230000035772 mutation Effects 0.000 claims abstract description 26
- 239000002773 nucleotide Substances 0.000 claims abstract description 19
- 125000003729 nucleotide group Chemical group 0.000 claims abstract description 19
- 238000000034 method Methods 0.000 claims abstract description 16
- 108091027544 Subgenomic mRNA Proteins 0.000 claims abstract description 15
- 230000034431 double-strand break repair via homologous recombination Effects 0.000 claims abstract description 9
- 238000010453 CRISPR/Cas method Methods 0.000 claims abstract description 7
- 108091028043 Nucleic acid sequence Proteins 0.000 claims abstract description 5
- 239000013598 vector Substances 0.000 claims description 40
- 210000004027 cell Anatomy 0.000 claims description 33
- 230000008685 targeting Effects 0.000 claims description 21
- 239000013612 plasmid Substances 0.000 claims description 17
- 108091033409 CRISPR Proteins 0.000 claims description 15
- 210000004263 induced pluripotent stem cell Anatomy 0.000 claims description 11
- 238000002360 preparation method Methods 0.000 claims description 7
- 101150092724 CLCN7 gene Proteins 0.000 claims description 6
- 230000006801 homologous recombination Effects 0.000 claims description 4
- 238000002744 homologous recombination Methods 0.000 claims description 4
- 206010007027 Calculus urinary Diseases 0.000 claims description 2
- 238000002659 cell therapy Methods 0.000 claims description 2
- 238000012216 screening Methods 0.000 claims description 2
- 208000008281 urolithiasis Diseases 0.000 claims description 2
- 238000004519 manufacturing process Methods 0.000 claims 1
- 230000008439 repair process Effects 0.000 abstract description 22
- 208000005368 osteomalacia Diseases 0.000 abstract description 6
- 206010031252 Osteomyelitis Diseases 0.000 abstract description 4
- 238000013461 design Methods 0.000 abstract description 4
- 108020005004 Guide RNA Proteins 0.000 abstract description 2
- 230000009286 beneficial effect Effects 0.000 abstract description 2
- 230000008569 process Effects 0.000 abstract description 2
- 230000014509 gene expression Effects 0.000 description 8
- 208000015924 Lithiasis Diseases 0.000 description 7
- 108020004414 DNA Proteins 0.000 description 6
- 238000010586 diagram Methods 0.000 description 6
- 102100028685 H(+)/Cl(-) exchange transporter 7 Human genes 0.000 description 5
- 101000766971 Homo sapiens H(+)/Cl(-) exchange transporter 7 Proteins 0.000 description 5
- 239000002609 medium Substances 0.000 description 5
- 238000010354 CRISPR gene editing Methods 0.000 description 4
- 238000012163 sequencing technique Methods 0.000 description 4
- 210000000130 stem cell Anatomy 0.000 description 4
- 238000011144 upstream manufacturing Methods 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 239000012634 fragment Substances 0.000 description 3
- 230000003211 malignant effect Effects 0.000 description 3
- 108010082117 matrigel Proteins 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- 230000008672 reprogramming Effects 0.000 description 3
- 102100022900 Actin, cytoplasmic 1 Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 238000012408 PCR amplification Methods 0.000 description 2
- 206010043276 Teratoma Diseases 0.000 description 2
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 239000004202 carbamide Substances 0.000 description 2
- 210000000845 cartilage Anatomy 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 210000003981 ectoderm Anatomy 0.000 description 2
- 230000000694 effects Effects 0.000 description 2
- 210000001900 endoderm Anatomy 0.000 description 2
- 239000013613 expression plasmid Substances 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 239000012737 fresh medium Substances 0.000 description 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 210000002490 intestinal epithelial cell Anatomy 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 210000003716 mesoderm Anatomy 0.000 description 2
- 230000006780 non-homologous end joining Effects 0.000 description 2
- 238000002660 stem cell treatment Methods 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 238000002636 symptomatic treatment Methods 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000012795 verification Methods 0.000 description 2
- 238000010356 CRISPR-Cas9 genome editing Methods 0.000 description 1
- 238000007400 DNA extraction Methods 0.000 description 1
- 230000033616 DNA repair Effects 0.000 description 1
- 101001139134 Homo sapiens Krueppel-like factor 4 Proteins 0.000 description 1
- 101000687905 Homo sapiens Transcription factor SOX-2 Proteins 0.000 description 1
- 208000026350 Inborn Genetic disease Diseases 0.000 description 1
- 102100020677 Krueppel-like factor 4 Human genes 0.000 description 1
- 241000711408 Murine respirovirus Species 0.000 description 1
- 241000594446 Neurocalyx Species 0.000 description 1
- 102100035423 POU domain, class 5, transcription factor 1 Human genes 0.000 description 1
- 101710126211 POU domain, class 5, transcription factor 1 Proteins 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102100024270 Transcription factor SOX-2 Human genes 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 208000021018 autosomal dominant inheritance Diseases 0.000 description 1
- 230000037182 bone density Effects 0.000 description 1
- 101150038500 cas9 gene Proteins 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 230000005782 double-strand break Effects 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 238000007877 drug screening Methods 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 238000010363 gene targeting Methods 0.000 description 1
- 208000016361 genetic disease Diseases 0.000 description 1
- 210000001654 germ layer Anatomy 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 238000011134 hematopoietic stem cell transplantation Methods 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000013600 plasmid vector Substances 0.000 description 1
- 210000001778 pluripotent stem cell Anatomy 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 238000002689 xenotransplantation Methods 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/465—Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0696—Artificially induced pluripotent stem cells, e.g. iPS
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
- C12N9/22—Ribonucleases RNAses, DNAses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/20—Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2800/00—Nucleic acids vectors
- C12N2800/10—Plasmid DNA
- C12N2800/106—Plasmid DNA for vertebrates
- C12N2800/107—Plasmid DNA for vertebrates for mammalian
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- Molecular Biology (AREA)
- Medicinal Chemistry (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Microbiology (AREA)
- Public Health (AREA)
- Biophysics (AREA)
- Physical Education & Sports Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Gastroenterology & Hepatology (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- General Chemical & Material Sciences (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Immunology (AREA)
- Cell Biology (AREA)
- Toxicology (AREA)
- Transplantation (AREA)
- Developmental Biology & Embryology (AREA)
- Rheumatology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
The invention discloses a gene editing system for osteomalacia and application thereof. The gene editing system comprises: the CRISPR/Cas editing system comprises sgRNAs, wherein the nucleotide sequences of the sgRNAs are shown in SEQ ID No. 1; and the exogenous template comprises at least one nucleotide sequence shown in SEQ ID No. 2-3. The gene editing system according to the embodiment of the application has at least the following beneficial effects: the gene editing system provided by the application designs a guide RNA and a repair template aiming at a specific osteomyelitis mutation site, utilizes a CRISPR/Cas editing system to specifically cut a target sequence, and guides a homologous recombination repair process, thereby efficiently completing the repair of the R286W mutation site.
Description
Technical Field
The application relates to the technical field of induced pluripotent stem cells, in particular to a gene editing system for treating a osteomalacia and application thereof.
Background
Osteomalacia is a rare human genetic disease, and its diagnosis relies mainly on radiographic confirmation of abnormal bone density. As a heterogeneous disease, different patients can be roughly classified into asymptomatic or slightly symptomatic benign types and potentially severely life threatening malignant types. Because the etiology and the pathology of the malignant osteomyelitis are not clear at present, the treatment of the malignant osteomyelitis can only adopt a symptomatic treatment method. The currently available symptomatic treatment is allogeneic Hematopoietic Stem Cell Transplantation (HSCT) therapy, and approximately 73% of patients receiving treatment can achieve a disease-free survival period of 5 years. Although this treatment has been greatly developed over the past few years, implantation of stem cells from donors still presents a number of difficulties. Transplant rejection is one of the important problems, and it is almost impossible for two different individuals to have identical HLA, thus requiring the recipient to match the donor as closely as possible, which also greatly limits the available donors for the patient.
Aiming at the problem, the autologous stem cells can not generate any rejection or toxicity and risk to the body of the patient, and the safety of the autologous stem cells can be greatly improved. Induced pluripotent stem cells (ipscs) derived from patients are ideal autologous stem cells, but pathogenic genes are also present in the cells, and thus the cells need to be genetically repaired before they can be used. The CRISPR-Cas9 technology enables efficient editing at specific positions of the genome in various biological systems, and after the CRISPR system is cleaved to generate double-strand breaks, DNA repair inside cells follows the following two mechanisms: one is non-homologous end joining (NHEJ), and the broken DNA is directly connected; and the other is Homologous Directed Repair (HDR), and the broken DNA is subjected to homologous recombination repair according to a DNA template. Taking HDR as an example, the efficiency of HDR is affected by various factors, such as the chosen PAM site, the type of repair template, and the design of the template sequence, etc., which together result in a large difference in HDR efficiency among different cell lines. In earlier studies, it was identified that the mutation R286W in CLCN7 gene may be the cause of an autosomal dominant inheritance type 2 osteomalacia (ADO2) family, and ADO2-iPSC containing the mutation site was constructed. Therefore, it is necessary to provide a gene editing system capable of efficiently repairing the mutation site of the lithiasis by means of the CRISPR technology.
Disclosure of Invention
The present application is directed to solving at least one of the problems in the prior art. Therefore, the application provides a gene editing system aiming at the lithiasis and application thereof, and the gene repairing system can be used for efficiently repairing the mutation site of the lithiasis R286W.
In a first aspect of the present application, there is provided a gene editing system for a lithiasis, the gene editing system comprising:
the CRISPR/Cas editing system comprises sgRNAs, wherein the nucleotide sequences of the sgRNAs are shown in SEQ ID No. 1;
and the exogenous template comprises at least one nucleotide sequence shown in SEQ ID No. 2-3.
The gene editing system according to the embodiment of the application has at least the following beneficial effects:
the gene editing system provided by the application designs a guide RNA and a repair template aiming at a specific osteomyelitis mutation site, specifically cuts a target sequence by using a CRISPR/Cas editing system, and guides a homologous recombination repair process, thereby efficiently completing the repair of the R286W mutation site.
In some embodiments of the present application, the exogenous template is a double-stranded template comprising nucleotide sequences as set forth in SEQ ID Nos. 2-3, respectively.
Wherein, the nucleotide sequence shown in SEQ ID No.1 is as follows: CCGCAGAGACACAGAGAAGT, respectively;
the nucleotide sequence shown in SEQ ID No.2 is as follows:
ACTTCCGCAGAGACACAGAGAAGCGGGACTTCGTCTCCGCAGGGGCTGCGGC;
the nucleotide sequence shown in SEQ ID No.3 is as follows:
TGAAGGCGTCTCTGTGTCTCTTCGCCCTGAAGCAGAGGCGTCCCCGACGCCG。
in some embodiments of the present application, the exogenous template comprises upstream and downstream homology arm sequences and a repair sequence located between the upstream and downstream homology arm sequences, and the nucleotide sequence of the repair sequence is shown in at least one of SEQ ID Nos. 2-3. The homologous arm sequence refers to a sequence of an upstream fragment and a downstream fragment which are homologous with a region to be repaired where a target sequence is located, and when the target sequence is sheared by the CRISPR/Cas editing system, the homologous arm of the template is complementarily paired with the sheared double strands for repair.
In some embodiments of the present application, the gene editing system includes a targeting vector comprising a nucleotide sequence of the sgRNA and an exogenous template vector comprising a nucleotide sequence of the exogenous template.
The targeting vector is a vector for gene targeting, which can be recombined with a specific site of a recipient cell and repaired. Exogenous template vectors refer to vectors that are used to provide templates for gene editing repair. In some embodiments, the targeting vector and the exogenous template vector are plasmid vectors.
In some embodiments of the present application, the targeting vector is a Cas9 expression plasmid with the sgRNA integrated on the Cas9 expression plasmid.
In some embodiments of the present application, the targeting vector is a px461 plasmid and the exogenous template vector is a pUC57 plasmid.
In some embodiments of the present application, the targeting vector further comprises a nucleotide sequence of a Cas9 protein.
In a second aspect of the present application, there is provided a method for preparing a homologous recombination repair cell, the method comprising the steps of:
constructing a targeting vector and an exogenous template vector, wherein the targeting vector comprises a nucleotide sequence shown as SEQ ID No.1, and the exogenous template vector comprises at least one nucleotide sequence shown as SEQ ID No. 2-3;
and co-transfecting the targeting vector and the exogenous template vector to the cell to be repaired, and screening to obtain a gene editing and repairing cell, wherein the cell to be repaired has R286W mutation of the CLCN7 gene.
In some embodiments of the present application, the cell to be repaired is an induced pluripotent stem cell. By utilizing the preparation method, the induced pluripotent stem cells from the patient with the lithiasis with the R286W mutation can be restored to the wild normal induced pluripotent stem cells, so that the induced pluripotent stem cells can be applied to autologous stem cell treatment of the patient with the lithiasis, and the problems of rejection, toxicity and the like caused by xenotransplantation are avoided.
In some embodiments of the present application, the targeting vector further comprises a nucleotide sequence of a Cas9 protein.
In a third aspect of the present application, there is provided a homologous recombination repair cell, which is prepared by the aforementioned preparation method.
In some embodiments of the present application, the homologous recombination repair cell is an induced pluripotent stem cell. The homologous recombination repairing cell is an induced pluripotent stem cell derived from autologous cells of a patient, so that the homologous recombination repairing cell can be applied to stem cell treatment of the patient and effectively relieves the lithiasis of the patient.
In a fourth aspect of the present application, there is provided the use of the aforementioned homologous recombination repair cell in the preparation of a cell therapy product.
Additional aspects and advantages of the present application will be set forth in part in the description which follows and, in part, will be obvious from the description, or may be learned by practice of the present application.
Drawings
Fig. 1 is a schematic diagram of a gene editing method in example 1 of the present application, in which a is a schematic diagram of the principle of the method, b is a schematic diagram of the position of the mutation site, and c is a schematic diagram of the specific repair principle of the mutation site.
FIG. 2 is a map of the targeting vector plasmid and the foreign template vector plasmid constructed in example 1 of the present application.
FIG. 3 is a gel electrophoresis image of PCR amplification products of different clones in example 1 of the present application.
FIG. 4 shows the sequencing results of some of the clones in example 1 of the present application.
FIG. 5 shows the results of identifying the clones in example 1 of the present application, wherein A is an image of a GC-ADO2-iPSCs colony cultured on Matrigel, B is the result of karyotyping GC-ADO2-iPSCs, C and D are the results of flow cytometry for identifying the expression of GC-ADO2-iPSCs pluripotency markers SSEA-4 and Tra-1-81, E, F and G are the formation of GC-ADO2-iPSCs teratoma, E is endoderm (intestinal epithelial cells), F is mesoderm (cartilage), and G is ectoderm (neurocalyx).
Fig. 6 is western blot detection results of different ipscs before and after repair of the present application, wherein a is the imaging result of western blot analysis and B is the relative level of CLCN7 expression in the different ipscs.
Detailed Description
The conception and the resulting technical effects of the present application will be clearly and completely described below in conjunction with the embodiments to fully understand the objects, features and effects of the present application. Obviously, the described embodiments are only a part of the embodiments of the present application, and not all embodiments, and other embodiments obtained by a person skilled in the art without making any inventive effort based on the embodiments of the present application are within the protection scope of the present application.
The following detailed description of embodiments of the present application is provided for illustrative purposes only and is not intended to be limiting of the present application.
In the description of the present application, the meaning of a plurality is one or more, the meaning of a plurality is two or more, and the above, below, exceeding, etc. are understood as excluding the present number, and the above, below, within, etc. are understood as including the present number. If there is a description of first and second for the purpose of distinguishing technical features only, this is not to be understood as indicating or implying a relative importance or implicitly indicating the number of technical features indicated or implicitly indicating the precedence of technical features indicated.
In the description of the present application, reference to the description of the terms "one embodiment," "some embodiments," "an illustrative embodiment," "an example," "a specific example," or "some examples," etc., means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present application. In this specification, the schematic representations of the terms used above do not necessarily refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples.
Example 1
The embodiment provides a gene editing and repairing method of a osteodynia-induced pluripotent stem cell, which relates to an osteodynia-induced pluripotent stem cell (ADO2-iPSCs), and the preparation process comprises the following steps:
(1) a30 mL urine sample from a patient with urolithiasis who had been identified as the R286W mutation was taken and centrifuged at 300 Xg for 10 minutes at room temperature, the supernatant was discarded, the buffer was resuspended and transferred to a 50mL tube and centrifuged again at 300 Xg for 10 minutes at room temperature. The supernatant was discarded, the cells at the bottom of the tube were washed and resuspended in 0.5mL urea medium (Cellapy, Beijing, China), seeded onto culture plates, and cultured in 5% CO 2 And cultured at 37 ℃ with medium replacement every 60 hours. The cultured cells are inoculated to a 6-well plate, and reprogramming is carried out when the confluence degree reaches 50-80%.
(2) Reprogramming was performed by infecting cells with a non-integrative Sendai virus reprogramming kit (Cytotunne 2.0 Sendai vector, Thermo Scientific) containing 4 transcription factors (OCT4, SOX2, KLF4, and C-MYC). After 24 hours of incubation following transfection, cells were harvested, inoculated into fresh medium (day 1) and cultured for 6 days (medium was changed every 2 days). Cells were harvested on day 7 and plated on Matrigel-coated urea medium. The iPSCs were grown in a simple medium containing growth factors on day 8, and then replaced with fresh medium every day to observe the cloning of iPSCs. And (5) carrying out passage after the iPSCs clone grows to a proper size. In this example, ADO2-iPSCs cell line was established using clones selected on day 17 after plasmid infection. Through the combination of methods such as morphology, surface markers, pluripotency, karyotype and the like and sequencing, the ADO2-iPSCs with the R286W mutation are identified and then used for repairing the CRISPR/Cas gene editing system.
The gene editing method is shown in figure 1, wherein a is a schematic diagram of the principle of the method, b is a schematic diagram of the position of the mutation site, and ADO2-iPSCs which are derived from a patient and carry CLCN7 gene R286W mutation are subjected to repair of the mutation site in the ADO2-iPSCs through a CRISPR/Cas9 editing system by means of an HDR exogenous template to obtain iPSCs with normal CLCN7 gene expression homologous to the patient. c is a specific repair principle of the mutation site, the mutation site is caused by mutation of TGG on the 10 th exon into CGG, the Cas9 protein is used for cutting nearby the mutation site, and homologous directional repair is completed through an exogenous template (repair sequences are respectively shown as SEQ ID No. 2-3).
sgRNAs were designed according to the upstream and downstream sequences of TGG mutation on exon 10 via website http:// www.deephf.com/index/#/cas9, wherein scores are shown in Table 1 below according to the sgRNAs of the first five arranged from high to low, and sgRNA5 was selected as the sgRNA of the subsequent experiment. sgRNA5 was chosen because the sequence was closest to the mutation site.
TABLE 1 sgRNA obtained by design
The gene editing and repairing method of the homologous recombination and repair cell comprises the following steps:
(1) respectively constructing plasmids by the screened sgRNA5 and an exogenous template, wherein a sequence of the sgRNA5 is connected to a px461 plasmid to obtain a targeting vector plasmid for simultaneously expressing sgRNA and Cas9 proteins; the sequence of the foreign template was ligated to the pUC57 plasmid to give a foreign template vector plasmid expressing CLCN7 template. Referring to A and B of FIG. 2, maps of the constructed targeting vector plasmid and exogenous template vector plasmid are shown, respectively.
(2) And (3) electrically transferring the targeting vector plasmid and the exogenous template vector plasmid into the ADO2-iPSCs prepared above, carrying out drug screening after transfection, carrying out culture and amplification, selecting a monoclonal, carrying out PCR, and carrying out PCR product sequencing verification.
The iPSC clones edited by this method (total 11 clones) were collected, subjected to DNA extraction, PCR amplification, and gel electrophoresis, and the results are shown in fig. 3, with 9 clones (clones C13, C14, C16, C17, C18, C19, C20, C22, and C23) having a band 724bp in the size of the target fragment. The cells of these clones were collected, and partial results of sequencing verification are shown in fig. 4, and in combination with the figure, the mutant site TGG in the edited iPSC clone was successfully repaired to be the wild-type CGG.
The edited iPSCs were further identified, and the result is shown in fig. 5, wherein a is a morphological photograph, and it can be seen from the figure that clustered cells are visible on Matrigel, and have a clearer outline. B is the result of karyotype analysis, which shows that it has normal karyotype. C and D are expression results of identifying GC-ADO2-iPSCs pluripotency markers SSEA-4 and Tra-1-81 by flow cytometry respectively, and as can be seen in the figure, the cells can effectively express specific markers SSEA-4E and Tra-1-81 of pluripotent stem cells. E. F and G are the formation of GC-ADO2-iPSCs teratoma, and it can be seen from the figure that GC-ADO2-iPSC has the capacity of three germ layers to differentiate: endoderm (intestinal epithelial cells), mesoderm (cartilage) and ectoderm (neurorosettes). The results are combined, and the edited iPSC has the typical iPSC characteristics and is complete in chromosome.
The expression of CLCN7 in wild-type iPSCs (NC-iPSC), ADO2-iPSCs and GC-ADO2-iPSCs was analyzed by Western blotting, and beta-actin was used as a control, and as a result, as shown in FIG. 6, A is a gel image of Western blotting of different cells (2 cases of NC-iPSC, 3 cases of each of ADO2-iPSCs and GC-ADO 2-iPSCs), and B is the expression of CLCN7 in A relative to beta-actin, and it can be seen from the figure that the expression level of CLCN7 before and after repair did not change significantly. The results indicate that, at the iPSC stage, the expression level of the protein was not affected before and after CLCN7 gene repair.
By combining the method, the repair template plasmid is constructed, a target sequence is targeted through a specific sgRNA by virtue of a CRISPR/Cas9 technology, the repair template is utilized to carry out single-base mutation repair on a mutation site, so that corrected iPSCs based on ADO2 patients can be efficiently obtained, and the method can be further applied to treatment of the patients.
The present application has been described in detail with reference to the embodiments, but the present application is not limited to the embodiments described above, and various changes can be made within the knowledge of those skilled in the art without departing from the gist of the present application. Furthermore, the embodiments and features of the embodiments of the present application may be combined with each other without conflict.
SEQUENCE LISTING
<110> Shenzhen Lin research medicine Limited
<120> Gene editing System for osteomalacia and use thereof
<130> 1
<160> 3
<170> PatentIn version 3.5
<210> 1
<211> 20
<212> DNA
<213> Artificial sequence
<400> 1
ccgcagagac acagagaagt 20
<210> 2
<211> 52
<212> DNA
<213> Artificial sequence
<400> 2
acttccgcag agacacagag aagcgggact tcgtctccgc aggggctgcg gc 52
<210> 3
<211> 52
<212> DNA
<213> Artificial sequence
<400> 3
tgaaggcgtc tctgtgtctc ttcgccctga agcagaggcg tccccgacgc cg 52
Claims (10)
1. A gene editing system for a urolithiasis, comprising:
the CRISPR/Cas editing system comprises sgRNAs, and the nucleotide sequences of the sgRNAs are shown in SEQ ID No. 1;
the exogenous template comprises at least one nucleotide sequence shown in SEQ ID No. 2-3.
2. The gene editing system of claim 1, wherein the exogenous template is a double-stranded template comprising nucleotide sequences as set forth in SEQ ID nos. 2-3, respectively.
3. The gene editing system of claim 1, comprising a targeting vector comprising a nucleotide sequence of the sgRNA and an exogenous template vector comprising a nucleotide sequence of the exogenous template.
4. A gene editing system as claimed in claim 3, wherein the targeting vector is a px461 plasmid and the exogenous template vector is a pUC57 plasmid.
5. The gene editing system of claim 3, wherein the targeting vector further comprises a nucleotide sequence of a Cas9 protein.
6. The preparation method of the homologous recombination repairing cell is characterized by comprising the following steps:
constructing a targeting vector and an exogenous template vector, wherein the targeting vector comprises a nucleotide sequence shown as SEQ ID No.1, and the exogenous template vector comprises at least one nucleotide sequence shown as SEQ ID No. 2-3;
and co-transfecting the targeting vector and the exogenous template vector to a cell to be repaired, and screening to obtain a gene editing and repairing cell, wherein the cell to be repaired has R286W mutation of a CLCN7 gene.
7. The method according to claim 6, wherein the cells to be repaired are induced pluripotent stem cells.
8. The preparation method according to claim 6, wherein the targeting vector further comprises a nucleotide sequence of Cas9 protein.
9. A homologous recombination repair cell produced by the production method according to any one of claims 6 to 8.
10. Use of the homologous recombination repair cells of claim 9 for the preparation of a cell therapy product.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210455336.1A CN114934066B (en) | 2022-04-28 | 2022-04-28 | Gene editing system for stone bone disease and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210455336.1A CN114934066B (en) | 2022-04-28 | 2022-04-28 | Gene editing system for stone bone disease and application thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN114934066A true CN114934066A (en) | 2022-08-23 |
CN114934066B CN114934066B (en) | 2024-10-18 |
Family
ID=82862392
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202210455336.1A Active CN114934066B (en) | 2022-04-28 | 2022-04-28 | Gene editing system for stone bone disease and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN114934066B (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114369600A (en) * | 2022-01-28 | 2022-04-19 | 复旦大学附属眼耳鼻喉科医院 | For repairing Klhl18lowfCRISPR/Cas9 gene editing system of mutant gene and application |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN110418800A (en) * | 2017-03-15 | 2019-11-05 | 加利福尼亚大学董事会 | The method for treating lysosome disorder |
CN111235152A (en) * | 2020-03-31 | 2020-06-05 | 深圳市人民医院 | sgRNA specifically targeting CLCN7 and application thereof |
-
2022
- 2022-04-28 CN CN202210455336.1A patent/CN114934066B/en active Active
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN110418800A (en) * | 2017-03-15 | 2019-11-05 | 加利福尼亚大学董事会 | The method for treating lysosome disorder |
CN111235152A (en) * | 2020-03-31 | 2020-06-05 | 深圳市人民医院 | sgRNA specifically targeting CLCN7 and application thereof |
Non-Patent Citations (3)
Title |
---|
LI DANDAN等: "CRISPR/Cas9-Mediated Gene Correction in Osteopetrosis Patient-Derived iPSCs", FRONT. BIOSCI. (LANDMARK ED), vol. 28, no. 6, 30 June 2023 (2023-06-30), pages 1 - 9 * |
OU MINGLIN等: "Genotyping, generation and proteomic profiling of the first human autosomal dominant osteopetrosis type II-specific induced pluripotent stem cells", STEM CELL RESEARCH & THERAPY, vol. 10, pages 1 - 17 * |
孙国平等: "基于CRISPR/Cas9 技术构建人CLCN7 基因编辑载体", 实验与检验医学, vol. 38, no. 3, pages 470 - 472 * |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114369600A (en) * | 2022-01-28 | 2022-04-19 | 复旦大学附属眼耳鼻喉科医院 | For repairing Klhl18lowfCRISPR/Cas9 gene editing system of mutant gene and application |
CN114369600B (en) * | 2022-01-28 | 2024-02-13 | 复旦大学附属眼耳鼻喉科医院 | For repairing Klhl18 lowf CRISPR/Cas9 gene editing system of mutant gene and application |
Also Published As
Publication number | Publication date |
---|---|
CN114934066B (en) | 2024-10-18 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Shao et al. | Generation of iPS cells using defined factors linked via the self-cleaving 2A sequences in a single open reading frame | |
US20190264223A1 (en) | Novel method | |
EP2658965B1 (en) | Method of generating induced pluripotent stem cells and differentiated cells | |
KR20130101135A (en) | Method for preparing induced pluripotent stem cells and culture medium used for preparing induced pluripotent stem cells | |
US20230053028A1 (en) | Engineered cells for therapy | |
CN112204149A (en) | Reprogramming carrier | |
CN113881637A (en) | Human induced pluripotent stem cell and preparation method and application thereof | |
He et al. | Genetic correction and hepatic differentiation of hemophilia B-specific human induced pluripotent stem cells | |
CN114934066B (en) | Gene editing system for stone bone disease and application thereof | |
EP2481795A1 (en) | Method of generating induced pluripotent stem cells and differentiated cells | |
WO2024183423A1 (en) | Crispr/cas9-grna targeting plasmid, donor plasmid, and method for preparing immortalized mouse cell line | |
CN101492676B (en) | Method for producing derivational pluripotent stem cell with meninges cell and uses thereof | |
CN110499331A (en) | The preparation method of exogenous human telomerase reverse transcriptase genetic recombination lentiviral particle | |
Shi et al. | Efficient and rapid fluorescent protein knock-in with universal donors in mouse embryonic stem cells | |
WO2016052759A1 (en) | Method for reestablishing pluripotent stem cells | |
El-Sayed et al. | Pluripotent state induction in mouse embryonic fibroblast using mRNAs of reprogramming factors | |
CN113195724A (en) | Low-immunogenicity engineered human mesenchymal stromal cells, preparation method and kit | |
US20220364072A1 (en) | Fusion protein that improves gene editing efficiency and application thereof | |
CN116334000A (en) | Preparation method of low-immunogenicity IPSC (iPSC) cells and low-immunogenicity iPSC cells | |
CN106987562A (en) | A kind of preparation method of immortalized mouse candidate stem cell system | |
WO2012165740A1 (en) | Composition for improving dedifferentiation of cells and method for producing inducted pluripotent stem cells using the same | |
CN114540310B (en) | Mouse embryonic stem cell and preparation method and application thereof | |
CN113564203B (en) | Preparation method and application of HSV1-tk/GCV induced blood system defect mouse model | |
JP7369416B2 (en) | Methods for producing naive pluripotent stem cells, methods for naiveting stem cells, and methods for preserving them | |
CN115074388A (en) | Maternal factor induced 2C sample totipotent stem cell and transformation application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant |