WO1990014092A1 - Single-strand site-directed modification of mammalian genes in vivo - Google Patents

Single-strand site-directed modification of mammalian genes in vivo Download PDF

Info

Publication number
WO1990014092A1
WO1990014092A1 PCT/US1990/002773 US9002773W WO9014092A1 WO 1990014092 A1 WO1990014092 A1 WO 1990014092A1 US 9002773 W US9002773 W US 9002773W WO 9014092 A1 WO9014092 A1 WO 9014092A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
target site
site
cells
cell
Prior art date
Application number
PCT/US1990/002773
Other languages
French (fr)
Inventor
Raju S. Kucherlapati
Colin Campbell
Original Assignee
Cell Genesys, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cell Genesys, Inc. filed Critical Cell Genesys, Inc.
Publication of WO1990014092A1 publication Critical patent/WO1990014092A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • 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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)

Definitions

  • the field of this invention is site-directed modification of genes in mammalian cells.
  • Site-directed modification is achieved by employing single-stranded oligonucleotide fragments having at least 40 nucleotides of homology at the site of interest, where the sequence desirably has an internal marker for detecting the presence of the desired modification.
  • DESCRIPTION OF THE SPECIFIC EMBODIMENTS Methods and composition are provided for transformation with single-stranded oligonucleotides having at least 40 base homology with a target site. The transformation may be achieved under conventional transformation conditions in culture. After selection and amplification of the transformed cells, the cells may be screened. Desirably a marker is employed as part of the oligonucleotide to allow for determination of the copy number and the site of insertion of the oligonucleotide in the mammalian cell genome.
  • the oligonucleotides will be at least about 40 bases and not more than about 2 kb, usually not more than about 1 kb.
  • the sequences will have substantial homology with the site of insertion, but may differ by one or more bases, usually not more than about 10, more usually not more than 5 bases from the target sequence.
  • sequences will be selected to modify a particular sequence, normally to change the phenotype of the cell.
  • sequences may provide substi ⁇ tutions, both transitions and transversions, insertions and deletions, in order to change the sequence present in the host.
  • Sequences of interest will frequently be associated with mutations causing diseases. These sequences may be involved with the globin genes, in sickle-cell anemia, and ⁇ -thalassemia, with adenosine deaminase gene in severe combined immunodeficiency, etc.
  • the situations where genetic modification will be desirable include sickle cell anemia and thalassemias, as well as other genetic diseases. Therefore, in the subject invention, normally the sequence of interest of the properly functioning gene, as well as the mutation will be known. However, in many instances it may not be necessary to know the specific defect, so long as one knows the region of the defect, and the sequences flanking the defect.
  • the oligonucleotide may be designed to provide the desired amino acid sequence, while also providing for a restriction site which is not naturally present in the wild-type gene, nor in the defective gene. In this manner, transformed cells may be screened to identify the presence of fragments having homology to the oligonucleotide, where these fragments may be cleaved at the restriction site. One could then identify that the oligonucleotide had been inserted at that site.
  • flanking regions had the proper sequence
  • the oligonucleotide may have homology to a regulatory region, intron or other sequence which can affect the nature and amount of an expressed product. In this way one can modify splicing sites, inactivate or activate enhancers, promoters, inducible regulatory regions, etc.
  • the subject method may be used with any mammalian cells of interest, including primates, particularly humans, domestic animals, e.g. bovine, equine, feline, canine, etc.
  • the cells will normally be transformed in culture, usually as dispersed cells, although in some instances, tissue slices or chunks may be involved, particularly where one is not concerned with having all cells of the desired phenotype, but only having a sufficient number of cells having the desired pheno ⁇ type. About 0.1 - 100 ⁇ g of DNA/10 6 cells will usually be employed. Organs which may be involved or cells from such organs include blood, bone marrow, lymph node, skin, endothelium, muscle, brain, central nervous system, thymus, liver, kidney, pancreas, etc. Specific cells may be B-cells, T-cells, neurons, glial cells, macrophages, monocytes, stem cells, retinal pigment epithelial cells, etc.
  • the cells will normally be present in an appropriate medium, for example, DMEM supplemented with appropriate growth factors, conveniently components of fetal serum.
  • DMEM fetal serum
  • the cells may be transformed by any convenient technique, such as calcium phosphate DNA coprecipitates, electroporation, liposome endocytosis, microinjection, etc. The particular manner of transformation is not critical to this invention.
  • a temperature sensitive or inducible replication system may be used, where the plasmid may be readily cured from the host cells by maintaining the host cells at a non-permissive temperature or in the absence of the inducer.
  • various markers may be introduced in conjunction with the oligonucleotide sequence, so as to select for those cells which have been transformed. It is found that cells which accept DNA are likely to accept all forms of DNA present. There is thus a high probability that if the plasmid has been accepted, the oligonucleotide will also be present in the same cells.
  • Various markers may be present on the plasmid, particularly antibiotic resistance, e.g. G418 resistance.
  • Various replication systems may be used, such as adenovins, papilloma virus, simian virus, Epstein-Barr virus, etc.
  • these cells may be amplified and returned to the host as appropriate. Amplification can be achieved by growth in an appropriate culture medium in the presence of the proper growth factors. These cells may then be stored before administration to the host in an appropriate manner, depending upon the nature of the cells.
  • the cells may be introduced into the circulatory system by injection to provide for normal cells, hematopoietic cells may be transfused back to the patient; etc.
  • grafts may be involved, where tissue may be grafted onto existing tissue in the patient.
  • oligonucleotide is compared with the neomycin sequence with the Cla I insert.
  • the underlined nucleotide indicates the substitution.
  • the complementary double-stranded DNA substrate was pSV2neoIL, which was derived by insertion of a 14 bp Cla I linker into the coding region of a neomycin phosphotransferase gene. Insertion renders the neo gene inactive.
  • the oligonucleotide was a synthetic oligodeoxynucleotide of 40 nucleotides. It contained the wild-type sequence at the region corresponding to the insertion. In addition, it contained a single base change which is silent but creates a Taq I restriction endonuclease recognition site.
  • FBS fstal bovine serum
  • Each dish received 10 yg of plasmid DNA with or without an equal mass of oligonu ⁇ cleotide.
  • the precipitate was removed and the cells treated with 20% (v/v) dimethyl sulfoxide for 2 min, rinsed and placed immediately in DMEM with 10% FBS. The next day, the cells were transferred to 100 mm dishes in DMEM containing 10% FBS and 400 ⁇ g/ml G418. Colonies were counted after 14 days.
  • a refers only to the amount of plasmid DNA added. When included, an equal mass of oligonucleotide was present. By heterologous oligonucleotide is intended sequence heterologous to the neo gene. pSV2neoIL (Cla I) indicates the presence of the 14 bp linker in the neo gene.
  • Genomic DNA was purified from G418 resistant cell lines obtained from transfection of EJ cells with pSV2neoIL and the oligonucleotide. 1 ⁇ g of DNA was used in an enzymatic amplification procedure to amplify an 800 bp fragment spanning the region of interest.
  • the polymerase chain reaction was performed with a Perkin-Elmer Cetus DNA amplification kit. Thirty cycles of 94°, 1 min, and 65°, 5 min, were performed using an automated thermal cycler.
  • the amplified fragment (800 bp) along with the rest of the reaction mixture was electrophoresed on a 1% agarose gel.
  • the DNA was subsequently transferred to nitrocellulose and hybridized as described by Wood, et al. ((1985) Proc. Natl. Acad. Sci. (USA) 82:1585-8) under conditions which allow discrimination on the basis of a single mismatch with the probe.
  • the amplified products from 2 of the 4 cell lines examined hybridized to the oligonucleotide indicating that in these cell lines, the oligonucleotide participated in the recombination reaction.
  • Plasmid DNA was rescued by fusing the G418 R cell lines to monkey COS cells followed by isolation of low mol cular weight DNA. Digestion of pSV2neoIL with Cla I linearizes it, producing a 5.7 kb molecule. Neither wild-type (WT) pSV2neo nor the rescued plasmid are Cla I sensitive and they migrate as a mixture of closed and nicked circles. Wild-type pSV2neo digested with Taq I yields 3 major bands, 1 of 2.1 kb, and 2 of 1.4 kb. The appropriate nucleotide substitution encoded by the oligonucleotide results in the change of a 1.4 kb band to a 1.2 kb band.
  • the rescued DNA was used to transform recA ⁇ E. coli.
  • the DNA contained an additional Taq I site and also contained the silent mutation as determined by DNA sequencing.
  • 14 out of 50 or 28% of plasmids derived from colonies in which the pSV2neoIL was used in conjunction with the oligo ⁇ nucleotide contained the Taq I site. Since not all of the information present on the oligonucleotide is necessary for the correction of the mutant plasmid, it is possible that a larger proportion of the plasmids are the result of homologous recombination.
  • a single-stranded DNA may provide higher efficiency of transformation at homologous sites in mammalian cells as compared to non-homologous sites than double-stranded DNA.
  • single-stranded DNA may find preferred usage in site-directed modification.

Abstract

Single-stranded oligonucleotides are employed for site-directed modification in mammalian cells to change genes encoding proteins of interest. Desirably, a marker may be included with the gene to be able to detect the insertion site and transformation may be performed in conjunction with a plasmid having a marker, where the plasmid may be cured from the host.

Description

SINGLE-STRAND SITE-DIRECTED MODIFICATION OF MAMMALIAN GENES IN VIVO
This work was supported by grants from the National Institute of Health (GM33943 and GM36565, as well as GM11893-02).
INTRODUCTION
Technical Field The field of this invention is site-directed modification of genes in mammalian cells.
Background
There is a great need for genetically modified mammalian cells, so as to introduce a new phenotype, correct a mutated phenotype, or inhibit a particular gene expression. There are a large number of genetic diseases, where the mutation has been established, and the list should increase as further diseases and their etiology are investigated and determined. In many situations, particularly the hematopoietic system, there are opportunities to genetically modify cells and reintroduce the cells into the mammalian host, where the genetically modified cells may function and correct a genetic or physiological defect. Diseases such as α- and β-thalassemia, sickle-cell anemia, are immediately evident opportunities for genetic treatment.
In modifying cells, there are substantial concerns. Normally, the number of cells available may be fairly limited. Secondly, it is important that genetic modification occur at the target site and not at other sites. Unless the modification is specific, the insertion of the introduced DNA at other sites may result in cellular modifications which could be detrimental. It is therefore important in providing for transformation of a cellular host, that the cells be transformed with high efficiency, that methods be provided which allow for identification and isolation of the modified cells, with some certainty of there being a single insertion at the desired target site. It is, therefore, of substantial interest to provide techniques and reagents which allow for enhanced efficiency in site-directed gene modification.
Relevant Literature Site-directed gene insertion in mammalian chromosomes is described by Smithies, et al. (1985) Nature 317:230-4; Thomas, et al. (1986) Cell 44:419-28; Thomas and Capecchi (1987) Cell 5_1:503-12; Doetchman, et al. (1987) Nature 330:576-8; and Mansour, et al. (1988) Nature 3^6:348-352. The participation of single-stranded DNA in homologous recombination in mammalian cells has been reported by Rauth, e_t al. (1986) 3:5587-91, with as few as 25 bp of DNA sequence homology adequate for recombination (Rubnitz and Subranani (1984) Mol. Cell. Biol. :2253-58); Ayares, et al. (1986) Proc. Natl. Acad. Sci. (USA) 8^:5199- 5203. Moerschell, et al. (1988) Proc. Natl. Acad. Sci. (USA) 8_5_:524-8 describes the modification of yeast genes using single-stranded synthetic oligonucleotides.
SUMMARY OF THE INVENTION
Site-directed modification is achieved by employing single-stranded oligonucleotide fragments having at least 40 nucleotides of homology at the site of interest, where the sequence desirably has an internal marker for detecting the presence of the desired modification. DESCRIPTION OF THE SPECIFIC EMBODIMENTS Methods and composition are provided for transformation with single-stranded oligonucleotides having at least 40 base homology with a target site. The transformation may be achieved under conventional transformation conditions in culture. After selection and amplification of the transformed cells, the cells may be screened. Desirably a marker is employed as part of the oligonucleotide to allow for determination of the copy number and the site of insertion of the oligonucleotide in the mammalian cell genome.
The oligonucleotides will be at least about 40 bases and not more than about 2 kb, usually not more than about 1 kb. The sequences will have substantial homology with the site of insertion, but may differ by one or more bases, usually not more than about 10, more usually not more than 5 bases from the target sequence.
The sequences will be selected to modify a particular sequence, normally to change the phenotype of the cell. Thus, the sequences may provide substi¬ tutions, both transitions and transversions, insertions and deletions, in order to change the sequence present in the host.
Sequences of interest will frequently be associated with mutations causing diseases. These sequences may be involved with the globin genes, in sickle-cell anemia, and β-thalassemia, with adenosine deaminase gene in severe combined immunodeficiency, etc. The situations where genetic modification will be desirable include sickle cell anemia and thalassemias, as well as other genetic diseases. Therefore, in the subject invention, normally the sequence of interest of the properly functioning gene, as well as the mutation will be known. However, in many instances it may not be necessary to know the specific defect, so long as one knows the region of the defect, and the sequences flanking the defect. As already indicated, it will normally not be necessary to have complete homoduplexing in the regions where the amino acid sequence is to be conserved and in some instances it will be desirable to have one or a few mismatches. Since it will be desirable to be able to ascertain how many copies of the oligonucleotide became inserted and whether insertion was at the target site, the oligonucleotide may be designed to provide the desired amino acid sequence, while also providing for a restriction site which is not naturally present in the wild-type gene, nor in the defective gene. In this manner, transformed cells may be screened to identify the presence of fragments having homology to the oligonucleotide, where these fragments may be cleaved at the restriction site. One could then identify that the oligonucleotide had been inserted at that site.
By further showing that flanking regions had the proper sequence, one could establish whether the oligonucleotide was inserted at the appropriate site or at a different site. Thus, one could rapidly determine by employing gel electrophoresis, Southern hybridi¬ zation, or other screening technique, whether one had cells in which the proper modification had occurred. Particularly, one can employ a polymerase chain reaction, using primers to sequences which would flank the oligonucleotide. In this way, a relatively large amount of DNA could be obtained, which could be sequenced or hybridized to determine whether the desired modification had occurred.
Rather than have homology to a structural gene, the oligonucleotide may have homology to a regulatory region, intron or other sequence which can affect the nature and amount of an expressed product. In this way one can modify splicing sites, inactivate or activate enhancers, promoters, inducible regulatory regions, etc. The subject method may be used with any mammalian cells of interest, including primates, particularly humans, domestic animals, e.g. bovine, equine, feline, canine, etc. The cells will normally be transformed in culture, usually as dispersed cells, although in some instances, tissue slices or chunks may be involved, particularly where one is not concerned with having all cells of the desired phenotype, but only having a sufficient number of cells having the desired pheno¬ type. About 0.1 - 100 μg of DNA/106 cells will usually be employed. Organs which may be involved or cells from such organs include blood, bone marrow, lymph node, skin, endothelium, muscle, brain, central nervous system, thymus, liver, kidney, pancreas, etc. Specific cells may be B-cells, T-cells, neurons, glial cells, macrophages, monocytes, stem cells, retinal pigment epithelial cells, etc.
The cells will normally be present in an appropriate medium, for example, DMEM supplemented with appropriate growth factors, conveniently components of fetal serum. The cells may be transformed by any convenient technique, such as calcium phosphate DNA coprecipitates, electroporation, liposome endocytosis, microinjection, etc. The particular manner of transformation is not critical to this invention.
In some instances, it may be desirable to cotransfect with a plasmid which may be cured from the host. A temperature sensitive or inducible replication system may be used, where the plasmid may be readily cured from the host cells by maintaining the host cells at a non-permissive temperature or in the absence of the inducer. In this manner, various markers may be introduced in conjunction with the oligonucleotide sequence, so as to select for those cells which have been transformed. It is found that cells which accept DNA are likely to accept all forms of DNA present. There is thus a high probability that if the plasmid has been accepted, the oligonucleotide will also be present in the same cells. Various markers may be present on the plasmid, particularly antibiotic resistance, e.g. G418 resistance. Various replication systems may be used, such as adenovins, papilloma virus, simian virus, Epstein-Barr virus, etc.
Once the cells have been identified as having the proper modification, these cells may be amplified and returned to the host as appropriate. Amplification can be achieved by growth in an appropriate culture medium in the presence of the proper growth factors. These cells may then be stored before administration to the host in an appropriate manner, depending upon the nature of the cells. For bone marrow, the cells may be introduced into the circulatory system by injection to provide for normal cells, hematopoietic cells may be transfused back to the patient; etc. In some instances, grafts may be involved, where tissue may be grafted onto existing tissue in the patient.
The following examples are offered by way of illustration and not by way of limitation.
EXPERIMENTAL An oligonucleotide sequence was prepared having the following sequence:
GATC C C TAGG
GC GC -GATGGATTGCACGCAGGTTCTCCGGCCGCTTGGGTGGAGAGGCTAT- Neo IL GGATTGCACGCAGGTTCTCCGGCCGCTTGGGTCGAGAGGC Oligo The oligonucleotide is compared with the neomycin sequence with the Cla I insert. The underlined nucleotide indicates the substitution. The complementary double-stranded DNA substrate was pSV2neoIL, which was derived by insertion of a 14 bp Cla I linker into the coding region of a neomycin phosphotransferase gene. Insertion renders the neo gene inactive. The oligonucleotide was a synthetic oligodeoxynucleotide of 40 nucleotides. It contained the wild-type sequence at the region corresponding to the insertion. In addition, it contained a single base change which is silent but creates a Taq I restriction endonuclease recognition site. For each transfection 1 X 10° human EJ cells were plated in 60 mm dishes. The cells were grown in DMEM supplemented with 10% fstal bovine serum (FBS). One to 2 days later, calcium phosphate DNA coprecipi- tates (Lowy, e_t al. (1978) J. Virol. 6.:291~8) were added to each plate. Each dish received 10 yg of plasmid DNA with or without an equal mass of oligonu¬ cleotide. Four hours later, the precipitate was removed and the cells treated with 20% (v/v) dimethyl sulfoxide for 2 min, rinsed and placed immediately in DMEM with 10% FBS. The next day, the cells were transferred to 100 mm dishes in DMEM containing 10% FBS and 400 μg/ml G418. Colonies were counted after 14 days.
The following table indicates the results. TABLE 1
No. of Plasmid DNA G418R Colonies
Subtrates Expts useda (i9) Total per μg pSV2neoIL + oligo 11 326 17 0.052 pSV2neoIL 3 400 1 0.0025 pSV2neoIL + heterologous oligonucleotide 2 320 0 <0.0031 pSV2neoIL (Cla I) + oligonucleotide 4 64 121 1.89 pSV2neoIL (Cla I) 4 40 22 0.55
a Refers only to the amount of plasmid DNA added. When included, an equal mass of oligonucleotide was present. By heterologous oligonucleotide is intended sequence heterologous to the neo gene. pSV2neoIL (Cla I) indicates the presence of the 14 bp linker in the neo gene.
Genomic DNA was purified from G418 resistant cell lines obtained from transfection of EJ cells with pSV2neoIL and the oligonucleotide. 1 μg of DNA was used in an enzymatic amplification procedure to amplify an 800 bp fragment spanning the region of interest. The polymerase chain reaction was performed with a Perkin-Elmer Cetus DNA amplification kit. Thirty cycles of 94°, 1 min, and 65°, 5 min, were performed using an automated thermal cycler. The amplified fragment (800 bp) along with the rest of the reaction mixture was electrophoresed on a 1% agarose gel. The DNA was subsequently transferred to nitrocellulose and hybridized as described by Wood, et al. ((1985) Proc. Natl. Acad. Sci. (USA) 82:1585-8) under conditions which allow discrimination on the basis of a single mismatch with the probe.
The amplified products from 2 of the 4 cell lines examined hybridized to the oligonucleotide indicating that in these cell lines, the oligonucleotide participated in the recombination reaction.
Plasmid DNA was rescued by fusing the G418R cell lines to monkey COS cells followed by isolation of low mol cular weight DNA. Digestion of pSV2neoIL with Cla I linearizes it, producing a 5.7 kb molecule. Neither wild-type (WT) pSV2neo nor the rescued plasmid are Cla I sensitive and they migrate as a mixture of closed and nicked circles. Wild-type pSV2neo digested with Taq I yields 3 major bands, 1 of 2.1 kb, and 2 of 1.4 kb. The appropriate nucleotide substitution encoded by the oligonucleotide results in the change of a 1.4 kb band to a 1.2 kb band.
The rescued DNA was used to transform recA~ E. coli. The DNA contained an additional Taq I site and also contained the silent mutation as determined by DNA sequencing. Analysis of 36 plasmids recovered from different G418R cell lines derived from transfection of pSV2neoIL alone failed to reveal any molecules which contained the Taq I site. In contrast 14 out of 50 or 28% of plasmids derived from colonies in which the pSV2neoIL was used in conjunction with the oligo¬ nucleotide contained the Taq I site. Since not all of the information present on the oligonucleotide is necessary for the correction of the mutant plasmid, it is possible that a larger proportion of the plasmids are the result of homologous recombination.
Based on these results, a single-stranded DNA may provide higher efficiency of transformation at homologous sites in mammalian cells as compared to non-homologous sites than double-stranded DNA. Thus, single-stranded DNA may find preferred usage in site-directed modification.
All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method for obtaining site-directed modification iτ\ vivo at a genomic target site to change the phenotype of a cell, the modification at the target site occurring with high efficiency, said method comprising: combining under transforming conditions a single stranded oligodeoxynucleotide with mammalian cells, wherein said oligonucleotide is substantially homologous to a target site, but differs by at least one nucleotide; and selecting for cells comprising said oligonucleotide sequence at said target site.
2. A method according to Claim 1, wherein said transforming conditions comprise calcium phosphate precipitated DNA.
3. A method according to Claim 1, wherein said oligonucleotide sequence comprises a restriction site absent at said target site.
4. A method according to Claim 1, wherein said oligonucleotide has at least 40nt of homology with said target site.
5. A method according to Claim 4, wherein said oligonucleotide is from about 40nt to 2knt.
6. A method according to Claim 1, wherein said mammalian cell is an hematopoietic cell.
7. A method according to Claim 6, wherein said hematopoietic cell is a lymphocyte.
8. A method according to Claim 1, wherein said mammalian cell is a retinal pigment epithelial cell.
9. A method for obtaining site-directed modification iji vivo at a genomic target site to change the phenotype of a cell, the modification at the target site occurring with high efficiency, said method comprising: combining under transforming conditions a single stranded oligodeoxynucleotide of at least 40nt with mammalian cells, wherein said oligonucleotide is substantially homologous to a target site, but differs by at least one nucleotide and comprises a restriction site absent at said target site; and screening or selecting for cells comprising said oligonucleotide sequence at said target site by fragmenting the genome of said host cell, identifying fragments hybridizing with said oligonucleotide and identifying fragments having the target site sequence flanking said oligonucleotide sequence by means of said restriction site.
10. A method according to Claim 9, wherein said transforming conditions comprise calcium phosphate precipitated DNA.
11. A method according to Claim 9, wherein said oligonucleotide has at least about 40nt of homology with said target site.
PCT/US1990/002773 1989-05-18 1990-05-18 Single-strand site-directed modification of mammalian genes in vivo WO1990014092A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US35390989A 1989-05-18 1989-05-18
US353,909 1989-05-18

Publications (1)

Publication Number Publication Date
WO1990014092A1 true WO1990014092A1 (en) 1990-11-29

Family

ID=23391104

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/002773 WO1990014092A1 (en) 1989-05-18 1990-05-18 Single-strand site-directed modification of mammalian genes in vivo

Country Status (1)

Country Link
WO (1) WO1990014092A1 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0535144A1 (en) * 1990-06-12 1993-04-07 Baylor College Of Medicine Method for homologous recombination in animal and plant cells
US5272071A (en) * 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
EP0656747A1 (en) * 1992-08-21 1995-06-14 The Regents Of The University Of California Composition and method for altering dna sequences by homologous recombination
EP0733059A1 (en) * 1993-12-09 1996-09-25 Thomas Jefferson University Compounds and methods for site-directed mutations in eukaryotic cells
EP0747485A1 (en) * 1989-11-06 1996-12-11 Cell Genesys, Inc. Production of proteins using homologous recombination
US5686263A (en) * 1991-05-31 1997-11-11 Genentech, Inc. Method for enhancing gene expression
US6001968A (en) * 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6048837A (en) * 1994-08-17 2000-04-11 The Rockefeller University OB polypeptides as modulators of body weight
US6124448A (en) * 1994-08-17 2000-09-26 The Rockfeller University Nucleic acid primers and probes for the mammalian OB gene
US6124439A (en) * 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US6172211B1 (en) 1997-07-11 2001-01-09 Boehringer Ingelheim International Gmbh Nucleic acid encoding tag7 polypeptide
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
WO2002010364A2 (en) * 2000-07-27 2002-02-07 University Of Delaware Methods for enhancing targeted gene alteration using oligonucleotides
US6350730B1 (en) 1994-08-17 2002-02-26 The Rockefeller University OB polypeptides and modified forms as modulators of body weight
US6471956B1 (en) 1994-08-17 2002-10-29 The Rockefeller University Ob polypeptides, modified forms and compositions thereto
US6524818B1 (en) 1997-09-26 2003-02-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
WO2004015117A2 (en) * 2002-08-13 2004-02-19 Nederlands Kanker Instituut Targeted gene modification by single-stranded dna oligonucleotides
US6740503B1 (en) 1997-09-26 2004-05-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
EP1421187A2 (en) * 2001-07-27 2004-05-26 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Systems for in vivo site-directed mutagenesis using oligonucleotides
US6773911B1 (en) 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
US7063958B1 (en) 1996-01-16 2006-06-20 The Rockefeller University Nucleic acids db, the receptor for leptin
US7084252B1 (en) 1996-01-16 2006-08-01 The Rockefeller University DB, the receptor for leptin
US7148004B1 (en) 1997-01-16 2006-12-12 The Rockefeller University Oligonucleotides of the OB-R isoforms and methods of diagnosing body weight
US7205398B2 (en) 2002-05-24 2007-04-17 Schering-Plough Animal Health Corporation Eta-1 gene and methods for use
US7619079B2 (en) 1996-02-14 2009-11-17 The Rockefeller University Db, the receptor for leptin, nucleic acids encoding the receptor, and uses thereof
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
US8110185B2 (en) 2008-10-03 2012-02-07 St. Michael's Hospital Method for preventing and treating cardiovascular diseases with BRCA1
US10584363B2 (en) 2016-06-03 2020-03-10 Takara Bio Usa, Inc. Methods of producing and using single-stranded deoxyribonucleic acids and compositions for use in practicing the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4457918A (en) * 1982-05-12 1984-07-03 The General Hospital Corporation Glycosides of vitamins A, E and K
US4565863A (en) * 1983-12-08 1986-01-21 Hoffmann-La Roche Inc. Retinoid carbohydrates

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4457918A (en) * 1982-05-12 1984-07-03 The General Hospital Corporation Glycosides of vitamins A, E and K
US4565863A (en) * 1983-12-08 1986-01-21 Hoffmann-La Roche Inc. Retinoid carbohydrates

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Volume 73, Number 14, issued 05 October 1970, (Columbus, Ohio, USA), N. TAKABAYOSHI et al.; "Vitamin A Glucose Ether", see pages 228, column 1, the abstract number 698342; & JP,B,45 020 097, 09 July 1970. *
JOURNAL OF CELLULAR BIOCHEMISTRY, (New York, USA), Volume Supplement O, issue 13 part E, issued 03 April 1989, CAMPBELL et al.: "Homologous recombination involving single-stranded oligonucleotide in human cells", pages 277, abstract number WH113, see the entire abstract. *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCE, Volume 84, issued April 1987 (USA), M/H. FILE et al., "Introduction of Differentication of Human Promyelocytic Leukemia Cell Line HL-60 by Retinoyl Glucuronide, a Biological activity metabolite of Vitamin A", see pages 2208-2212. *
THE AMERICAN JOURNAL OF CLINICAL NUTRITION, Volume 43, issued April 1986 (USA), A.B. BARAA et al.; "Refinoyl B-glucuronice: an endogenous compound of human blood", see pages 481-485. *

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0747485A1 (en) * 1989-11-06 1996-12-11 Cell Genesys, Inc. Production of proteins using homologous recombination
US5981214A (en) * 1989-11-06 1999-11-09 Cell Genesys, Inc. Production of proteins using homologous recombination
US5272071A (en) * 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
EP0535144A1 (en) * 1990-06-12 1993-04-07 Baylor College Of Medicine Method for homologous recombination in animal and plant cells
EP0535144A4 (en) * 1990-06-12 1993-08-11 Baylor College Of Medicine Method for homologous recombination in animal and plant cells
US5686263A (en) * 1991-05-31 1997-11-11 Genentech, Inc. Method for enhancing gene expression
EP0656747A1 (en) * 1992-08-21 1995-06-14 The Regents Of The University Of California Composition and method for altering dna sequences by homologous recombination
EP0656747A4 (en) * 1992-08-21 1997-05-07 Univ California Composition and method for altering dna sequences by homologous recombination.
EP0733059A1 (en) * 1993-12-09 1996-09-25 Thomas Jefferson University Compounds and methods for site-directed mutations in eukaryotic cells
EP0733059A4 (en) * 1993-12-09 1997-05-21 Univ Jefferson Compounds and methods for site-directed mutations in eukaryotic cells
US6124448A (en) * 1994-08-17 2000-09-26 The Rockfeller University Nucleic acid primers and probes for the mammalian OB gene
US6001968A (en) * 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6124439A (en) * 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
EP2402444A1 (en) 1994-08-17 2012-01-04 The Rockefeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US6350730B1 (en) 1994-08-17 2002-02-26 The Rockefeller University OB polypeptides and modified forms as modulators of body weight
US6471956B1 (en) 1994-08-17 2002-10-29 The Rockefeller University Ob polypeptides, modified forms and compositions thereto
US6048837A (en) * 1994-08-17 2000-04-11 The Rockefeller University OB polypeptides as modulators of body weight
US7063958B1 (en) 1996-01-16 2006-06-20 The Rockefeller University Nucleic acids db, the receptor for leptin
US7084252B1 (en) 1996-01-16 2006-08-01 The Rockefeller University DB, the receptor for leptin
US7812137B2 (en) 1996-01-16 2010-10-12 The Rockefeller University Db, the receptor for leptin, nucleic acids encoding the receptor, and uses thereof
US7612171B2 (en) 1996-01-16 2009-11-03 The Rockefeller University DB, the receptor for leptin, nucleic acids encoding the receptor, and uses thereof
US7619079B2 (en) 1996-02-14 2009-11-17 The Rockefeller University Db, the receptor for leptin, nucleic acids encoding the receptor, and uses thereof
US7148004B1 (en) 1997-01-16 2006-12-12 The Rockefeller University Oligonucleotides of the OB-R isoforms and methods of diagnosing body weight
US6172211B1 (en) 1997-07-11 2001-01-09 Boehringer Ingelheim International Gmbh Nucleic acid encoding tag7 polypeptide
US6759211B1 (en) 1997-07-11 2004-07-06 Boehringer Ingelheim International Gmbh Tumor growth inhibition- and apoptosis-associated genes and polypeptides and methods of use thereof
US7569220B2 (en) 1997-09-26 2009-08-04 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US6623958B1 (en) 1997-09-26 2003-09-23 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US7842792B2 (en) 1997-09-26 2010-11-30 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US6524818B1 (en) 1997-09-26 2003-02-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US7785831B2 (en) 1997-09-26 2010-08-31 Abt Holding Company Compositions and methods for non-targeted activation of endogenous genes
US6541221B1 (en) 1997-09-26 2003-04-01 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US7033782B2 (en) 1997-09-26 2006-04-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6740503B1 (en) 1997-09-26 2004-05-25 Athersys, Inc. Compositions and methods for non-targeted activation of endogenous genes
US6602686B1 (en) 1997-09-26 2003-08-05 Athersys, Inc. Compositions and method for non-targeted activation of endogenous genes
US6773911B1 (en) 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
US7371834B2 (en) 1998-11-23 2008-05-13 Amgen Inc. Apoptosis-inducing factor
WO2002010364A3 (en) * 2000-07-27 2003-09-25 Univ Delaware Methods for enhancing targeted gene alteration using oligonucleotides
WO2002010364A2 (en) * 2000-07-27 2002-02-07 University Of Delaware Methods for enhancing targeted gene alteration using oligonucleotides
JP2005500841A (en) * 2001-07-27 2005-01-13 アメリカ合衆国 System for in vivo site-directed mutagenesis using oligonucleotides
EP1421187A4 (en) * 2001-07-27 2005-06-15 Us Gov Health & Human Serv Systems for in vivo site-directed mutagenesis using oligonucleotides
US7314712B2 (en) 2001-07-27 2008-01-01 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Systems for in vivo site-directed mutagenesis using oligonucleotides
EP1421187A2 (en) * 2001-07-27 2004-05-26 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Systems for in vivo site-directed mutagenesis using oligonucleotides
US7780965B2 (en) 2002-05-24 2010-08-24 Schering-Plough Animal Health Corp. Eta-1 gene and methods for use
US7205398B2 (en) 2002-05-24 2007-04-17 Schering-Plough Animal Health Corporation Eta-1 gene and methods for use
US8188236B2 (en) 2002-05-24 2012-05-29 Intervet Inc. Eta-1 gene and methods for use
WO2004015117A2 (en) * 2002-08-13 2004-02-19 Nederlands Kanker Instituut Targeted gene modification by single-stranded dna oligonucleotides
WO2004015117A3 (en) * 2002-08-13 2004-06-03 Nl Kanker I Targeted gene modification by single-stranded dna oligonucleotides
US8110185B2 (en) 2008-10-03 2012-02-07 St. Michael's Hospital Method for preventing and treating cardiovascular diseases with BRCA1
US8496928B2 (en) 2008-10-03 2013-07-30 St. Michael's Hospital Method for preventing and treating cardiovascular diseases with BRCA1
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
US10584363B2 (en) 2016-06-03 2020-03-10 Takara Bio Usa, Inc. Methods of producing and using single-stranded deoxyribonucleic acids and compositions for use in practicing the same

Similar Documents

Publication Publication Date Title
WO1990014092A1 (en) Single-strand site-directed modification of mammalian genes in vivo
AU740702B2 (en) Cell-free chimeraplasty and eukaryotic use of heteroduplex mutational vectors
AU2018370588B2 (en) Composition for Genome Editing Using Crispr/CPF1 System and Use Thereof
KR20200121782A (en) Uses of adenosine base editor
Reid et al. Cotransformation and gene targeting in mouse embryonic stem cells
JP7078946B2 (en) Genome editing method
US11833225B2 (en) Methods and compositions for efficient gene deletion
KR20230129230A (en) Compositions and methods for targeting BCL11A
WO2023023515A1 (en) Persistent allogeneic modified immune cells and methods of use thereof
Bandyopadhyay et al. Identification and characterization of satellite III subfamilies to the acrocentric chromosomes
KR20140092853A (en) Transcription unit and use thereof in (yb2/0) expression vectors
WO2000075299A1 (en) Hybrid yeast-bacteria cloning system and uses thereof
CN114560946A (en) Product, method and application of adenine single base editing without PAM limitation
Goncz et al. Small fragment homologous replacement-mediated modification of genomic β-globin sequences in human hematopoietic stem/progenitor cells
WO2011097618A1 (en) Loss of de novo dna methyltransferases promotes expansion of normal hematopoietic stem cells
WO1982000158A1 (en) System for amplification of eukaryotic genes
JP2001509395A (en) Method for identifying artificial transcription regulatory region specific to cell or tissue
US20060063179A1 (en) MIFL- a new partner gene of MLL and methods of use thereof
WO2023025767A1 (en) Method for cas9 nickase-mediated gene editing
Bühler et al. Efficient nonhomologous and homologous recombination in scid cells
König Gene therapy for STAT3-Associated Autosomal-Dominant Hyper-Ige-Syndrome (AD-HIES) using RNA-guided nucleases
CN112048522A (en) Construction method and application of TMEM173 gene humanized and modified animal model
EP4274893A1 (en) Method for producing genetically modified cells
CA3205138A1 (en) Compositions and methods for editing beta-globin for treatment of hemaglobinopathies
IL300563A (en) Nuclease-mediated nucleic acid modification

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB IT LU NL SE