WO1999060108A2 - Transgenic animals produced by homologous sequence targeting - Google Patents

Transgenic animals produced by homologous sequence targeting Download PDF

Info

Publication number
WO1999060108A2
WO1999060108A2 PCT/US1999/010731 US9910731W WO9960108A2 WO 1999060108 A2 WO1999060108 A2 WO 1999060108A2 US 9910731 W US9910731 W US 9910731W WO 9960108 A2 WO9960108 A2 WO 9960108A2
Authority
WO
WIPO (PCT)
Prior art keywords
gene
dna
sequence
targeting
reca
Prior art date
Application number
PCT/US1999/010731
Other languages
French (fr)
Other versions
WO1999060108A3 (en
Inventor
Sushma Pati
David Zarling
Original Assignee
Sri International
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 Sri International filed Critical Sri International
Priority to CA002341350A priority Critical patent/CA2341350A1/en
Priority to AU40797/99A priority patent/AU4079799A/en
Priority to NZ508665A priority patent/NZ508665A/en
Priority to EP99924251A priority patent/EP1078048A2/en
Publication of WO1999060108A2 publication Critical patent/WO1999060108A2/en
Publication of WO1999060108A3 publication Critical patent/WO1999060108A3/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4712Cystic fibrosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4746Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used p53
    • 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
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1003Transferases (2.) transferring one-carbon groups (2.1)
    • C12N9/1018Carboxy- and carbamoyl transferases (2.1.3)
    • 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)
    • 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
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the invention relates to methods for targeting an exogenous polynucleotide or exogenous complementary polynucleotide pair to a predetermined endogenous DNA target sequence in a target cell by homologous pairing, particularly for altering an endogenous DNA sequence, such as a chromosomal DNA sequence, typically by targeted homologous recombination
  • the invention relates to methods for targeting an exogenous polynucleotide having a linked chemical substituent to a predetermined endogenous DNA sequence in a metabolically active target cell, generating a DNA sequence-specific targeting of one or more chemical substituents in an intact nucleus of a metabolically active living target cell, generally for purposes of altering a predetermined endogenous DNA sequence in the cell
  • the invention also relates to compositions and formulations that contain exogenous targeting polynucleotides, complementary pairs of exogenous targeting polynucleotides, chemical substituents of such polynucleotides, and recombinase proteins, including
  • Homologous recombination is defined as the exchange of homologous segments anywhere along a length of two DNA molecules
  • An essential feature of general recombination is that the enzymes responsible for the recombination event can presumably use any pair of homologous sequences as substrates, although some types of sequence may be favored over others
  • Both genetic and cytological studies have indicated that such a crossing-over process occurs between pairs of homologous chromosomes during meiosis in higher organisms
  • targeted homologous recombination provides a basis for targeting and altering essentially any desired sequence in a duplex DNA molecule, such as targeting a DNA sequence in a chromosome for replacement by another sequence
  • Site-specific recombination hag been proposed as one method to integrate transfected DNA at chromosomal locations having specific recognition sites (O'Gorman et al (1991) Science 251 1351 , Onouchi et al (1991 ) Nucleic Acids Res 19 6373)
  • This approach requires the presence of specific target sequences and recombinases, its utility for targeting recombination events at any particular chromosomal location is severely limited in comparison to targeted general recombination
  • Human genetic diseases include (1) classical human genetic diseases wherein a disease allele having a mutant genetic lesion is inherited from a parent (e g , adenosine deaminase deficiency, sickle cell anemia, thalassemias), (2) complex genetic diseases like cancer, where the pathological state generally results from one or more specific inherited or acquired mutations, and (3) acquired genetic disease, such as an integrated provirus (e g , hepatitis B virus)
  • a provirus e g , hepatitis B virus
  • a primary step in homologous recombination is DNA strand exchange, which involves a pairing of a DNA duplex with at least one DNA strand containing a complementary sequence to form an intermediate recombination structure containing heteroduplex DNA (see. Radding, C M (1982) Ann Rev Genet 16 405, U S Patent 4,888,274)
  • the heteroduplex DNA may take several forms, including a three DNA strand containing triplex form wherein a single complementary strand invades the DNA duplex (Hsieh et al (1990) Genes and Development 4 1951 , Rao et al , (1991 ) PNAS 88 2984)) and, when two complementary DNA strands pair with a DNA duplex, a classical Hol day recombination joint or chi structure (Holhday, R (1964) Genet Res 5 282) may form, or a double-D loop ("Diagnostic Applications of Double-D Loop Formation" U S S N 07/755,462, filed 4 September 1991 , which is incorporated herein
  • homologous recombination gene conversion and classical strand breakage/rejoining
  • RecA is required for induction of the SOS repair response, DNA repair, and efficient genetic recombination in E coli RecA can catalyze homologous pairing of a linear duplex DNA and a homologous single strand DNA in vitro
  • proteins like recA which are involved in general recombination recognize and promote pairing of DNA structures on the basis of shared homology, as has been shown by several in vitro experiments (Hsieh and Came ⁇ ni- Otero (1989) J Biol Chem 264 5089, Howard-Flanders et al (1984) Nature 309 215, Stasiak et al (1984) Cold Spring Harbor Svmp Quant Biol 49 561 , Register et al (1987) J Biol Chem 262 12812)
  • recA protein jn vitro to promote homologously paired triplex DNA
  • Exogenous polynucleotides are localized (or targeted) to one or more predetermined DNA target sequence(s) by homologous pairing in vivo
  • Such targeted homologous pairing of exogenous polynucleotides to endogenous DNA sequences in vivo may be used (1 ) to target chemical substituents in a sequence-specific manner m vivo.
  • Some embodiments of the invention employ targeted exogenous polynucleotides to correct endogenous mutant gene alleles in human cells, the invention provides methods and compositions for correcting disease alleles involved in producing human genetic diseases, such as inherited genetic diseases (e g , cystic fibrosis) and neoplasia (e g , neoplasms induced by somatic mutation of an oncogene or tumor suppressor gene, such as p53, or viral genes associated with neoplasia, such as HBV genes)
  • inherited genetic diseases e g , cystic fibrosis
  • neoplasia e g , neoplasms induced by somatic mutation of an oncogene or tumor suppressor gene, such as p53, or viral genes associated with neoplasia, such as HBV genes
  • At least one exogenous polynucleotide is targeted to a predetermined endogenous DNA sequence and alters the endogenous DNA sequence, such as a chromosomal DNA sequence, typically by targeted homologous recombination within and/or flanking the predetermined endogenous DNA sequence
  • a predetermined endogenous DNA sequence such as a chromosomal DNA sequence
  • two complementary exogenous polynucleotides are used for targeting an endogenous DNA sequence
  • the targeting polynucleot ⁇ de(s) are introduced simultaneously or contemporaneously with one or more recombinase species
  • one or more recombinase species may be induced or produced in vivo, for example by expression of a heterologous expression cassette in a cell containing the preselected target DNA sequence
  • the methods of the invention can be used to target such a chemical substituent to a predetermined DNA sequence by homologous pairing for various applications, for example producing sequence-specific
  • the invention can be used to correct genetic mutations, such as base substitutions, additions, and/or deletions, by converting a mutant DNA sequence that encodes a non-functional, dysfunctional, and/or truncated polypeptide into a corrected DNA sequence that encodes a functional polypeptide (e g , has a biological activity such as an enzymatic activity, hormone function, or other biological property)
  • the methods and compositions of the invention may also be used to correct genetic mutations or dysfunctional alleles with genetic lesions in non-coding sequences (e g , promoters, enhancers, silencers, origins of replication, splicing signals)
  • the invention also can be used to target DNA sequences for inactivating gene expression, a targeting polynucleotide can be employed to make a targeted base substitution, addition, and/
  • targeting polynucleotides having at least one associated recombinase are targeted to cells in vivo (i e , in an intact animal) by exploiting the advantages of a receptor-mediated uptake mechanism, such as an asialoglycoprotein receptor-mediated uptake process
  • a targeting polynucleotide is associated with a recombinase and a cell-uptake component which enhances the uptake of the targeting polynucleotide- recombinase into cells of at least one cell type in an intact individual
  • a cell-uptake component typically consists of (I) a galactose-terminal (asialo-) glycoprotein (e g , asi
  • the invention also provides methods and compositions for diagnosis, treatment and prophylaxis of genetic diseases of animals, particularly mammals, wherein a recombinase and a targeting polynucleotide are used to produce a targeted sequence modification in a disease allele of an endogenous gene
  • the invention may also be used to produce targeted sequence mod ⁇ ficat ⁇ on(s) in a non-human animal, particularly a non-human mammal such as a mouse, which create(s) a disease allele in a non-human animal
  • Sequence-modified non-human animals harboring such a disease allele may provide useful models of human and veterinary d ⁇ sease(s)
  • the methods and compositions of the invention can be used to provide nonhuman animals having homologously- targeted human disease alleles integrated into a non-human genome, such non-human animals may provide useful experimental models of human or other animal genetic disease, including neoplastic and other pathogenic diseases
  • a targeting polynucleotide coated with a recombinase may homologously pair with an endogenous chromosomal sequence in a structural or regulatory sequence of a gene and form a stable multistrand complex which may (1 ) constitute a significant physical or chemical obstacle to formation of or procession of an active transcriptional complex comprising at least an RNA polymerase, or (2) alter the local chromatin structure so as to alter the transcription rate of gene sequences within about 1 to 500 kilobases of
  • acquired human and animal diseases particularly parasitic or viral diseases, such as human hepatitis B virus (HBV) hepatitis
  • compositions that contain exogenous targeting polynucleotides, complementary pairs of targeting polynucleotides, chemical substituents of such polynucleotides, and recombinase proteins used in the methods of the invention
  • Such compositions may include a targeting or cell-uptake components to facilitate intracellular uptake of a targeting polynucleotide, especially for jn vivo gene therapy and gene modification
  • the present invention provides methods for targeting and altering, by homologous recombination, a pre-selected target nucleic acid sequence in a cell to make a targeted sequence modification
  • the methods comprise introducing into at least one cell at least one recombinase and at least two single-stranded targeting polynucleotides which are substantially complementary to each other and comprise a homology clamp that substantially corresponds to or is substantially complementary to a preselected target nucleic acid sequence
  • compositions for producing targeted modifications of target sequences comprising two substantially complementary single-stranded targeting polynucleotides and at least one recombinase
  • Fig 1 Homologous targeting of recA-coated chromosome 1 alpha-satellite polynucleotides in metabolically active cell nuclei
  • the homologously targeted biotinylated polynucleotides were visualized by addition of FITC-avidin followed by washing to remove unbound FITC Signals were visualized using a Zeiss Confocal Laser Scanning Microscope (CLSM-10) with 488 nm argon laser beam illumination for FITC-DNA detection
  • Top left -localized FITC-DNA signals in the cell nucleus Lower left -enhanced image of FITC-DNA signals in the cell nucleus
  • Upper right - image of FITC- DNA signals overlaid on the phase image of nucleus Lower right - phase image of the center of the cell nucleus showing nucleoli Note all images except lower right were photographed at the same focus level (focus unchanged between these photos)
  • Fig 6 Tests for alteration of an insertion mutation in the lacZ gene of a eukaryotic expression vector NIH 3T3 cells were needle microinjected with five types of plasmids
  • Two plasmids contained a wild-type ⁇ -galactosidase gene (pMCI Iacpa or pSV- ⁇ -gal [Promega]), a plasmid with a mutant ⁇ -gal gene (pMCIIacXpa), pMCI IacXpa plasmid incubated with a protein uncoated wild-type 276-mer DNA, or pMCIIacXpa plasmid reacted and D-looped with RecA-coated wild-type 276-mer DNA
  • the wild-type 276-mer DNA was heat denatured and either coated or not coated with RecA protein in a standard RecA protein coating reaction protocol (Sena and Zarling, supra) Following a 10-m ⁇ n RecA coating reaction, the Rec
  • Fig 7A Southern hybridization analysis of the 687-bp fragment amplified from genomic DNA Electrophoretic migration of a 687-bp DNA fragment generated with primers CF1 and CF6 from genomic DNA of 3CFTE29o-cells which were capillary needle-micromjected with the 491 -nucleotide DNA fragment in the presence of recA protein (lane 2) or transfected as a protem-DNA-lipid complex where the 491 -nucleotide fragments were coated with recA protein (+, lane 3)
  • the control DNA was amplified from nontransfected 3CFTE29o-cultures (lane 1 )
  • Fig 7B Autoradiographic analysis of DNA transferred to Gene Screen Plus filters and hybridized with a 32 P-labeled oligonucleotide specific for normal exon 10 sequences in the region of the ⁇ F508 mutation Cells transfected by micro-injection or protein-lipid-DNA complexes both were positive for homologous targeting, whereas control cells were not
  • Fig 8A Analysis of DNA from cells electroporated or transfected with CSS DNA encapsulated in a protein- pid complex Allele-specific PCR amplification of the 687/684 bp DNA fragment amplified in the first round with primers CF1 and oligo N (N) or oligo ⁇ F ( ⁇ F) Ethidium bromide-stained 300 bp DNA fragment separated by electrophoresis in a 1% agarose gel
  • the DNA in each lane is as follows lane 1 , 100-bp marker DNA, lane 2, control 16HBE14o-cell DNA amplified with the CF1/N primer pair, lane 3, nontransfected ECFTE29o-cell DNA amplified with CF1/N primers, lane 4, nontransfected
  • ECFTE29o-cell DNA amplified with CF1/ ⁇ F primers lane 5
  • DNA from ECFTE29o-cells electroporated with recA-coated 49l-nucleot ⁇ de fragments and amplified with CF1/N primers lane 6
  • Fig 8B Autoradiographic analysis of the DNA in Fig HA transferred to Gene Screen Plus filters and hybridized with 32 P-labeled oligo N probe Samples in lanes 1-5 for the autoradiographic analysis are equivalent to samples in lanes 2-6 in Fig IIA
  • Figure 10 depicts the scheme for the recombination assay used in Example 4.
  • Fig 11 shows RecA mediated cssDNA targeting to dsDNA with deletions produces a mixed population of probe target hybrids
  • the biotinylated cssDNA probes were denatured and coated with RecA at 37°C as described in Material The reaction mixture was incubated for 60 minutes at 37°C All reactions were stopped by deprotemization with 1 2% SDS and separated by electrophoresis on a 20 cm X 25 cm 1% agarose gel The gel was run overnight at 30V then blotted onto a positively charged Tropilon Plus (TROPIX) membrane The DNA was monitored for the presence of unhybridized probe or probe target hybrids using an alkaline phosphatase based chemiluminescent detection of biotin When the membranes were exposed to X-ray film and developed, it is evident that cssDNA probes will hybridize to dsDNA targets which are completely homologous, as well as dsDNA targets which contain a deletion (lanes 3 and 6, respectively) RecA
  • Fig 12 shows data for the enhanced homologous recombination (EHR) of cssDNA probe target hybrids in E coli, as per Example 4
  • the homologously targeted probe target hybrids have enhanced homologous recombination frequencies in recombination proficient cells
  • cssDNA probe target hybrids were formed as described in the legend of Figure 11 and were introduced into RecA+ and RecA-E coli as in described Figure 12
  • the molar ratio of cssDNA probe target in the in vitro targeting reaction varied from 1 1 to 1 5 6
  • the % recombinant/total colonies is the percentage of blue colonies in the total population of ampicillin-resistant colonies Groups with 0% recombinants did not produce any blue colonies in at least 10 5 plated colonies
  • Plasmid DNA was isolated from blue colonies that were serially propagated for three generations to determine if homologous recombination stably occurred in the lacZ gene
  • Fig 13 shows double D-loop hybrids with internal homology clamps
  • Duplex target DNA (thin line) is completely homologous to the cssDNA probe (thick) and each probe strand can pair with its complementary strand in the target
  • Duplex target has a deletion with respect to the cssDNA probe The deleted region is indicated with a dashed line
  • the region of the cssDNA probes homologous to the deleted region in the target can re-pair with each other forming a stable hybrid complex
  • C
  • Duplex target has an insertion (dashed line) with respect to the cssDNA probe Structures on the left show the re-annealing of cssDNA probe or target strands to form internal homology clamps Structures on the right show the presence of unpaired regions in comparable single D-loop hybrids
  • FIG. 14A depicts the Maps of Plasmids pRD 0 and pRD 59 Relative positions of cssDNA probes IP290 and CP443, PCR primers 1 A and 4B, restriction endonuclease sites EcoRI, Seal, and Dral are indicated The alpha peptide sequence of the LacZ gene is indicated Note the deletion ()) in pRD 59 is approximately equidistant from the ends of primers 1 A and 4B Figure 14B) Time course for cssDNA probe target hybrid formation with linear dsDNA targets Biotinylated, RecA coated cssDNA probe IP290 was hybridized as described to Seal-digested plasmids pRD 0 and pRD 59 carrying 0 or 59 bp deletion, respectively at the EcoRI site in pRD 0 Probe IP290 is completely homologous to pRD 0, but has a 59 bp insertion with respect to pRD 59
  • Fig 15 depicts the formation of cssDNA probe target hybrids formed with linear dsDNA targets containing small deletions
  • Plasmid constructs and probes used in this study A series of plasmids with defined deletions were constructed from the EcoRI site of pRD 0 (pbluesc ⁇ ptllSK+ (Stratagene) as described in Example 5 Each plasmid is named for the size of the deletion, as indicated on the left
  • a series of cssDNA probes were labelled and constructed by PCR from various primers which flank the deleted region Probes were made from either pRD 0 or the deleted plasmids and named for the size of the probe when made from pRD 0 (2960 bp)
  • p527 is 527 bp long
  • IP527 to indicate that the insertion probe (IP) has an insertion with respect to the target
  • IP insertion probe
  • Fig 16 depicts that insertions and deletions have the same effect on the relative efficiency of probe target hybrid formation
  • RecA-coated cssDNA probes IP215 made from pRD 0 was targeted to Seal-digests of plasmids pRD 0, pRD 8, pRD 25, and pRD 59 and compared to similar reactions of DP215 cssDNA probes made from pRD 0, pRD 8, pRD 25, and pRD 59 and targeted to pRD 0
  • the effect of insertions in the cssDNA probe (dark line) is compared with deletions in the cssDNA probe (shaded line) of the same size
  • the relative level of hybrid formation for each cssDNA probe with a heterologous target is normalized by the level of hybridization with the homologous target, respectively
  • the data represents an average of three experiments Error bars are indicated
  • FIG. 17A depicts the formation of stable double-D-Loop hybrids in linear dsDNA targets containing large deletions
  • Biotinylated cssDNA probe IP1246 was coated with RecA protein and targeted to Seal digests of the indicated plasmids as described herein
  • the relative amount of hybrid formation formed between RecA-coated cssDNA probes and plasmids with deletions ranging from 0-967 bp was normalized to the amount of probe target hybrids formed with control probe CP443
  • Autoradiograph (17A) shows the biotinylated cssDNA probes or probe target hybrids
  • the position of the untargeted Seal-digested (Formlll) marker for each of the plasmids are indicated on the right
  • the relative level of hybrid formation (B) of each of the bands in (A) was normalized to the level of hybrid formation with control cssDNA probe CP443, as described herein
  • Figs 18A, 18B, 18C and 18D depict the formation of restriction endonuclease sites in probe target hybrids
  • the probe target hybrids formed between probe IP290 and pRD 0 and pRD 59 targets were deprotemized by extraction with chloroform phenol isoamyl alcohol and chloroform Restriction enzyme treated DNA samples were incubated with EcoRI for three hours before separation on a 1% agarose gel and transferred onto a nylon membrane
  • the ethidium bromide stained DNA of the products of the targeting reactions formed between cssDNA probe IP290 and circular plasmid targets pRD 0 or pRD 59 (A and B) and autoradiographs showing the positions of biotinylated cssDNA probe target hybrids (C and D) are shown
  • the positions of form I and form III markers of pRD 0 are shown on the right
  • the positions of the pRD59 hybrids I* (form I) and rl* (relaxed) are shown on the left
  • Fig 19 depicts the thermal stability of relaxed and non-relaxed probe target hybrids
  • the RecA mediated cssDNA targeting reaction was performed with the cssDNA probe IP290 and the dsDNA target pRD 59, as described herein
  • the probe target hybrids were deprotemized with 1 2% SDS and then incubated for 5 minutes at the indicated temperatures
  • the thermally melted products were then separated on a 1% agarose gel and blotted onto a positively charged Tropilon membrane Autoradiograph shows the position of biotinylated cssDNA probe target hybrids I * (forml) and rl * (relaxed) as shown on the left
  • FIGs 20A and 20B The organization of the mouse OTC gene Sequence of cssDNA probes and PCR primers used in this study are indicated Sizes of the exons in base pairs are indicated The relative position of PCR primers M9, M8 and M11 are shown B) Map of plasmid pTAOTCI A 250 bp fragment containing the normal OTC exon 4 sequence and surrounding introns were cloned into the EcoRV site of pbluesc ⁇ pt SK (+) (Stratagene)
  • Anucleic ac ⁇ d@, Aoi ⁇ gonucleot ⁇ de@, and Apolynucleot ⁇ de@ or grammatical equivalents herein means at least two nucleotides covalently linked together
  • a nucleic acid of the present invention will generally contain phosphodiester bonds, although in some cases nucleic acid analogs are included that may have alternate backbones, comprising, for example, phosphoramide (Beaucage et al , Tetrahedron 49(10) 1925 (1993) and references therein, Letsmger, J Org Chem 35 3800 (1970), Sp ⁇ nzl et al , Eur J Biochem 81 579 (1977), Letsmger et al ,
  • the nucleic acids may be single stranded or double stranded, as specified, or contain portions of both double stranded or single stranded sequence
  • the nucleic acid may be DNA, both genomic and cDNA, RNA or a hybrid, where the nucleic acid contains any combination of deoxy ⁇ bo-and ribonucleotides, and any combination of bases, including uracil, adenme, thymine, cytosme, guanme, inosine, xathanme and hypoxathanme, etc
  • chime ⁇ c DNA-RNA molecules may be used such as described in Cole-Strauss et al , Science 273 1386 (1996) and Yoon et al , PNAS USA 93 2071 (1996), both of which are hereby incorporated by reference
  • the targeting polynucleotides may comprise any number of structures, as long as the changes do not substantially effect the functional ability of the targeting polynucleotide to result in homologous recombination For example, recombinase coating of alternate structures should still be able to occur
  • the terms Apredeterm ed endogenous DNA sequence" and "predetermined target sequence” refer to polynucleotide sequences contained in a target cell Such sequences include, for example, chromosomal sequences (e g , structural genes, regulatory sequences including promoters and enhancers, recombinato ⁇ al hotspots, repeat sequences, integrated proviral sequences, hairpins, palindromes), episomal or extrachromosomal sequences (e g , rephcable plasmids or viral or parasitic replication intermediates) including chloroplast and mitochond ⁇ al DNA sequences
  • predetermined or Apre-selected@ it is meant that the target sequence may be selected at the discretion of the practitioner on the basis of known or predicted sequence information, and is not constrained to specific sites recognized by certain site-specific recombinases (e g , FLP recombinase or CRE recombinase)
  • the predetermined end e.g , FLP
  • a polynucleotide sequence is homologous (i e , may be similar or identical, not strictly evolutiona ⁇ ly related) to all or a portion of a reference polynucleotide sequence, or that a polypeptide sequence is identical to a reference polypeptide sequence
  • the term “complementary to” is used herein to mean that the complementary sequence is homologous to all or a portion of a reference polynucleotide sequence
  • the homology is at least 50-70%, preferably 85%, and more preferably 95% identical
  • the complementarity between two single-stranded targeting polynucleotides need not be perfect
  • the nucleotide sequence "TATAC” corresponds to a reference sequence "TATAC@ and is perfectly complementary to a reference sequence "GTATA”
  • nucleic acid sequence has at least about 60 percent sequence identity as compared to a reference sequence, typically at least about 75 percent sequence identity, and preferably at least about 95 percent sequence identity as compared to a reference sequence
  • the percentage of sequence identity is calculated excluding small deletions or additions which total less than 25 percent of the reference sequence
  • the reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome However, the reference sequence is at least 12-18 nucleotides long, typically at least about 30 nucleotides long, and preferably at least about 50 to 100 nucleotides long
  • ASubstantially complementary refers to a sequence that is complementary to a sequence that substantially corresponds to a reference sequence In general, targeting efficiency increases with the length of the targeting polynu.
  • naturally-occurring® refers to the fact that an object can be found in nature
  • a polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring
  • a metabolically-active cell is a cell, comprising an intact nucleoid or nucleus, which, when provided nutrients and incubated in an appropriate medium carries out DNA synthesis and RNA for extended periods (e g , at least 12-24 hours)
  • Such metabolically-active cells are typically undifferentiated or differentiated cells capable or incapable of further cell division (although non-dividing cells many undergo nuclear division and chromosomal replication), although stem cells and progenitor cells are also metabolically-active cells
  • disease allele@ refers to an allele of a gene which is capable of producing a recognizable disease
  • a disease allele may be dominant or recessive and may produce disease directly or when present in combination with a specific genetic background or pre-existing pathological condition
  • a disease allele may be present in the gene pool or may be generated de novo in an individual by somatic mutation
  • disease to alleles include activated oncogenes, a sickle cell anemia allele, a Tay-Sachs allele, a cystic fibrosis allele, a Lesch-Nyhan allele, a retinoblastoma-susceptibihty allele, a Fabry's disease allele, and a Huntington's chorea allele
  • a disease allele encompasses both alleles associated with human diseases and alleles associated with recognized veterinary diseases
  • the ⁇ F508 CFTR allele in a human disease allele which is
  • cell-uptake component® refers to an agent which, when bound, either directly or indirectly, to a targeting polynucleotide, enhances the intracellular uptake of the targeting polynucleotide into at least one cell type (e g , hepatocytes)
  • a cell-uptake component may include, but is not limited to, the following specific cell surface receptors such as a galactose-terminal (asialo-) glycoprotem capable of being internalized into hepatocytes via a hepatocyte asialoglycoprotein receptor, a polycation (e g , poly-L-lysme), and/or a protein-hpid complex formed with the targeting polynucleotide
  • asialo- galactose-terminal glycoprotem
  • polycation e g , poly-L-lysme
  • mice are derived according to Hogan, et al , "Manipulating the Mouse Embryo A Laboratory Manual®, Cold Spring Harbor Laboratory (1988) which is incorporated herein by reference
  • Embryonic stem cells are manipulated according to published procedures (Teratocarcinomas and embryonic stem cells a practical approach, E J Robertson, ed , IRL Press, Washington, D C , 1987, Zjilstra et al , Nature 342 435-438 (1989), and Schwartzberg et al , Science 246 799-803 (1989), each of which is incorporated herein by reference)
  • Zygotes are manipulated according to known procedures, for example see U S Patent No 4,873,191 , B ⁇ nster et al , PNAS 86 7007 (1989), Susuhc et al , J Biol Chem 49 29483 (1995), and Cavard et al , Nucleic Acids Res 16 2099 (1988), hereby incorporated by reference
  • Oligonucleotides can be synthesized on an Applied Bio Systems oligonucleotide synthesizer according to specifications provided by the manufacturer Modified oligonucleotides and peptide nucleic acids are made as is generally known in the art
  • the present invention provides methods for targeting and altering, by homologous recombination, a pre-selected target nucleic acid sequence in a target cell, to make targeted sequence modifications
  • the methods comprise introducing into the target cells a recombinase and at least two single-stranded targeting polynucleotides which are substantially complementary to each other
  • the targeting polynucleotides each comprise at least one homology clamp that substantially corresponds to or is substantially complementary to the preselected target nucleic acid sequence
  • the target cells are then screened to identify target cells containing the targeted sequence modification
  • Targeting polynucleotides may be produced by chemical synthesis of oligonucleotides, nick-translation of a double-stranded DNA template, polymerase chain-reaction amplification of a sequence (or ligase chain reaction amplification), purification of prokaryotic or target cloning vectors harboring a sequence of interest (e g , a cloned cDNA or genomic clone, or portion thereof) such as plasmids, phagemids, YACs, cosmids, bacte ⁇ ophage DNA, other viral DNA or replication intermediates, or purified restriction fragments thereof, as well as other sources of single and double-stranded polynucleotides having a desired nucleotide sequence
  • Targeting polynucleotides are generally ssDNA or dsDNA, most preferably two complementary single-stranded DNAs
  • Targeting polynucleotides are generally at least about 2 to 100 nucleotides long, preferably at least about 5-to 100 nucleotides long, at least about 250 to 500 nucleotides long, more preferably at least about 500 to 2000 nucleotides long, or longer, however, as the length of a targeting polynucleotide increases beyond about 20,000 to 50,000 to 400,000 nucleotides, the efficiency or transferring an intact targeting polynucleotide into the cell decreases
  • the length of homology may be selected at the discretion of the practitioner on the basis of the sequence composition and complexity of the predetermined endogenous target DNA sequence(s) and guidance provided in the art, which generally indicates that 1 3 to 6 8 kilobase segments of homology are preferred (Hasty et al (1991 ) Molec Cell Biol 11 5586, Shulman et al (1990) Molec Cell Biol 10 4466, which are incorporated herein by reference)
  • Targeting polynucleotides have
  • targeting polynucleotides of the invention have homology clamps that are highly homologous to the predetermined target endogenous DNA sequence(s), most preferably isogenic
  • targeting polynucleotides of the invention have at least one homology clamp that is at least about 18 to 35 nucleotides long, and it is preferable that homology clamps are at least about 20 to 100 nucleotides long, and more preferably at least about 100-500 nucleotides long, although the degree of sequence homology between the homology clamp and the targeted sequence and the base composition of the targeted sequence will determine the optimal and minimal clamp lengths (e g , G-C rich sequences are typically more thermodynamically stable and will generally require shorter clamp length) Therefore, both homology clamp length and the degree of sequence homology can only be determined with reference to a particular predetermined sequence, but homology clamps generally must be at least about 12 nucleotides long and must also substantially correspond or be substantially complementary to a predetermined target sequence
  • a homology clamp is
  • the targeting polynucleotides form a double stranded hybrid, which may be coated with recombinase, although when the recombinase is recA, the loading conditions may be somewhat different from those used for single stranded nucleic acids
  • two substantially complementary single-stranded targeting polynucleotides are used The two complementary single-stranded targeting polynucleotides are usually of equal length, although this is not required However, as noted below, the stability of the four strand hybrids of the invention is putatively related, in part, to the lack of significant unhybridized single-stranded nucleic acid, and thus significant unpaired sequences are not preferred Furthermore, as noted above, the complementarity between the two targeting polynucleotides need not be perfect
  • the two complementary single-stranded targeting polynucleotides are simultaneously or contemporaneously introduced into a target cell harboring a predetermined endogenous target sequence, generally with at lease one recombinase protein (e g , recA) Under most circumstances, it is preferred that the targeting polynucleotides are incubated with recA or other recombinase prior to introduction into a target cell, so that the recombinase prote ⁇ n(s)
  • the use of two complementary single-stranded targeting polynucleotides allows the use of internal homology clamps as depicted in Figure 13
  • the use of internal homology clamps allows the formation of stable deprotemized cssDNA probe target hybrids with homologous DNA sequences containing either relatively small or large insertions and deletions within a homologous DNA target
  • these probe target hybrids, with heterologous inserts in the cssDNA probe are stabilized by the re-anneahng of cssDNA probes to each other within the double-D-loop hybrid, forming a novel DNA structure with an internal homology clamp
  • stable double-D-loop hybrids formed at internal sites with heterologous inserts in the linear DNA targets are equally stable Because cssDNA probes are kmetically trapped within
  • the length of the internal homology clamp (i e the length of the insertion or deletion) is from about 1 to 50% of the total length of the targeting polynucleotide, with from about 1 to about 20% being preferred and from about 1 to about 10% being especially preferred, although in some cases the length of the deletion or insertion may be significantly larger
  • the complementarity within the internal homology clamp need not be perfect
  • a targeting polynucleotide is introduced into a target cell simultaneously or contemporaneously with a recombinase protein, typically in the form of a recombinase coated targeting polynucleotide as outlined herein (i e , a polynucleotide pre-mcubated with recombinase wherein the recombinase is noncovalently bound to the polynucleotide, generally referred to in the art as a nucleoprotein filament)
  • a targeting polynucleotide used in a method of the invention typically is a single-stranded nucleic acid, usually a DNA strand, or derived by denaturation of a duplex DNA, which is complementary to one (or both) strand(s) of the target duplex nucleic acid
  • one of the complementary single stranded targeting polynucleotides is complementary to one strand of the endogeneous target sequence (i e Watson) and the other complementary single stranded targeting polynucleotide is complementary to the other strand of the endogeneous target sequence (i e Crick)
  • the homology clamp sequence preferably contains at least 90-95% sequence homology with the target sequence, to insure sequence-specific targeting of the targeting polynucleotide to the endogenous DNA target
  • Each single-stranded targeting polynucleotide is typically about 50-600 bases long, although a shorter or longer polynucleotide may also be employed Alternatively, targeting polynucleotides may
  • Recombinase Proteins are proteins that, when included with an exogenous targeting polynucleotide, provide a measurable increase in the recombination frequency and/or localization frequency between the targeting polynucleotide and an endogenous predetermined DNA sequence
  • increases in recombination frequency from the normal range of 10 "8 to 10 "4 , to 10 "4 to 10 1 , preferably 10 3 to 10 1 , and most preferably 10 2 to 10 1 may be acheived
  • recombinase refers to a family of RecA-hke recombination proteins all having essentially all or most of the same functions, particularly (i) the recombinase protein's ability to properly bind to and position targeting polynucleotides on their homologous targets and (n) the ability of recombinase protein/targeting polynucleotide complexes to efficiently find and bind to complementary endogenous sequences
  • the best characterized recA protein is from E coli, in addition to the wild-type protein a number of mutant recA-hke proteins have been identified (e g , recA803, see Madiraju et al , PNAS USA 85(18) 6592 (1988), Madiraju et al, Biochem 31 10529 (1992), Lavery et al , J Biol Chem 267 20648 (1992)) Further, many organisms have recA-hke recombinases with strand-transfer activities (e g
  • RecA may be purified from E coli strains, such as E coli strains JC12772 and JC15369 (available from A J
  • the recA803 protein is a high-activity mutant of wild-type recA
  • the art teaches several examples of recombinase proteins, for example, from Drosophila, yeast, plant, human, and non-human mammalian cells, including proteins with biological properties similar to recA (i e , recA-hke recombinases), such as Rad51 from mammals and yeast, and Pk-rec (see Rashid et al , Nucleic Acid Res 25(4) 719 (1997), hereby incorporated by reference)
  • the recombinase may actually be a complex of proteins, i e a Arecomb ⁇ nosome@
  • included within the definition of a recombinase are portions or fragments of recombinases which retain recombina
  • recA or rad51 is used
  • recA protein is typically obtained from bacterial strains that overproduce the protein wild-type E coli recA protein and mutant recA803 protein may be purified from such strains
  • recA protein can also be purchased from, for example, Pharmacia (Piscataway, NJ)
  • this nucleoprotein filament one monomer of recA protein is bound to about 3 nucleotides
  • This property of recA to coat single-stranded DNA is essentially sequence independent, although particular sequences favor initial loading of recA onto a polynucleotide (e g , nucleation sequences)
  • the nucleoprotein filament(s) can be formed on essentially any DNA molecule and can be formed in cells (e g , mammalian cells), forming complexes with both single-stranded and double-stranded DNA, although the loading conditions for dsDNA are somewhat different than for ssDNA
  • RecA protein coating of targeting polynucleotides is typically carried out as described in U S S N 07/910,791 , filed 9 July 1992 and U S S N 07/755,462, filed 4 September 1991 , which are incorporated herein by reference Briefly, the targeting polynucleotide, whether double-stranded or single-stranded, is denatured by heating in an aqueous solution at 95-100°C for five minutes, then placed in an ice bath for 20 seconds to about one minute followed by centrifugation at 0°C for approximately 20 sec, before use When denatured targeting polynucleotides are not placed in a freezer at -20°C they are usually immediately added to standard recA coating reaction buffer containing ATP(S, at room temperature, and to this is added the recA protein Alternatively, recA protein may be included with the buffer components and ATP(S before the polynucleotides are added
  • RecA coating of targeting polynucleot ⁇ de(s) is initiated by incubating polynucleotide-recA mixtures at 37°C for 10-15 mm RecA protein concentration tested during reaction with polynucleotide varies depending upon polynucleotide size and the amount of added polynucleotide, and the ratio of recA molecule nucleotide preferably ranges between about 3 1 and 1 3
  • the mM and ⁇ M concentrations of ATP(S and recA, respectively can be reduced to one-half those used with double-stranded targeting polynucleotides (i e , recA and ATP(S concentration ratios are usually kept constant at a specific concentration of individual polynucleotide strand, depending on whether a smgle- or double-stranded polynucleotide is used)
  • RecA protein coating of targeting polynucleotides is normally carried out in a standard 1X RecA coating reaction buffer 10X RecA reaction buffer (i e , 10x AC buffer) consists of 100 mM T ⁇ s acetate (pH 7 5 at 37°C), 20 mM magnesium acetate, 500 mM sodium acetate, 10 mM DTT, and 50% glycerol) All of the targeting polynucleotides, whether double-stranded or single-stranded, typically are denatured before use by heating to 95-100°C for five minutes, placed on ice for one minute, and subjected to centrifugation (10,000 rpm) at 0°C for approximately 20 seconds (e g , in a Tomy centrifuge) Denatured targeting polynucleotides usually are added immediately to room temperature RecA coating reaction buffer mixed with ATP(S and diluted with buffer or double-distilled H 2 0 as necessary
  • a reaction mixture typically contains the following components (i) 0 2-4 8 mM ATP(S, and (n) between 1-100 ng/ ⁇ l of targeting polynucleotide
  • To this mixture is added about 1-20 ⁇ l of recA protein per 10- 100 ⁇ l of reaction mixture, usually at about 2-10 mg/ml (purchased from Pharmacia or purified), and is rapidly added and mixed
  • the final reaction volume-for RecA coating of targeting polynucleotide is usually in the range of about 10-500 ⁇ l RecA coating of targeting polynucleotide is usually initiated by incubating targeting polynucleotide-RecA mixtures at 37°C for about 10-15 mm
  • RecA protein concentrations in coating reactions varies depending upon targeting polynucleotide size and the amount of added targeting polynucleotide recA protein concentrations are typically in the range of 5 to 50 ⁇ M
  • concentrations of ATP(S and recA protein may optionally be reduced to about one-half of the concentrations used with double-stranded targeting polynucleotides of the same length that is, the recA protein and ATP(S concentration ratios are generally kept constant for a given concentration of individual polynucleotide strands
  • the coating of targeting polynucleotides with recA protein can be evaluated in a number of ways
  • protein binding to DNA can be examined using band-shift gel assays (McEntee et al , (1981 ) J_ Biol Chem 256 8835)
  • Labeled polynucleotides can be coated with recA protein in the presence of ATP(S and the products of the coating reactions may be separated by agarose gel electrophoresis Following incubation of recA protein with denatured duplex DNAs the recA protein effectively coats single-stranded targeting polynucleotides derived from denaturing a duplex DNA
  • the ratio of recA protein monomers to nucleotides in the targeting polynucleotide increases from 0, 1 27, 1 2 7 to 3 7 1 for 121-mer and 0, 1 22, 1 2 2 to 4 5 1 for 159-mer
  • targeting polynucleotide's electrophoretic mobility decreases, i e , is retarded, due to
  • a second method for evaluating protein binding to DNA is in the use of nitrocellulose filter binding assays (Leahy et al , (1986) J Biol Chem 261 6954. Woodbury, et al , (1983) Biochemistry 22(20) 4730-4737
  • the nitrocellulose filter binding method is particularly useful in determining the dissociation-rates for protein DNA complexes using labeled DNA
  • DNA protein complexes are retained on a filter while free DNA passes through the filter
  • This assay method is more quantitative for dissociation-rate determinations because the separation of DNA protein complexes from free targeting polynucleotide is very rapid
  • recombinase prote ⁇ n(s) may be exogenously induced or administered to a target cell simultaneously or contemporaneously (i e , within about a few hours) with the targeting polynucleot ⁇ de(s) Such administration is typically done by micro-injection, although electroporation, lipofection, and other transfection methods known in the art may also be used
  • recombinase-proteins may be produced m vivo For example, they may be produced from a homologous or heterologous expression cassette in a transfected cell or transgenic cell, such as a transgenic totipotent cell (e g a fertilized zygote) or an embryonal stem cell (e g , a mu ⁇ ne ES cell such as AB-1 ) used to generate a transgenic non-human animal line or a somatic cell or a plu ⁇ potent hematopoietic stem cell
  • a targeting polynucleotide of the invention may optionally be conjugated, typically by covalently or preferably noncovalent binding, to a cell-uptake component
  • a targeting polynucleotide of the invention can be conjugated to essentially any of several cell-uptake components known in the art
  • a targeting polynucleotide can be conjugated to an asialoorosomucoid (ASOR)-poly-L- lysme conjugate by methods described in the art and incorporated herein by reference (Wu GY and Wu CH (1987) J Biol Chem 262 4429, Wu GY and Wu CH (1988) Biochemistry 27 887, Wu GY and Wu CH (1988) J Biol Chem 263 14621.
  • a cell-uptake component may be formed by incubating the targeting polynucleotide with at least one lipid species and at least one protein species to form protein-hpid-polynucleotide complexes consisting essentially of the targeting polynucleotide and the lipid-protem cell-uptake component
  • Lipid vesicles made according to Feigner (W091/17424, incorporated herein by reference) and/or cationic hpidization (WO91/16024, incorporated herein by reference) or other forms for polynucleotide administration (EP 465,529, incorporated herein by reference) may also be employed as cell-uptake components
  • Nucleases may also be used as cell-uptake components
  • targeting components such as nuclear localization signals may be used, as is known in the art
  • the targeting polynucleotides may include chemical substituents Exogenous targeting polynucleotides that have been modified with appended chemical substituents may be introduced along with recombinase (e g , recA) into a metabolically active target cell to homologously pair with a predetermined endogenous DNA target sequence in the cell
  • the exogenous targeting polynucleotides are derivatized, and additional chemical substituents are attached, either during or after polynucleotide synthesis, respectively, and are thus localized to a specific endogenous target sequence where they produce an alteration or chemical modification to a local DNA sequence
  • Preferred attached chemical substituents include, but are not limited to cross-linking agents (see Podyminogm et al , Biochem 34 13098 (1995) and 35 7267 (1996), both
  • a targeting polynucleotide of the invention is coated with at least one recombinase and is conjugated to a cell-uptake component, and the resulting cell targeting complex is contacted with a target cell under uptake conditions (e g , physiological conditions) so that the targeting polynucleotide and the recomb ⁇ nase(s) are internalized in the target cell
  • a targeting polynucleotide may be contacted simultaneously or sequentially with a cell-uptake component and also with a recombinase, preferably the targeting polynucleotide is contacted first with a recombinase, or with a mixture comprising both a cell-uptake component and a recombinase under conditions whereby, on average, at least about one molecule of recombinase is noncovalently attached per targeting polynucleotide molecule and at least about one cell-uptake component also is noncovalently attached Most preferably, coating of both re
  • Cell-uptake components are included with recombmase-coated targeting polynucleotides of the invention to enhance the uptake of the recombmase-coated targeting polynucleot ⁇ de(s) into cells, particularly for in vivo gene targeting applications, such as gene therapy to treat genetic diseases, including neoplasia, and targeted homologous recombination to treat viral infections wherein a viral sequence (e g , an integrated hepatitis B virus (HBV) genome or genome fragment) may be targeted by homologous sequence targeting and inactivated
  • a targeting polynucleotide may be coated with the cell-uptake component and targeted to cells with a contemporaneous or simultaneous administration of a recombinase (e g , liposomes or immunohposomes containing a recombinase, a viral-based vector encoding and expressing a recombinase)
  • the recombinase-targeting polynucleotide compositions are formulated, they are introduced or administered into target cells
  • the administration is typically done as is known for the administration of nucleic acids into cells, and, as those skilled in the art will appreciate, the methods may depend on the choice of the target cell Suitable methods include, but are not limited to, microinjection, electroporation, lipofection, etc
  • Atarget cells® herein is meant prokaryotic or eukaryotic cells
  • Suitable prokaryotic cells include, but are not limited to, bacteria such as E coli, Bacillus species, and the extremophile bacteria such as thermophiles, etc
  • the procaryotic target cells are recombination competent Suitable eukaryotic cells include, but are not limited to, fungi such as yeast and filamentous fungi, including species of Aspergillus, Tnchoderma, and Neurospora, plant cells including those of corn, sorghum, tobacco, canola, soybean, cotton, tomato, potato,
  • procaryotic cells are used in this embodiment, a pre-selected target DNA sequence is chosen for alteration
  • the pre-selected target DNA sequence is contained within an extrachromosomal sequence
  • Aextrachromosomal sequence® herein is meant a sequence separate from the chromosomal or genomic sequences
  • Preferred extrachromosomal sequences include plasmids (particularly procaryotic plasmids such as bacterial plasmids), P1 vectors, viral genomes, yeast, bacterial and mammalian artificial chromosomes (YAC, BAC and MAC, respectively), and other autonomously self-rephcating sequences, although this is not required
  • a recombinase and at least two single stranded targeting polynucleotides which are substantially complementary to each other, each of which contain a homology clamp to the target sequence contained on the extrachromosomal sequence, are added to the extrachromosomal sequence, preferably in vitro The two single strand
  • the pre-selected target DNA sequence is a chromosomal sequence
  • the recombinase with the targeting polynucleotides are introduced into the target cell, preferably eukaryotic target cells
  • it may be desirable to bind (generally non-covalently) a nuclear localization signal to the targeting polynucleotides to facilitate localization of the complexes in the nucleus See for example Kido et al , Exper Cell Res 198 107-114 (1992), hereby expressly incorporated by reference
  • the targeting polynucleotides and the recombinase function to effect homologous recombination, resulting in altered chromosomal or genomic sequences
  • eukaryotic cells are used for making transgenic non-human animals (which include homologously targeted non-human animals) embryonal stem cells (ES cells) and fertilized zygotes are preferred
  • embryonal stem cells are used Mu ⁇ ne ES cells, such as AB-1 line grown on mitotically inactive SNL76/7 cell feeder layers (McMahon and Bradley, Cell 62 1073-1085 (1990)) essentially as described (Robertson, E J (1987) in Teratocarcinomas and Embryonic Stem Cells A Practical Approach E J Robertson, ed (oxford IRL Press), p 71-112) may be used for homologous gene targeting
  • Other suitable ES lines include, but are not limited to, the E14 line (Hooper et al (1987) Nature 326 292-295), the D3 line (Doetschman et al (1985) J Embrvol Exp Morph 87 21-45), and the CCE line (Robertson
  • the plunpotence of any given ES cell line can vary with time in culture and the care with which it has been handled
  • the only definitive assay for plunpotence is to determine whether the specific population of ES cells to be used for targeting can give rise to chimeras capable of germhne transmission of the ES genome
  • a portion of the parental population of AB-1 cells is injected into C57B1/6J blastocysts to ascertain whether the cells are capable of generating chime ⁇ c mice with extensive ES cell contribution and whether the majority of these chimeras can transmit the ES genome to progeny
  • non-human zygotes are used, for example to make transgenic animals, using techniques known in the art (see U S Patent No 4,873,191 )
  • Preferred zygotes include, but are not limited to, animal zygotes, including fish, avian and mammalian zygotes
  • Suitable fish zygotes include, but are not limited to, those from species of salmon, trout, tuna, carp, flounder, halibut, swordfish, cod, tulapia and zebrafish
  • Suitable bird zygotes include, but are not limited to, those of chickens, ducks, quail, pheasant, turkeys, and other jungle fowl and game birds
  • Suitable mammalian zygotes include, but are not limited to, cells from horses, cattle, buffalo, deer, sheep, rabbits, rodents such as mice, rats, hamsters and guinea pigs, goats, pigs, primates, and marine mammals including dolphins and whales See
  • the compositions of the invention find use in a number of applications, including the creation of transgenic plants and animals
  • transgenic animals can be any of the animals, fish and birds outlined above as suitable for zygotes
  • the transgenic animals are mammals, including, but not limited to, farm animals such as cattle, buffalo, goats, including BELE® goats, sheep, and pigs or other transgenic animals such as mice, rabbits, monkeys, etc
  • the animals or mammals are non-human
  • transgenic animals are made with any number of changes Exogeneous sequences, or extra copies of endogeneous sequences, including structural genes and regulatory sequences, may be added to the animal, as outlined below Endogeneous sequences (again, either genes or regulatory sequences) may be disrupted, i e via insertion, deletion or substitution, to prevent expression of endogeneous proteins Alternatively, endogeneous sequences may be modified to alter their biological function, for example via mutation of the endogeneous sequence by insertion, deletion or substitution
  • exogenous DNA sequences such as exogenous genes or regulatory sequences, extra copies of endogenous genes or regulatory sequences, or exogeneous genes or regulatory sequences
  • exogenous DNA sequences such as exogenous genes or regulatory sequences, extra copies of endogenous genes or regulatory sequences, or exogeneous genes or regulatory sequences
  • a transgenic plant or animal This may be done for a number of reasons for example, adding one or more copies of a wild-type gene can increase the production of a desirable gene product, adding or deleting one or more copies of a therapeutic gene can alleviate a disease state, or to create an animal model of disease Adding one or more copies of a modified wild type gene may be done for the same reasons Adding therapeutic genes or proteins may yield superior transgenic animals, for example for the production of therapeutic or nut ⁇ ceutical proteins Adding human genes to non-human mammals may facilitate production of human proteins and adding regulatory sequences derived from human or non-human mammals may be useful to increase or decrease the expression of endogenous or exogenous genes Such inserted genes may
  • Suitable endogenous gene targets include, but are not limited to, genes which encode peptides or proteins including enzymes, structural or soluble proteins, as well as endogeneous regulatory sequences including, but not limited to, promoters, transcriptional or translational sequences, repetitive sequencs including ol ⁇ go[d(A-C) n « d(G-T) n ], ohgo[d(A-T)] n , ol ⁇ go[d(C-T)] n , etc
  • Examples of such endogenous gene targets include, but are not limited to, genes which encode lactoglobuhns including both ⁇ -lactoglobuhn and $-lactoglobuhn, casein, including both ⁇ -casem, ⁇ -casem and ⁇ -case ⁇ n, albumins, including serum albumin, particularly human and bovine, immunoglobuhns, including IgE, IgM, I
  • Endogeneous genes may be modified in several ways, including disruptions and alterations
  • the endogenous target gene may be disrupted in a variety of ways
  • Adisrupt® as used herein comprises a change in the coding or non-coding sequence of an endogenous nucleic acid that alters the transcription or translation of an endogenous gene
  • a disrupted gene will no longer produce a functional gene product
  • disruption may occur by either the insertion, deletion or frame shifting of nucleotides
  • Ainsertion sequence® as used herein means one or more nucleotides which are inserted into an endogenous gene to disrupt it
  • insertion sequences can be as short as 1 nucleotide or as long as a gene, as outlined below
  • the sequences are at least 1 nucleotide, with from about 1 to about 50 nucleotides being preferred, and from about 10 to 25 nucleotides being particularly preferred
  • An insertion sequence may comprise a polylinker sequence, with from about 1 to about 50 nucleotides being preferred, and from about 10 to 25 nucleotides being particularly preferred
  • an insertion sequence comprises a gene which not only disrupts the endogenous gene, thus preventing its expression, but also can result in the expression of a new gene product
  • the disruption of an endogenous gene by an insertion sequence gene is done in such a manner to allow the transcription and translation of the insertion gene
  • An insertion sequence that encodes a gene may range from about 50 bp to 5000 bp of cDNA or about 5000 bp to 50000 bp of genomic DNA As will be appreciated by those in the art, this can be done in a variety of ways
  • the insertion gene is targeted to the endogenous gene in such a manner as to utilize endogenous regulatory sequences, including promoters, enhancers or a regulatory sequence
  • the insertion sequence gene includes its own regulatory sequences, such as a promoter, enhancer or other regulatory sequence etc
  • insertion sequence genes include, but are not limited to, genes which encode therapeutic and nut ⁇ ceutical proteins, and reporter genes Suitable insertion sequence genes which may be inserted into endogenous genes include, but are not limited to, nucleic acids which encode those genes listed as suitable endogeneous genes for alterations, above, particularly mammalian enzymes, mammalian antibodies, mammalian proteins including serum albumin as well as mammalian therapeutic genes
  • the inserted mammalian gene is a human gene
  • Suitable reporter genes are those genes which encode detectable proteins, such as the genes encoding luciferase, ⁇ -galactosidase (both of which require the addition of reporter substrates), and the fluorescent proteins, including green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), and red fluorescent protein (RFP)
  • the targeted sequence modification creates a sequence that has a biological activity or encodes a polypeptide having a biological activity
  • the polypeptide is an enzyme with enzymatic activity
  • the polypeptide is an antibody
  • the polypeptide is a structural protein
  • the insertion sequence genes may be modified or variant genes, i e they contain a mutation from the wild-type sequence
  • modified genes including, but not limited to, improved therapeutic genes, modified "-lactalbumm genes that do not encode any phenylalanme residues, or human enzyme or human antibody genes that do not encode any phenylalanme residues
  • Adeletion® as used herein comprises removal of a portion of the nucleic acid sequence of an endogenous gene Deletions range from about 1 to about 100 nucleotides, with from about 1 to 50 nucleotides being preferred and from about 1 to about 25 nucleotides being particularly preferred, although in some cases deletions may be much larger, and may effectively comprise the removal of the entire endogenous gene and/or its regulatory sequences Deletions may occur in combination with substitutions or modifications to arrive at a final modified endogenous gene
  • endogenous genes may be disrupted simultaneously by an insertion and a deletion
  • some or all of an endogenous gene, with or without its regulatory sequences may be removed and replaced with an insertion sequence gene
  • all but the regulatory sequences of an endogenous gene may be removed, and replaced with an insertion sequence gene, which is now under the control of the endogenous gene's regulatory elements
  • Aregulatory element® is used herein to describe a non-coding sequence which affects the transcription or translation of a gene including, but are not limited to, promoter sequences, ⁇ bosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, enhancer or activator sequences, or dime ⁇ zing sequences
  • the regulatory sequences include a promoter and transcriptional start and stop sequence
  • Promoter sequences encode either constitutive or inducible promoters
  • the promoters may be either naturally occurring promoters or hybrid promoters Hybrid promoters, which combine elements of more than one promoter, are also known in the art, and are useful in the present invention
  • the endogeneous genes may be altered by substitutions, insertions or deletions of nucleotides that do not completely eliminate the biological function of the sequence, but rather alter it That is, targeted gene modifications may be made to alter gene function
  • defective genes may be fixed, or the activity of a gene may be modulated, either increasing or decreasing the activity of the sequence (either the nucleic acid sequence, for example in the case of regulatory nucleic acid, or of the gene product, i e the ammo acid sequence of the protein may be altered)
  • Suitable targets for such fully or partially modified regulatory sequences include, but are not limited to, regulatory sequences that regulate any of the suitable endogeneous genes listed above, with preferred embodiments altering the endogeneous regulatory sequences that control the genes which encode "-lactoglobulm, $-lactoglobuhn, casein, ⁇ -casem, ⁇ - casein, ⁇ -case ⁇ n, serum albumin, globin, IgG, integ ⁇ n, lactofer ⁇ n, a refroviral provirus, a p ⁇ on, alpha-galactosyl transferase (galT), a sugar transferase or a milk or urine production gene
  • Examples of such fully or partially modified endogenous regulatory sequences include, but are not limited to, a modified regulatory element for an endogenous gene, a modified transcriptional regulation cassette or start site for an endogenous gene, a modified promoter, transcription initiation site, or enhance
  • substitutions are less conservative than those shown in Chart I
  • substitutions may be made which more significantly affect the structure of the polypeptide backbone in the area of the alteration, for example the ⁇ -hehcal or ⁇ -sheet structure, the charge or hydrophobicity of the molecule at the target site, or the bulk of the side chain
  • substitutions which in general are expected to produce the greatest changes in the polypeptide's properties are those in which (a) a hydrophilic residue, e g seryl or threonyl, is substituted for (or by) a hydrophobic residue, e g leucyl, isoleucyl, phenylalanyl, valyl or alanyl, (b) a cysteme or prolme is substituted for (or by) any other residue, (c) a residue having an electropositive side chain, e g lysyl, argmyl, or histidyl, is substituted
  • Preferred embodiments of the present invention include, but are not limited to (1 ) a farm animal including cattle, sheep, pigs, horses and goats with a 1-25 base pair deletion, or a 10-25 base pair insertion of a poly nker sequence, or insertion of a reporter gene such as a luciferase gene, a ⁇ - galactosidase gene or a green fluorescent (GFP) protein gene in an endogenous gene or sequence encoding ornithme transcarbamylase (OTC), lactoglobulm, casein, ⁇ -casein, ⁇ -casein, ⁇ -case ⁇ n, albumin, globin, immunoglobulm, IgG, mterleukin, a sugar transferase, integ ⁇ n, a milk protein, a urine protein, a retroviral provirus, an endogenous virus, a p ⁇ on, a leptm, or cystic fibrosis transmembrane regulator (CFTR), (2) a
  • Additional preferred embodiments comprise a pig, monkey or cow with a 1-25 to 1-50 base pair insertion, examples of which include a hormone receptor gene, a viral receptor gene or a G-protein coupled receptor gene, or a 1-25 to 1-50 bp deletion in a sugar transferase gene including the ⁇ - galactosyl transferase gene (galT) or the fucosyl transferase gene, a BELE® goat with a human gene, and a pig, goat, sheep or cow with a 1-25 base pair insertion or a 1-25 base pair deletion in a endogenous refroviral provirus gene such as deletion of the sequence for proviral KC
  • Further specific preferred embodiments include, a cow with a modified milk production gene such as, a cow with a lactase gene insertion in a milk promoter, a cow with the human lactofer ⁇ n gene replacing the bovine lactofer ⁇ n gene, a monkey with a human therapeutic gene, or a human antibody gene, a
  • the vectors containing the DNA segments of interest can be transferred into the host cell by well- known methods, depending on the type of cellular host
  • micro-injection is commonly utilized for target cells, although calcium phosphate treatment, electroporation, lipofection, biolistics or viral-based transfection also may be used
  • Other methods used to transform mammalian cells include the use of Polybrene, protoplast fusion, and others (see, generally, Sambrook et al Molecular Cloning A Laboratory Manual, 2d ed , 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N Y , which is incorporated herein by reference)
  • Direct injection of DNA and/or recombmase-coated targeting polynucleotides into target cells, such as skeletal or muscle cells also may be used (Wolff et al (1990) Science 247 1465, which is incorporated herein by reference)
  • compositions of the invention find use in a number of applications, including the site directed modification of endogeneous sequences within any target cell, the creation of transgenic plants and animals, and the use of the compositions to do site-directed mutagenesis or modifications of target sequences
  • any predetermined endogenous DNA sequence such as a gene sequence
  • the target polynucleotides have at least one homology clamp which substantially corresponds to or is substantially complementary to a predetermined endogenous DNA target sequence and are introduced with a recombinase (e g , recA) into a target cell having the predetermined endogenous DNA sequence
  • a targeting polynucleotide (or complementary polynucleotide pair) has a portion or region having a sequence that is not present in the preselected endogenous targeted sequence(s) (i e , a nonhomologous portion or mismatch) which may be as small as a single mismatched nucleotide, several mismatches, or may span up to about several kilobases or more of nonhomo
  • a targeting polynucleotide is used to repair a mutated sequence of a structural gene by replacing it or converting it to a wild-type sequence (e g , a sequence encoding a protein with a wild-type biological activity)
  • a wild-type sequence e g , a sequence encoding a protein with a wild-type biological activity
  • such applications could be used to convert a sickle cell trait allele of a hemoglobin gene to an allele which encodes a hemoglobin molecule that is not susceptible to sickhng, by altering the nucleotide sequence encoding the $-subun ⁇ t of hemoglobin, so that the codon at position 6 of the $-subun ⁇ t is converted fromVal$6->Glu$6 (Shesely et al (1991) op cit )
  • Other genetic diseases can be corrected, either partially or totally, by replacing, inserting, and/or deleting sequence information in a disease allele using appropriately selected exogenous targeting polynucle
  • the combination of (I) a targeting polynucleot ⁇ de(s), (2) a recombinase (to provide enhanced efficiency and specificity of correct homologous sequence targeting), and (3) a cell- uptake component (to provide enhanced cellular uptake of the targeting polynucleotide), provides a means for the efficient and specific targeting of cells m vivo, making in vivo homologous sequence targeting, and gene therapy, practicable
  • Several disease states may be amenable to treatment or prophylaxis by targeted alteration of heptocytes m vivo by homologous gene targeting
  • the following diseases are expected to be amenable to targeted gene therapy hepatocellular carcinoma, HBV infection, familial hypercholesterolemia (LDL receptor defect), alcohol sensitivity (alcohol dehydrogenase and/or aldehyde dehydrogenase insufficiency), hepatoblastoma, Wilson's disease, congenital hepatic porphy ⁇ as, inherited disorders of hepatic metabolism, ornithine transcarbamylase (OTC) alleles, HPRT alleles associated with Lesch Nyhan syndrome, etc
  • a cell-uptake component consisting essentially of an asialoglycoprotem-poly-L- lysme conjugate is preferred
  • the targeting complexes of the invention which may be used to target
  • the methods and compositions of the invention are used for gene inactivation That is, in addition to correcting disease alleles, exogenous targeting polynucleotides can be used to inactivate, decrease or alter the biological activity of one or more genes in a cell (or transgenic nonhuman animal) This finds particular use in the generation of animal models of disease states, or in the elucidation of gene function and activity, similar to Aknock out® experiments These techniques may be used to eliminate a biological function, for example, a galT gene (alpha galactosyl transferase genes) associated with the xenoreactivity of animal tissues in humans may be disrupted to form transgenic animals (e g pigs) to serve as organ transplantation sources without associated hyperacute rejection responses Alternatively, the biological activity of the wild-type gene may be either decreased, or the wild-type activity altered to mimic disease states This includes genetic manipulation of non-coding gene sequences that affect the transcription of genes, including, promoters, repressors, enhancers and
  • Plasmids are engineered to contain an appropriately sized gene sequence with a deletion or insertion in the gene of interest and at least one flanking homology clamp which substantially corresponds or is substantially complementary to an endogenous target DNA sequence
  • Vectors containing a targeting polynucleotide sequence are typically grown in E coli and then isolated using standard molecular biology methods, or may be synthesized as oligonucleotides Direct targeted inactivation which does not require vectors may also be done When using microinjection procedures it may be preferable to use a transfection technique with linearized sequences containing only modified target gene sequence and without vector or selectable sequences
  • the modified gene site is such that a homologous recombinant between the exogenous targeting polynucleotide and the endogenous DNA target sequence can be identified by using carefully chosen primers and PCR, followed by analysis to detect if PCR products specific to the desired targeted event are present (
  • the methods of the present invention are useful to add exogeneous DNA sequences, such as exogeneous genes or extra copies of endogeneous genes, to an organism
  • this may be done for a number of reasons, including to alleviate disease states, for example by adding one or more copies of a wild-type gene or add one or more copies of a therapeutic gene, to create disease models, by adding disease genes such as oncogenes or mutated genes or even just extra copies of a wild-type gene, to add therapeutic genes and proteins, for example by adding tumor suppressor genes such as p53, Rb1 , Wt1 , NF1 , NF2, and APC, or other therapeutic genes, to make superior transgenic animals, for example superior livestock, or to produce gene products such as proteins, for example for protein production, in any number of host cells
  • Suitable gene products include, but are not limited to, Rad51 , alpha-antitrypsin, casein, hormones, antithrombin III, alpha glucosidase, collagen
  • the targeted sequence modification creates a sequence that has a biological activity or encodes a polypeptide having a biological activity
  • the polypeptide is an enzyme with enzymatic activity
  • a preferred embodiment utilizes the methods of the present invention to create novel genes and gene products
  • fully or partially random alterations can be incorporated into genes to form novel genes and gene products, to produce rapidly and efficiently a number of new products which may then be screened, as will be appreciated by those in the art
  • compositions and methods of the invention are useful in site-directed mutagenesis techniques to create any number of specific or random changes at any number of sites or regions within a target sequence (either nucleic acid or protein sequence), similar to traditional site-directed mutagenesis techniques such as cassette mutagenesis and PCR mutagenesis
  • the techniques and compositions of the invention may be used to generate site specific variants in any number of systems, including E coli, Bacillus, Archebacte ⁇ a, Thermus, yeast (Sacchromyces and Pichia), insect cells (Spodoptera, Tnchoplusia, Drosophila), Xenopus, rodent cell lines including CHO, NIH 3T3 and primate cell lines including COS, or human cells, including HT1080 and BT474, which are traditionally used to make variants
  • the techniques can be used to make specific changes, or random changes, at a particular site or sites, within a particular region or regions of the sequence, or over the entire sequence
  • suitable target sequences include nucleic acid sequences encoding therapeutically or commercially relevant proteins, including, but not limited to, enzymes (proteases, recombinases, lipases, kinases, carbohyd rases, isomerases, peptides tautomerases, nucleases etc ), hormones, receptors, transcription factors, growth factors, antibodies, cytokmes, globin genes, immunosupppressive genes, tumor suppressors, oncogenes, complement-activating genes, milk proteins (casein, "-lactalbumin, ⁇ -lactoglobuhn, whey proteins, serum albumin), immunoglobuhns, urine proteins, milk proteins, esterases, pharmaceutical proteins and vaccines
  • enzymes proteases, recombinases, lipases, kinases, carbohyd rases, isomerases, peptides tautomerases, nucleases etc
  • hormones proteases, recomb
  • the methods of the invention are used to generate pools or libraries of variant nucleic acid sequences, and cellular libraries containing the variant libraries
  • a plurality of targeting polynucleotides are used
  • the targeting polynucleotides each have at least one homology clamp that substantially corresponds to or is substantially complementary to the target sequence
  • the targeting polynucleotides are generated in pairs, that is, pairs are made of two single stranded targeting polynucleotides that are substantially complementary to each other (i e a Watson strand and a Crick strand)
  • less than a one to one ratio of Watson to Crick strands may be used, for example, an excess of one of the single stranded target polynucleotides (i e Watson) may be used
  • sufficient numbers of each of Watson and Crick strands are used to allow the majority of the targeting polynucleotides to form double D-loops, which are
  • the plurality of pairs preferably comprise a pool or library of mismatches
  • the size of the library will depend on the number of residues to be mutagenized, as will be appreciated by those in the art Generally, a library in this instance preferably comprises at least 40% different mismatches, with at least 30% mismatches being preferred and at least 10% being particularly preferred That is, the plurality of pairs comprise a pool of random and preferably degenerate mismatches over some regions or all of the entire targeting sequence
  • Amismatches® include substitutions, insertions and deletions
  • a pool of degenerate variant targeting polynucleotides covering some, or preferably all, possible mismatches over some region are generated, as outlined above, using techniques well known in the art
  • the variant targeting polynucleotides each comprise only one or a few mismatches (less than 10), to allow complete multiple randomization, as outlined below
  • the introduction of a pool of variant targeting polynucleotides (in combination with recombinase) to a target sequence can result in a large number of homologous recombination reactions occu ⁇ ng over time That is, any number of homologous recombination reactions can occur on a single target sequence, to generate a wide variety of single and multiple mismatches within a single target sequence, and a library of such variant target sequences, most of which will contain mismatches and be different from other members of the library This thus works to generate a library of mismatches
  • the variant targeting polynucleotides are made to a particular region or domain of a sequence (i e a nucleotide sequence that encodes a particular protein domain) For example, it may be desirable to generate a library of all possible variants
  • the variant targeting polynucleotides are made to the entire target sequence In this way, a large number of single and multiple mismatches may be made in an entire sequence
  • the methods of the invention may be used to create superior recombinant reporter genes such as lacZ,iziciferase and green fluorescent protein (GFP), superior antibiotic and drug resistance genes, superior recombinase genes, superior recombinant vectors, and other superior recombinant genes and proteins, including peptides, immunoglobuhns, vaccines or other proteins with therapeutic value
  • superior recombinant reporter genes such as lacZ,iziciferase and green fluorescent protein (GFP), superior antibiotic and drug resistance genes, superior recombinase genes, superior recombinant vectors, and other superior recombinant genes and proteins, including peptides, immunoglobuhns, vaccines or other proteins with therapeutic value
  • targeting polynucleotides containing any number of alterations may be made to one or more functional or structural domains of a protein, and then the products of homologous recombination evaluated
  • the target cells may be screened to identify a cell that contains the targeted sequence modification This will be done in any number of ways, and will depend on the target gene and targeting polynucleotides, as will be appreciated by those in the art
  • the screen may be based on phenotypic, biochemical, genotypic, or other functional changes, depending on the target sequence
  • selectable markers or marker sequences may be included in the targeting polynucleotides to facilitate later identification
  • kits containing the compositions of the invention include the compositions, particularly those of libraries or pools of degenerate cssDNA probes, along with any number of reagents or buffers, including recombinases, buffers, ATP, etc.
  • Homologously targeted exogenous targeting polynucleotides specifically target human DNA sequences in intact nuclei of metabolically active cells RecA-coated complementary exogenous targeting polynucleotides were introduced into metabolically active human cells encapsulated in agarose microbeads and permeabihzed to permit entry of DNA/protem complexes using the Jackson- Cook method (Cook, P R (1984) EMBO J 3 1837, Jackson and Cook (I985) EMBO J 4 919, Jackson and Cook (1985) EMBO J 4 913, Jackson and Cook (1986) J Mol Biol 192 65, Jackson et al (1988) J Cell Sci 90 365, which are incorporated herein by reference) These experiments were designed to specifically target homologous DNA sequences with recA protein in intact nuclei of metabolically active human HEp-2 cells
  • the nuclear membranes of human or other cells may be permeabihzed without loss of metabolic function when the cells are first encapsulated in a gel of agarose microbeads
  • the agarose microbead coat contains the cell constituents and preserves native conformation of chromosomal DNA, while permitting diffusion of macromolecules into and out of the cell compartment Wittig et al (1991) Proc Natl Acad Sci (U S A ).
  • RecA-mediated homologous gene targeting with complementary oligonucleotides in intact human cell nuclei was verified directly by homologous targeting using targeting polynucleotides that were biotinylated These were subsequently labeled with a fluorescent reporter compound to verify homologous pairing at specific locations having the predetermined sequence(s) RecA-coated targeting polynucleotides for human chromosome 1 pe ⁇ centromet ⁇ c alpha-satellite DNA sequences were specifically targeted to chromosome 1 centromere sequences in living human cell nuclei that were permeabihzed and suspended in agarose
  • the recA-coated targeting polynucleo ⁇ des were targeted to the repetitive alpha satellite sequences of chromosome 1 This result showed DNA targeting in intact nuclei to specific human chromosome 1 sequences (data not shown)
  • HEp-2 cells were embedded in agarose prepared with I x PBS, New Buffer treated, then treated with 100 ng of biotinylated targeting polynucleotide complementary to chromosome 1 alpha-satellite DNA Controls in this experiment also included reactions without recA protein and additional control reactions supplemented with an identical amount of BSA protein to replace the recA protein Additionally, cells were also embedded in agarose prepared with I x AC buffer Examples of specific targeting to endogenous target sequences were recorded
  • recA-coated biotinylated targeting polynucleotides for human chromosome 1 alpha- satellite DNA were specifically targeted to human HEp-2 epithelial carcinoma chromosomal DNA in intact cell nuclei of metabolically active cells that had been suspended in agarose, then treated with buffers and recA-coated targeting polynucleotides under suitable reaction conditions (supra and U S S N 07/755,462, U S S N 07/755,462, and U S S N 07/520,321 , incorporated herein by reference)
  • suitable reaction conditions supra and U S S N 07/755,462, U S S N 07/755,462, and U S S N 07/520,321 , incorporated herein by reference
  • Nuclei were washed in 1 x AC buffer and incubated with recA-coated complementary single-stranded DNA oligonucleotides (i e , exogenous targeting polynucleotides) for 3 5 hours at 37°C
  • the alpha satellite DNA targeting polynucleotides for chromosome 1 were previously described and were nick- translated with biotinylated deoxyribonucleotides (b ⁇ o-14-dATP)
  • the p53 tumor suppressor gene polynucleotide was obtained from Oncor (209 Perry Parkway, Gaithersburg, MD 20877) and is a 1 2 kilobase cDNA fragment from a wild-type human p53 gene (Fields and Jang, (1990) Science 242 1046, Miller et al (1986) Nature 319 783, Zakut-Houre et al (1985) EMBO J 4 1251 )
  • the 1 2 kilobase human p53 DNA was nick-tran
  • the targeting technique employs the unique properties of recA-mediated DNA sequence targeting with single- stranded (complementary) short targeting polynucleotides Native intact nuclei were incubated with labeled, heat-denatured targeting polynucleotides coated with recA protein The DNA hybridized to the predetermined targeted homologous sequences
  • This in vivo targeting by recA-mediated homologous targeting polynucleotides shows the targeting specificity and therapeutic potential for this new in vivo methodology
  • Application of recA or other recombmase-mediated targeting of (complementary) ssDNA or denatured dsDNA targeting polynucleotides to predetermined endogenous DNA targets is important for gene eatry, gene knockout, gene replacement, and gene mutation or correction
  • EXAMPLE 2 Correcting a Mutant Gene to Produce a Functional Gene Product Homologously targeted complementary DNA oligonucleotides were used to correct 11 bp insertion mutations in vector genes and restore vector gene expression and vector protein function in microinje ⁇ te mammalian cells
  • Expe ⁇ ments were designed to testwhether homologously targeted complementary 276-bp oligonucleote! targeting polynucleotides could correct an 11-bp insertion mutation in the lacZ gene of a mammahalMA vector, which encoded a nonfunctional $-galactos ⁇ dase, so that a corrected lacZ gene encoded and expressed a functional enzyme Functional enzyme ($-galactos ⁇ dase) was detected by an X-gal assay that turns cells expressing a revertant (i e , corrected) lacZ gene a blue color
  • NIH3T3 cells microinjected with the mutant test vector bearing an 11 basepair insertion mdflacZ coding sequence do not produce any detectable functional $-galactos ⁇ dase enzyme
  • cells microinjected with the wild type test vector do produce functional enzyme
  • pMCIIacpA for use as a positive control for expression of $- galactosidase
  • pMCI IacXpA is the target test mutant plasmid (shown in Figure 3) It is identical to pMCI IacpA (shown in F ⁇ gure4) but has a 11-bp Xbal linker insertional mutation
  • This plasmid does not express $-galactos ⁇ dase activity in rouse NIH3T3 cells when introduced by electroporation It does not produce blue color in the presence of X-gal indicative of $-galactos ⁇ dase production following vector micro-injection Negative controls with mock or noninjected cells we also done Using these conditions and NIH3T3 cells have no detectable background blue staining
  • the plasmid pMC1 lacpA (8 4 kb) contains the strong polyoma virus promoter of transcription plus ATG placed in front of the lacZ gene The polyadenylation signal from SV40 virus was placed in back of the lacZ gene
  • the plasmid vector was plB130 from IBI (New Haven, CT)
  • the mutant vector pMCIIacpA has a 11-bp inserton in the Xbal site consisting of the inserted sequence CTCTAGACGCG (see Figure
  • NIH3T3 cells were capillary needle microinjected with targeting polynucleotide-target DNA mixtures ade in glass pipettes freshly pulled into microneedles using a Sutter instruments microprocessor controlled apparatus
  • An ECET Eppendorf microinjection pump ad computerized micromanipulator were used for computer-assisted microinjection using an Olympus IMT-2 inverted microscope Cells were carefully microinjected under controlled pressure and time NIH3T3 cells injected with pMCIIacpA showed approximately 9% of the injected cells were blue None (0%) of the celfeijected with pMCIIacXpA DNA in reactions containing the 271 bp oligonucleotide but without recA protein showed a blue color In marfeie contrast, approximately 3 6% of the cells microinjected with the recA-coated 271-bp targeting polynucleotide targeted to-the pMCIIacXpA target hybd were blue ( Figure 6),
  • CF cystic fibrosis
  • CFTR CF transmembrane conductance regulator
  • Targeting polynucleotides that were coated with recA protein were introduced into transformed CF airway epithelial cells, homozygous for bothers ⁇ F508 CFTR gene mutation, by either intranuclear microinjection, ele ⁇ tporation, or by transfection with a protein-DNA-hpid complex
  • An immortalized CF tracheobronchial epithelial huma ⁇ cell line, ECFTE290-, is homozygous for the ⁇ F508 mutation (Kunzelmann et al (1993) Am J Respir Cell Mol Biol . 8 522) These cells are useful as targets for homologous recombination analysis, because they contain the same 3 basepair deletion in CFTR allele on all copies of chomosome 7 Replacement of the ⁇ F508 allele with wild-type CFTR DNA in indicated only when homologous recombination has occurred
  • the 491 bp region of the CFTR gene spanning exon 11 and containing 3' and 5' flanking mtron sequences was selected from sequence data published previously (Zielenski et al (1991) Genomics 10 214, incorporated herein by reference) and used as a targeting polynucleotide
  • TheDNA fragment was PCR amplified in preparative quantities and then denatured for introduction into cells as recA-coated complementary s
  • the 491 bp fragment was generated by PCR amplification from the T6/20 plasmid (Rommene-t al (1989) op cit . incorporated herein by reference) ad verified by restriction enzyme mapping and propagated as described previously After digestion with EcoRI and Hindlll, a 860 bp insert was isolated following electrophoresis in 0 8% SeaPlaque agarose gel
  • the 860 bp fragmti contained CFTR exon 10, as well as 5' and 3' mtron sequences, as defined by the restriction enzyme cleavagates (Zielenski et al (1991 ) op cit )
  • a 50 ng aliquot of the fragment was amplified by PCR using primers CF1 and CF5 (Table 1 ) to generate a 491 bp fragment
  • the conditions for amplification were denaturation, 94°C for 1 annealing, 53°C for 30 sec, extension, 72°C for 30 sec witha 4 sec
  • the 491 nucleotide fragments were coaid with recA protein using the reaction buffer of Cheng (Cheng, et al (1988) J Biol Chem 263 15110, incorporated herein by reference)
  • the 491 bp DNA fragment (5 ⁇ g) was denatured at 95°C for 10 mn, then added to a 63 ⁇ l of coating buffer containing 200 ⁇ g of recA protein, 4 8 mM ATP(S, and 1 7 ⁇ l reaction buffer (100 mM T ⁇ s-Ac, pH 7 5 at 37°C, 10 m M dithiothreitol, 500 mM NaOAc, 20 mM MgOAc, 50 percent glycerol) and incubated for 10 mm at 37°C Next, the MgOAc concentration was increased to a final concentration of about 22 mM by addition of 7 ⁇ l of 200 mM MgOAc Under these conditions, the 491 nucleotide fragment was coated witbcrA protein at a
  • Microinjection when used, was performed with an Eppendorf 5242 microinjection pump fitted to an Eppendorf 5170 micromanipulator using borosihcate pipete (Brunswick, 1 2 OD x 1 9ID) fabricated into a microneedle with a Sutter instruments (P-87) micropipette puller
  • the micropipettes were filled by capillary force from the opposite side of the needle Approximately 100 pipettes were used for injecting 4000 cells Cells were injected with approximately 1,000-10,000 fragments per cell by intranuclear injection witi 120 hPa for 0 1-0 3 s at a volume of 1-10 fl/nucleus Microinjected cells were viewed with an Olympus IMT-2 inverted microscope during the injection The area of the petri dish containing icjed cells was marked with 2 to 5 mm diameter rings Needlsnicromjection was performed in cells grown on 10 separate 60 mm petri dishes Cells were injected at room temperature in culture medium after two washes in phosphat
  • Electroporation experiments were performed using recA-coated 491 -mer ssDNA as described above Approximately 1 x 10? exponentially growing cells were supended in 400 ⁇ l of coating buffer with 5 ⁇ g of recA coated-DNA The cell suspension wa pre-mcubated on ice for 10 mm and electroporated at room temperature with 400 V and 400 ⁇ F in a BTX 300 electroporator (BTX Corporation, San Diego, CA) Afte electroporation, cells were incub ed on ice for an additional 10 mm, diluted in Eagle's minimal essential medium (MEM) supplemented with 10% fetal bovine serum (FBS) and HD ⁇ g/ml streptomycin, 100 U/ml penicillin (Cozens et al (1992Proc Natl Acad Sci (U S A )89 5171 , Gruenert et al (1988) Proc Natl Acad Sci (U S A ) 85 5951 , Kunzelmann, (1992) o
  • Protein DNA-lipid complexes were prepared Briefly, dioleoylphosphatidyl-ethanolamine (PtdEtn, DOPE) was used for preparing liposomes by ng 4 ⁇ M solutions of the lipid under nitrogen at room temperature The lipid film was rehydrated with 4 ml of 30 mM T ⁇ s-HC1 buffer (pH 9), then sonicated for 15 minutes under an atmosphere or argon The protem-DNA complex was prepared in polystyrene tubes by diluting 20 ⁇ g of recA-coated 491-base DNA in 30 mM T ⁇ s-HC1 , (pH 9) buffer Gramicidin S protein (GmS) was also diluted with 30 mM T ⁇ s HC1 (pH 9) to a final concentration of 2 mg/ml from a 20 mg/ml stock solution prepared in dimethyl sulfoxide The protein (40 ⁇ g) was added to the DNA and rapidly mixed Next, 175 ⁇ l of the h
  • Genomic DNA was isolated and purified from cells as described in Maniatisp cit to test for homologous DNA recombination
  • Cellular DNA was firsPCR-amphfied with primers CF1 and CF6 (Table 1 )
  • CF1 is within the region of homologydefined at the 5' end of the 491 bp CFTR fragment
  • CF6 is outside the regro of homology at the 3' end of this fragment
  • the conditions for the PCR amplification were as follows CF1/CF6, 684/687 bp fragment, primers, 0 5 ⁇ M, DNA, 1-2 ⁇ g, denaturation, 94°C for 1 mm, annealing, 53°C for 45 s, extension, 72°C for 90 s wit h a 4-s/cycle increase in extension time for 40 cycles, Mg +2 1 5 mM DNA fragments were separated by agarose electrophoresis and visualized by staining with ethidium bromide, then transferred to Gene Screen Plus filters (DuPont) The DNA was then hybridized with the allele-specific normal CFTW>-end- labeled DNA probe defined by oligo N as described by Cozens et al (1992) op cit . Kunzelmann (1992) op cit , incorporated herein by reference The presence of wild-type (WT) sequences was determined autoradiographically by hybridization with the radiolabeled DNA probe
  • Cytoplasmic RNA was isolated and denatured at 95°C for 2 mm, then reveestranscnbed using the DNA polymerase provided in a PCR RNA Gene Amp kit according to manufacturer's instructions (Perkin- Elmer/Cetus)
  • First strand cDNA was amplified by using primer CF17 at the 5' end of exon 9 and the allele-specific oligo N or ohgo ⁇ F primers
  • the length of the PCR fragments is 322 bp (CF17/ohgo J tand 321 bp (CF17/ohgo ⁇ F)
  • the conditions for PCR amplification are CF17/ohgo N/ ⁇ F, 322/321 bp fragment, primers, 1 ⁇ M, denaturation, 94°C forl mm, annealing, 51 °C for 30s, extension, 72°C for 20s with a 4-s/cycle increase in extension time for 40 cycles, Mg +2 , 0 8 mM DNA fragments were visualized after electrophoresis on ethidium bromide-stained 1 % agarose gels
  • Southern hybridization was performed as described above Fragments were transferred to Gene Screen Plus filtes then hybridized with allele-specific oligo N probe under the same conditions used for the Southern analysis of the genomic DNA (Kuretmann et al (1992) op cit .
  • the CF17 primer is located at the 5' end of exon 9 and was used for amplification of first strand cDNA derived from CFTR mRNA
  • Oligo N and Oligo ⁇ F are allele-specific probes and can also be used as allele-specific PCR primers for amplifying the 300/299 bp fragments (DNA analysis) and the 322/321 bp fragments (RNA analysis)
  • Sense (S) and antisense (A) primers are designated under DNA Strand and indicate the sense of the strand relative to the transcribed direction (i e , the CFTR mRNA)
  • cytoplasmic CFTR mRNA was analyzed for the presence of a normal CFTR RNA sequence in the ⁇ F508 region of exon 10 Cytoplasmic RNA was isolated from the cells, reverse- transcribed with DNA polymerase and PCR-amphfied as first-strand cDNA This amplification was performed with a PCR primer located in exon 9 (CF17, sense) and CFTR allele-specific PCR primer in exon 10 (oligo N or ⁇ F, antisense) The exon 10 primer contains the CF mutation site, and the resulting fragment is 322 bp in normal DNA or 321 bp in DNA containing the ⁇ F508 mutation Amplification of genomic DNA is eliminated by using primers that require amplification across mtron/exon boundaries Amplified cDNA generated from normal control 16HBE140- cells and experimentally transfected cells yielded DNA product fragments with the CF17/o
  • CFTR DNA can homologously recombme with targeting polynucleotides comprising small fragments of WT CFTR DNA resulting in a corrected genomic CFTR allele, and that a recA-coated targeting polynucleotide can be used in transfection reactions in cultured human cells, and that cystic fibrosis ⁇ F508 mutations can be corrected in genome DNA resulting in the production of normal CFTR cytoplasmic mRNA
  • the plasmid pRD 59 was made from the 2 9 kb cloning vector pBluesc ⁇ pt IISK(-) (pRD 0) (Stratagene)
  • the pRD 0 DNA was linearized at a unique EcoRI site in the poiyhnker region of the lacZ gene and digested with mung bean nuclease (Boeh ⁇ nger-Mannheim)
  • the plasmids were then ligated and transformed into the RecA(-) E coli host XL1-Blue (Stratagene)
  • the resulting alpha peptide mutant clones were screened for lack of alpha-complementation of ⁇ - galactosidase activity, which results in white colonies when grown on plates containing X-gal and IPTG (Sambrook et al , 1989) Plasmid DNAs recovered from white colonies by a mini-prep procedure (Qiagen) lacked the unique EcoRI site, as well as the
  • Biotinylated cssDNA probes were made from a fragment of the normal pBluesc ⁇ pt IISK(-) plasmid
  • the plasmid DNA was linearized with Bgll and run on a 1% agarose gel in 1X TAE After ethidium bromide staining, the 1 6 kB fragment band was excised from the gel and purified using the Qiaex II gel purification method (Qiagen) This 1 6 kb fragment was diluted 1 20 and then used as a template for PCR
  • the PCR reaction mixture contained b ⁇ ot ⁇ n-14-dATP (GIBCO-BRL) in order to synthesize IP290, a 290 bp biotinylated cssDNA probe homologous to the LacZ region of pRD 0
  • pRD 59 was linearized with Bgll and the 1 55 kb fragment was purified in the same manner as the pRD 0 1 6 kb fragment Using the same primer
  • biotinylated cssDNA probes 70 ng were denatured by heat at 98°C for 10 minutes, cooled immediately in an ice-water bath, and then cent ⁇ fuged at 4°C for 10 seconds to recover all liquids Reactions without cssDNA probe contained equivalent volumes of water
  • the denatured cssDNA probes were then coated with RecA protein (Boeh ⁇ nger-Mannheim) in T ⁇ s-acetate reaction buffer (Cheng et al , 1988, 10 mM T ⁇ s-acetate (pH 7 5), 1 mM dithiothreitol, 50 mM sodium acetate, 2 mM magnesium acetate, 5% (v/v) glycerol) with 2 43 mM ATPS for 15 minutes at 37°C in a 10 ⁇ l volume Reactions without the RecA protein contained equivalent volumes of RecA storage buffer (20 mM Tri
  • the RecA mediated targeting reactions were performed by adding 1- 4 ⁇ g of the appropriate plasmid DNA in an aqueous solution containing 22 mM magnesium acetate, bringing the final magnesium concentration to 11 mM and the final reaction volume to 20 ⁇ l The reaction was incubated for another 60 minutes at 37°C
  • Biotinylated cssDNA probes and probe target hybrids were detected using the Southern-Light System (Tropix)
  • the nylon bound DNA blots were treated with avidin conjugated to alkaline phosphatase, followed by the chemiluminescent substrate, CDP-Star (Tropix), in conditions described by the manufacturer Blots were exposed to X-ray film (Kodak) for varying times (1 minute to 8 minutes) and developed Electroporation of probe target DNA hybrids into metabolically active E coli cells After purification of targeted complexes, 40 ⁇ l of electro-competent RecA(+) and/or RecA(-) E coli (Dower et al , 1988) was added to 30-200 ng of the targeted complexes in a chilled microfuge tube The RecA(+) cells were BB4 (Stratagene) and the RecA(-) cells were XL1-Blue (Stratagene) The mixture was incubated on ice for 1 minute This mixture was then transferred
  • the presence of a sizable region of non-homologous nucleotide sequences (59 bp) in the cssDNA probe IP290 does not significantly affect the ability of the RecA coated cssDNA probe IP290 to form stable probe target hybrids with pRD 59 in comparison to completely homologous dsDNA pRD 0 ( Figure 11 , lane 3 and 6) In each reaction, under these conditions, the presence of the RecA protein was absolutely required for hybrid detection ( Figure 11 , lane 2 and 5) Probe
  • Figure 12 shows the percentage of potential recombinant blue colonies formed when IP290 probe pRD 59 target hybrids were electroporated into RecA+ and RecA- cells Blue colonies only arose when deprotemized hybrids formed with pRD 59 and cssDNA probe IP290 are introduced into RecA+ E coli cells Control experiments performed with cssDNA probes homologous to the mutant LacZ region of pRD 59 (DP231) and homologous to a region outside of the LacZ gene (CP443) did not yield any blue colonies ( Figure 12) In addition, when all of these hybrids were transformed into RecA(-) hosts, no blue colonies were produced from any type of hybrid, indicating the the recombinogenic effect is also dependent on endogenous RecA protein produced in the cell Thus only the cssDNA probe containing the 59 base pair correction produces recombinogenic clones in bacterial host cells that are RecA(+)
  • ds target substrates A series of plasmid DNA targets with defined deletions were constructed by linearization of the plasmid vector pBluescnpt IISK(-) (Stratagene) at a unique EcoRI restriction site in the polylinker region following digestion with mung bean exonuclease (Boeh ⁇ nger- Mannheim), DNA ligation, and subsequent transformation into XL1-Blue E coli (Stratagene) by standard methods The resulting clones were sequenced using Sanger dideoxy sequencing methods (Sequenase Kit version 2, USB) to determine the extent of deletion A series of plasmids with deletions ranging from 4 to 967 bp were prepared and named for the extent of size of the deletion (see Figure 15) The size of the parent plasmid, pBluescnpt IISK(-), referred to as pRD 0 in this study, is 2960 bp Plasmid DNA was prepared by a modified alkaline lysis procedure with anion
  • Biotin-labeled probes homologous to pRD 0 were synthesized by PCR with incorporation of b ⁇ ot ⁇ n-14-dATP using previously described methods where the molar ratio of unlabelled dATP to biotm-labelled dATP was 3 1 (Griffin & Griffin, 1995)
  • Primer pairs flanking the polylinker region of pRD 0 or analogous plasmids with a deletion were chosen to produce PCR fragments which span the deletion in the target plasmids
  • a control PCR fragment (CP443) primer pair flanking sequences outside the polylinker was selected for production of a probe homologous to all clones in the plasmid series
  • the oligonucleotide products were purified by membrane ultrafiltration using Microcon 100 filters (Amicon)
  • the relative level of hybrid formation with heterologous targets was expressed as a percentage of the level of hybrid formation of standardized reactions with a completely homologous probe and target These values were normalized to the level of hybrid formation that occured with control probe CP443 which hybridizes to all of the plasmid targets in a region away from the heterology
  • the data generally represent averages of at least three separate measurements from three independent targeting reactions
  • Plasmid targets (pbluesc ⁇ ptllSK(+), Stratagene Figure 14A) Plasmid targets (pRD 4 - pRD 967) are named for the size of deletion in bp at the EcoRI site CssDNA probes were made and labelled with b ⁇ ot ⁇ n-14-dATP by PCR using primers which symet ⁇ cally flank the deleted region of plasmids in the pRD series CssDNA probes made from pRD 0 that were targeted to plasmids containing deletions are called insertion probes and named for the length of the probe in bp
  • IP290 is a 290 bp cssDNA probe that contains an insertion with respect to a target containing a deletion, but is completely homologous to pRD 0
  • a cssDNA probe made from pRD 59 and targeted to pRD 0 is called DP231 , since it contains a deletion with respect to pRD 0, but is completely homologous to pRD
  • Stable cssDNA probe target hybrids are formed in linear dsDNA targets with deletions at internal sites.
  • cssDNA probe IP290 with either a completely homologous linear target, pRD 0 or a target carrying a 59 bp deletion, pRD 59
  • Probe IP290 symet ⁇ cally spans the 59 bp deletion in pRD 59
  • Figure 15B shows that in steady state hybrid reactions, the maximum level of stable hybrid formation when RecA-coated IP290 is targeted to pRD 59 is 62% of the steady state level obtained with the fully homologous target pRD 0 Furthermore steady state levels of hybrid formation occurs within 45 minutes with fully homologous pRD 0 targets, but requires 2 hours for pRD 59 targets Thus, in all subsequent experiments RecA-coated probes were hybridized
  • Ornithme transcarbamylase is a mitochondnal matrix enzyme that catalyzes the synthesis of citrulline from ornithme and carbamylphosphate in the second step of the mammalian urea cycle
  • OTC deficiency in humans is the most common and severe defect of the urea cycle disorders
  • OTC is an X-lmked gene that is primarily expressed in the liver and to a lesser extent in the small intestine
  • Affected males develop hyperammonemia, acidosis, orotic acidu ⁇ a, coma and death occurs in up to 75% of affected males, regardless of intervention
  • Two allehc mutations at the OTC locus are known in mice spf and spf-ash, (sparse fur-abnormal skin and hair)
  • spf-ash mice can be readily identified by the abnormal skin and hair phenotype
  • the spf-ash mutation is a single-base substitution at the end
  • RecA-coated probe A 230 bp fragment from the normal mouse OTC gene was amplified by PCR with primers M9 and M8 from pTAOTC ( Figure 20)
  • the PCR fragment was purified on M ⁇ crocon-100 columns (Amicon) and then extensively dialyzed
  • the M9-M8 amphcon was denatured by heating the fragments to 98°C and then coated with RecA protein (Boeh ⁇ nger- Mannheim) at a ratio 3 nucleotides/ protein monomer
  • the final concentration of RecA-coated DNA in coating buffer (5 mM T ⁇ sOAc, pH 7 5, 0 5 mM DTT, 10 mM MgOAc, 1 22 mM ATP(S, 5 5 ⁇ M RecA) was 5 ng/ ⁇ L RecA-coated filaments were made on the day of microinjection and then stored on ice until use
  • Plasmid pTAOTCI carries a 250 bp segment of exon4 and surrounding mtron sequences from the normal mouse OTC gene
  • a 230 bp cssDNA probe OTC1 was prepared by PCR amplification of pTAOTCI with primers M9 and M8 cssDNA probe
  • OTC1 was denatured and coated with RecA protein as described herein
  • Homozygous spf-ash/spf-ash female and hemizygous (spf-ash/y) males can be phenotypically identified by the appearance of sparse fur and wrinkled skin early in development
  • a cross between homozygous spf-ash/spf-ash B6C3H females and normal B6D2F1 males yields heterozygous phenotypically normal females and hemizygous males with sparse fur and wrinkled skin
  • RecA-coated cssDNA probe OTC1 recombines with the homologous chromosomal copy of the OTC gene in fertilized mouse zygotes
  • genomic DNA was isolated from tail biopsies
  • Genomic DNA was amplified with either the primer set M10-M11 or M54-M11 to produce either a 250 bp or 314 bp amphcon
  • the DNA amphcon represents DNA from the endogenous OTC gene PCR fragments from all of the eight mice and several female mice were sequenced to determine the base sequence at the spf-ash locus to determine if a normal allele (G) or a mutant allele (A) was present in the genomic DNA
  • Figure 21 shows sequencing gels of representative reactions The panel on the left side shows the sequence of the homozygous spf-ash females that donated the eggs to produce the fertilized zygotes where only the mutant base A is present at the
  • mouse #16 was mated with homozygous (spf-ash/spf-ash) mutant females
  • the resultant progeny will either be hemizygous (spf-ash/Y) mutant males or homozygous (spf-ash/spf-ash) mutant females, both of which are phenotypically mutant
  • heterozygous (spf-ash/+) females that are phenotypically normal will be produced
  • two litters were produced that consisted of a total of 5 hemizygous (spf-ash/Y) mutant males, 7 homozygous (spf-ash/spf-ash) mutant females,

Abstract

The invention relates to methods and compositions for producing transgenic animals by targeted homologous recombination comprising targeting an exogenous polynucleotide or exogenous complementary polynucleotide pair to a predetermined endogenous DNA target sequence in a target cell by homologous pairing, particularly for altering an endogenous DNA sequence, such as a chromosomal DNA sequence. In certain embodiments, the invention relates to compositions that contain exogenous targeting polynucleotides, complementary pairs of exogenous targeting polynucleotides, chemical substituents of such polynucleotides, and recombinase proteins used in the methods of the invention.

Description

TRANSGENIC ANIMALS PRODUCED BY HOMOLOGOUS SEQUENCE TARGETING
FIELD OF THE INVENTION
The invention relates to methods for targeting an exogenous polynucleotide or exogenous complementary polynucleotide pair to a predetermined endogenous DNA target sequence in a target cell by homologous pairing, particularly for altering an endogenous DNA sequence, such as a chromosomal DNA sequence, typically by targeted homologous recombination In certain embodiments, the invention relates to methods for targeting an exogenous polynucleotide having a linked chemical substituent to a predetermined endogenous DNA sequence in a metabolically active target cell, generating a DNA sequence-specific targeting of one or more chemical substituents in an intact nucleus of a metabolically active living target cell, generally for purposes of altering a predetermined endogenous DNA sequence in the cell The invention also relates to compositions and formulations that contain exogenous targeting polynucleotides, complementary pairs of exogenous targeting polynucleotides, chemical substituents of such polynucleotides, and recombinase proteins, including recombinosome proteins and other targeting proteins, used in the methods of the invention
BACKGROUND
Homologous recombination (or general recombination) is defined as the exchange of homologous segments anywhere along a length of two DNA molecules An essential feature of general recombination is that the enzymes responsible for the recombination event can presumably use any pair of homologous sequences as substrates, although some types of sequence may be favored over others Both genetic and cytological studies have indicated that such a crossing-over process occurs between pairs of homologous chromosomes during meiosis in higher organisms
Alternatively, in site-specific recombination, exchange occurs at a specific site, as in the integration of phage 8 into the E coli chromosome and the excision of 8 DNA from it Site-specific recombination involves specific sequences of the phage DNA and bacterial DNA Within these sequences there is only a short stretch of homology necessary for the recombination event, but not sufficient for it The enzymes involved in this event generally cannot recombine other pairs of homologous (or nonhomologous) sequences, but act specifically on the particular phage and bacterial sequences
Although both site-specific recombination and homologous recombination are useful mechanisms for genetic engineering of DNA sequences, targeted homologous recombination provides a basis for targeting and altering essentially any desired sequence in a duplex DNA molecule, such as targeting a DNA sequence in a chromosome for replacement by another sequence Site-specific recombination hag been proposed as one method to integrate transfected DNA at chromosomal locations having specific recognition sites (O'Gorman et al (1991) Science 251 1351 , Onouchi et al (1991 ) Nucleic Acids Res 19 6373) Unfortunately, since this approach requires the presence of specific target sequences and recombinases, its utility for targeting recombination events at any particular chromosomal location is severely limited in comparison to targeted general recombination
For these reasons and others, targeted homologous recombination has been proposed for treating human genetic diseases Human genetic diseases include (1) classical human genetic diseases wherein a disease allele having a mutant genetic lesion is inherited from a parent (e g , adenosine deaminase deficiency, sickle cell anemia, thalassemias), (2) complex genetic diseases like cancer, where the pathological state generally results from one or more specific inherited or acquired mutations, and (3) acquired genetic disease, such as an integrated provirus (e g , hepatitis B virus) However, current methods of targeted homologous recombination are inefficient and produce desired homologous recombinants only rarely, necessitating complex cell selection schemes to identify and isolate correctly targeted recombinants
A primary step in homologous recombination is DNA strand exchange, which involves a pairing of a DNA duplex with at least one DNA strand containing a complementary sequence to form an intermediate recombination structure containing heteroduplex DNA (see. Radding, C M (1982) Ann Rev Genet 16 405, U S Patent 4,888,274) The heteroduplex DNA may take several forms, including a three DNA strand containing triplex form wherein a single complementary strand invades the DNA duplex (Hsieh et al (1990) Genes and Development 4 1951 , Rao et al , (1991 ) PNAS 88 2984)) and, when two complementary DNA strands pair with a DNA duplex, a classical Hol day recombination joint or chi structure (Holhday, R (1964) Genet Res 5 282) may form, or a double-D loop ("Diagnostic Applications of Double-D Loop Formation" U S S N 07/755,462, filed 4 September 1991 , which is incorporated herein by reference) Once formed, a heteroduplex structure may be resolved by strand breakage and exchange, so that all or a portion of an invading DNA strand is spliced into a recipient DNA duplex, adding or replacing a segment of the recipient DNA duplex Alternatively, a heteroduplex structure may result in gene conversion, wherein a sequence of an invading strand is transferred to a recipient DNA duplex by repair of mismatched bases using the invading strand as a template (Genes. 3rd Ed (1987) Lewin, B , John Wiley New York, NY, Lopez et al (1987) Nucleic Acids Res 15 5643) Whether by the mechanism of breakage and rejoining or by the mechanιsm(s) of gene conversion, formation of heteroduplex DNA at homologously paired joints can serve to transfer genetic sequence information from one DNA molecule to another
The ability of homologous recombination (gene conversion and classical strand breakage/rejoining) to transfer genetic sequence information between DNA molecules makes targeted homologous recombination a powerful method in genetic engineering and gene manipulation
The ability of mammalian and human cells to incorporate exogenous genetic material into genes residing on chromosomes has demonstrated that these cells have the general enzymatic machinery for carrying out homologous recombination required between resident and introduced sequences These targeted recombination events can be used to correct mutations at known sites, replace genes or gene segments with defective ones, or introduce foreign genes into cells The efficiency of such gene targeting techniques is related to several parameters the efficiency of DNA delivery into cells, the type of DNA packaging (if any) and the size and conformation of the incoming DNA, the length and position of regions homologous to the target site (all these parameters also likely affect the ability of the incoming homologous DNA sequences to survive intracellular nuclease attack), the efficiency of recombination at particular chromosomal sites and whether recombinant events are homologous or nonhomologous Over the past 10 years or so, several methods have been developed to introduce DNA into mammalian cells direct needle microinjection, transfection, electroporation, electroincorporation, retroviruses, adenoviruses, adeno-associated viruses, Herpes viruses, and other viral packaging and delivery systems, polyamidoamine dendimers, liposomes, and most recently techniques using DNA-coated microprojectiles delivered with a gene gun (called a biolistics device), or narrow-beam lasers (laser-poration) The processes associated with some types of gene transfer have been shown to be both mutagenic and carcinogenic (Bardwell, (1989) Mutagenesis 4 245), and these possibilities must be considered in choosing a transfection approach
The choice of a particular DNA transfection procedure depends upon its availability to the researcher, the technique's efficiency with the particular chosen target cell type, and the researchers concerns about the potential for generating unwanted genome mutations For example, refroviral integration requires dividing cells, always results in nonhomologous recombination events, and refroviral insertion within a coding sequence of nonhomologous (i e , non-targeted) gene could cause cell mutation, by inactivating the gene's coding sequence (Fπedmann, (1989) Science 244 1275) Newer retroviral- based DNA delivery systems are being developed using defective retroviruses However, these disabled viruses must be packaged using helper systems, are often obtained at low titer, and recombination is still not site-specific, thus recombination between endogenous cellular retrovirus sequences and disabled virus sequences could still produce wild-type retrovirus capable of causing gene mutation Adeno- or polyoma virus based delivery systems appear very promising (Samulski et al . (1991) EMBO J 10 2941. Gareis et al . (1991) Cell Molec Biol 37 191, Rosenfeld et al (1992) Ceil 68 143) although they still require specific cell membrane recognition and binding characteristics for target cell entry Liposomes often show a narrow spectrum of cell specificities, and when DNA is coated externally on to them, the DNA is often sensitive to cellular nucleases Newer polycationic pospermines compounds exhibit broad cell ranges (Behr et al , (1989) Proc Natl Acad Sci USA 86 6982) and DNA is coated by these compounds In addition, a combination of neutral and cationic lipid has been shown to be highly efficient at transfection of animal cells and showed a broad spectrum of effectiveness in a variety of cell lines (Rose et al , (1991) BioTechniques 10 520) Galactosylated bis- acπdine has also been described as a carrier for delivery of polynucleotides to liver cells (Haensler JL and Szoka FC (1992), Abstract V211 in J Cell Biochem Supplement 16F, April 3-16, 1992, incorporated herein by reference) Electroporation also appears to be applicable to most cell types The efficiency of this procedure for a specific gene is variable and can range from about one event per 3 x 104 transfected cells (Thomas and Capecchi, (1987) CeN 51 503) to between one in 107 and 108 cells receiving the exogenous DNA (Koller and Smithies, (1989) Proc Natl Acad Sci (U S A ) 86 8932) Microinjection of exogenous DNA into the nucleus has been reported to result in a high frequency of stable transfected cells Zimmer and Gruss (Zimmer and Gruss (1989) Nature 338 150) have reported that for the mouse hoxl 1 gene, 1 per 150 microinjected cells showed a stable homologous site specific alteration
Several methods have been developed to detect and/or select for targeted site-specific recombinants between vector DNA and the target homologous chromosomal sequence (see, Capecchi, (1989) Science 244 1288 for review) Cells which exhibit a specific phenotype after site-specific recombination, such as occurs with alteration of the hprt gene, can be obtained by direct selection on the appropriate growth medium Alternatively, a selective marker sequence such as neo can be incorporated into a vector under promoter control, and successful transfection can be scored by selecting G418r cells followed by PCR to determine whether neo is at the targeted site (Joyner et al , (1989) Nature 338 153) A positive-negative selection (PNS) procedure using both neo and HSV-tk genes allows selection for transfectants and against nonhomologous recombination events, and significantly enriched for desired disruption events at several different mouse genes (Mansour et al , (1988) Nature 336 348) This procedure has the advantage that the method does not require that the targeted gene be transcribed If the targeted gene is transcribed, a promoter-less marker gene can be incorporated into the targeting construct so that the gene becomes activated after homologous recombination with the target site (Jasin and Berg, (1988) Genes and Development 2 1353,
Doetschman et al (1988) Proc Natl Acad Sci (U S A ) 85 8583. Donni et al . (1989) Science 243 1357, Itzhaki and Porter, (1991) Nucl Acids Res 19 3835) Recombinant products produced using vectors with selectable markers often continue to retain these markers as foreign genetic material at the site of transfection, although loss does occur Valancius and Smithies (Valancius and Smithies, (1991) Molec Cellular Biol 11 1402) have described an "in-out" targeting procedure that allowed a subtle 4-bp insertion modification of a mouse hprt target gene The resulting transfectant contained only the desired modified gene sequence and no selectable marker remained after the "out" recombination step Cotransformation of cells with two different vectors, one vector contained a selectable gene and the other used for gene disruption, increases the efficiency of isolating a specific targeting reaction (Reid et al , (1991 ) Molec Cellular Biol 11 2769) among selected cells that are subsequently scored for stable recombinants
Unfortunately, exogenous sequences transferred into eukaryotic cells undergo homologous recombination with homologous endogenous sequences only at very low frequencies, and are so inefficiently recombined that large numbers of cells must be transfected, selected, and screened in order to generate a desired correctly targeted homologous recombinant (Kucherlapati et al (1984) Proc Natl Acad Sci (U S A ) 81 3153, Smithies, 0 (1985) Nature 317 230, Song et al (1987) Proc Natl Acad Sci (U S A ) 84 6820, Doetschman et al (1987) Nature 330 576, Kim and Smithies (1988) Nucleic Acids Res 16 8887, Doetschman et al (1988) op cιt . Koller and Smithies (1989) op cit . Sheselv et al (1991) Proc Natl Acad Sci (U S A ) 88 4294, Kim et al (1991) Gene 103 227, which are incorporated herein by reference)
Koller et al (1991 ) Proc Natl Acad Sci (U S A ). 88 10730 and Snouwaert et al (1992) Science 257 1083, have described targeting of the mouse cystic fibrosis transmembrane regulator (CFTR) gene for the purpose of inactivating, rather than correcting, a munne CFTR allele Koller et al employed a large (7 8kb) homology region in the double-stranded DNA targeting construct, but nonetheless reported a low frequency for correct targeting (only 1 of 2500 G418-resιstant cells were correctly targeted) Thus, even targeting constructs having lone homology regions are inefficiently targeted
Several proteins or purified extracts having the property of promoting homologous recombination (i e , recombinase activity) have been identified in prokaryotes and eukaryotes (Cox and Lehman (1987) Ann Rev Biochem 56 229, Radding, C M (1982) op at . Madiraju et al (1988) Proc Natl Acad Sci (U S A ) 85 6592, McCarthy et al (1988) Proc Natl Acad Sci (U S A ) 85 5854, Lopez et al (1987) op cit . which are incorporated herein by reference) These general recombinases presumably promote one or more steps in the formation of homologously-paired intermediates, strand-exchange, gene conversion, and/or other steps in the process of homologous recombination The frequency of homologous recombination in prokaryotes is significantly enhanced by the presence of recombinase activities Several purified proteins catalyze homologous pairing and/or strand exchange m vitro, including E coli recA protein, the T4 uvsX protein, and the red protein from Ustilago maydis Recombinases, like the recA protein of E coli are proteins which promote strand pairing and exchange The most studied recombinase to date has been the recA recombinase of E coli, which is involved in homology search and strand exchange reactions (see. Cox and Lehman (1987) op cit ) RecA is required for induction of the SOS repair response, DNA repair, and efficient genetic recombination in E coli RecA can catalyze homologous pairing of a linear duplex DNA and a homologous single strand DNA in vitro In contrast to site-specific recombinases, proteins like recA which are involved in general recombination recognize and promote pairing of DNA structures on the basis of shared homology, as has been shown by several in vitro experiments (Hsieh and Cameπni- Otero (1989) J Biol Chem 264 5089, Howard-Flanders et al (1984) Nature 309 215, Stasiak et al (1984) Cold Spring Harbor Svmp Quant Biol 49 561 , Register et al (1987) J Biol Chem 262 12812) Several investigators have used recA protein jn vitro to promote homologously paired triplex DNA (Cheng et al (1988) J Biol Chem 263 15110. Ferπn and Cameπni-Otero (1991 ) Science 354 1494, Ramdas et al (1989) J Biol Chem 264 11395, Strobel et al (1991) Science 254 1639, Hsieh et al (1990) op cit . Riqas et al (1986) Proc Natl Acad Sci (U S A ) 83 9591 , and Cameπni-Otero et al U S 7,611,268 (available from Derwent), which are incorporated herein by reference) Unfortunately many important genetic engineering manipulations involving homologous recombination, such as using homologous recombination to alter endogenous DNA sequences in a living cell, cannot be done jn vitro Further, gene therapy requires highly efficient homologous recombination of targeting vectors with predetermined endogenous target sequences, since selectable marker selection schemes, such as those currently available in the art, are not usually practicable in human beings
Thus, there exists a need in the art for methods of efficiently altering predetermined endogenous genetic sequences by homologous pairing and homologous recombination m vivo by introducing one or more exogenous targeting polynucleotιde(s) that efficiently and specifically homologously pair with a predetermined endogenous DNA sequence There exists a need in the art for high-efficiency gene targeting, so as to avoid complex in vitro selection protocols (e g , neo gene selection with G418), which are of limited utility for in vivo gene therapy on affected individuals
SUMMARY OF THE INVENTION
It is an object of the present invention to provide methods for targeting an exogenous polynucleotide to a predetermined endogenous DNA target sequence in a target cell with high efficiency and with sequence specificity Exogenous polynucleotides, are localized (or targeted) to one or more predetermined DNA target sequence(s) by homologous pairing in vivo Such targeted homologous pairing of exogenous polynucleotides to endogenous DNA sequences in vivo may be used (1 ) to target chemical substituents in a sequence-specific manner m vivo. (2) to correct or to generate genetic mutations in endogenous DNA sequences by homologous recombination and/or gene conversion, (3) to produce homologously targeted transgenic animals and plants at high efficiency, and (4) in other applications (e g , targeted drug delivery) based on in vivo homologous pairing Some embodiments of the invention employ targeted exogenous polynucleotides to correct endogenous mutant gene alleles in human cells, the invention provides methods and compositions for correcting disease alleles involved in producing human genetic diseases, such as inherited genetic diseases (e g , cystic fibrosis) and neoplasia (e g , neoplasms induced by somatic mutation of an oncogene or tumor suppressor gene, such as p53, or viral genes associated with neoplasia, such as HBV genes)
In one embodiment, at least one exogenous polynucleotide is targeted to a predetermined endogenous DNA sequence and alters the endogenous DNA sequence, such as a chromosomal DNA sequence, typically by targeted homologous recombination within and/or flanking the predetermined endogenous DNA sequence Generally, two complementary exogenous polynucleotides are used for targeting an endogenous DNA sequence Typically, the targeting polynucleotιde(s) are introduced simultaneously or contemporaneously with one or more recombinase species Alternatively, one or more recombinase species may be induced or produced in vivo, for example by expression of a heterologous expression cassette in a cell containing the preselected target DNA sequence
It is another object of the invention to provide methods whereby at least one exogenous polynucleotide containing a chemical substituent can be targeted to a predetermined endogenous DNA sequence in a metabolically-active or intact living target cell, permitting sequence-specific targeting of chemical substituents such as, for example cross-linking agents, metal chelates (e g , iron/EDTA chelate for iron catalyzed cleavage), topoisomerases, endonucleases, exonucleases, ligases, phosphodiesterases, photodynamic porphyπns, free-radical generating drugs, chemotherapeutic drugs (e g , adriamycin, doxirubicin), intercalating agents, base-modification agents, immunoglobulin chains, oligonucleotides, and other substituents The methods of the invention can be used to target such a chemical substituent to a predetermined DNA sequence by homologous pairing for various applications, for example producing sequence-specific strand scιssιon(s), producing sequence-specific chemical modifications (e g , base methylation, strand cross-linking), producing sequence-specific localization of polypeptides (e g , topoisomerases, he cases, proteases), producing sequence-specific localization of polynucleotides (e g , loading sites for transcription factors and/or RNA polymerase), and other applications
It is another object of the present invention to provide methods for correcting a genetic mutation in an endogenous DNA target sequence, such as a sequence encoding an RNA or a protein For example, the invention can be used to correct genetic mutations, such as base substitutions, additions, and/or deletions, by converting a mutant DNA sequence that encodes a non-functional, dysfunctional, and/or truncated polypeptide into a corrected DNA sequence that encodes a functional polypeptide (e g , has a biological activity such as an enzymatic activity, hormone function, or other biological property) The methods and compositions of the invention may also be used to correct genetic mutations or dysfunctional alleles with genetic lesions in non-coding sequences (e g , promoters, enhancers, silencers, origins of replication, splicing signals) In contradistinction, the invention also can be used to target DNA sequences for inactivating gene expression, a targeting polynucleotide can be employed to make a targeted base substitution, addition, and/or deletion in a structural or regulatory endogenous DNA sequence to alter expression of one or more genes, typically by knocking out at least one allele of a gene (i e , making a mutant, nonfunctional allele) The invention can also be used to correct disease alleles, such as a human or non-human animal CFTR gene allele associated with cystic fibrosis, by producing a targeted alteration in the disease allele to correct a disease-causing lesion (e g , a deletion)
It is a further object of the invention to provide methods and compositions for high-efficiency gene targeting of human genetic disease alleles, such as a CFTR allele associated with cystic fibrosis or an LDL receptor allele associated with familial hypercholesterolemia In one aspect of the invention, targeting polynucleotides having at least one associated recombinase are targeted to cells in vivo (i e , in an intact animal) by exploiting the advantages of a receptor-mediated uptake mechanism, such as an asialoglycoprotein receptor-mediated uptake process In this variation, a targeting polynucleotide is associated with a recombinase and a cell-uptake component which enhances the uptake of the targeting polynucleotide- recombinase into cells of at least one cell type in an intact individual For example, but not limitation, a cell-uptake component typically consists of (I) a galactose-terminal (asialo-) glycoprotein (e g , asialoorosomucoid) capable of being recognized and internalized by specialized receptors (asialoglycoprotein receptors) on hepatocvtes in vivo, and (2) a polycation, such as poly-L-lysine, which binds to the targeting polynucleotide, usually by electrostatic interaction Typically, the targeting polynucleotide is coated with recombinase and cell-uptake component simultaneously so that both recombinase and cell-uptake component bind to the targeting polynucleotide, alternatively, a targeting polynucleotide can be coated with recombinase prior to incubation with a cell-uptake component, alternatively the targeting polynucleotide can be coated with the cell-uptake component and introduced into cells contemporaneously with a separately delivered recombinase (e g , by targeted liposomes containing one or more recombinase)
The invention also provides methods and compositions for diagnosis, treatment and prophylaxis of genetic diseases of animals, particularly mammals, wherein a recombinase and a targeting polynucleotide are used to produce a targeted sequence modification in a disease allele of an endogenous gene The invention may also be used to produce targeted sequence modιficatιon(s) in a non-human animal, particularly a non-human mammal such as a mouse, which create(s) a disease allele in a non-human animal Sequence-modified non-human animals harboring such a disease allele may provide useful models of human and veterinary dιsease(s) Alternatively, the methods and compositions of the invention can be used to provide nonhuman animals having homologously- targeted human disease alleles integrated into a non-human genome, such non-human animals may provide useful experimental models of human or other animal genetic disease, including neoplastic and other pathogenic diseases
It is also an object of the invention to provide methods and compositions for recombinase- enhanced positioning of a targeting polynucleotide to a homologous sequence in an endogenous chromosome to form a stable multistrand complex, and thereby alter expression of a predetermined gene sequence by interfering with transcription of sequence(s) adjacent to the multistrand complex Recombιnase(s) are used to ensure correct homologous pairing and formation of a stable multistrand complex, which may include a double-D loop structure For example, a targeting polynucleotide coated with a recombinase may homologously pair with an endogenous chromosomal sequence in a structural or regulatory sequence of a gene and form a stable multistrand complex which may (1 ) constitute a significant physical or chemical obstacle to formation of or procession of an active transcriptional complex comprising at least an RNA polymerase, or (2) alter the local chromatin structure so as to alter the transcription rate of gene sequences within about 1 to 500 kilobases of the multistrand complex
It is another object of the invention to provide methods and compositions for treating or preventing acquired human and animal diseases, particularly parasitic or viral diseases, such as human hepatitis B virus (HBV) hepatitis, by targeting viral gene sequences with a recombmase-associated targeting polynucleotide and thereby inactivating said viral gene sequences and inhibiting viral-induced pathology
It is a further object of the invention to provide compositions that contain exogenous targeting polynucleotides, complementary pairs of targeting polynucleotides, chemical substituents of such polynucleotides, and recombinase proteins used in the methods of the invention Such compositions may include a targeting or cell-uptake components to facilitate intracellular uptake of a targeting polynucleotide, especially for jn vivo gene therapy and gene modification
In accordance with the above objects, the present invention provides methods for targeting and altering, by homologous recombination, a pre-selected target nucleic acid sequence in a cell to make a targeted sequence modification The methods comprise introducing into at least one cell at least one recombinase and at least two single-stranded targeting polynucleotides which are substantially complementary to each other and comprise a homology clamp that substantially corresponds to or is substantially complementary to a preselected target nucleic acid sequence
In an additional aspect, the invention provides compositions for producing targeted modifications of target sequences, including disease alleles, comprising two substantially complementary single-stranded targeting polynucleotides and at least one recombinase
BRIEF DESCRIPTION OF THE DRAWINGS
Fig 1 Homologous targeting of recA-coated chromosome 1 alpha-satellite polynucleotides in metabolically active cell nuclei The homologously targeted biotinylated polynucleotides were visualized by addition of FITC-avidin followed by washing to remove unbound FITC Signals were visualized using a Zeiss Confocal Laser Scanning Microscope (CLSM-10) with 488 nm argon laser beam illumination for FITC-DNA detection Top left -localized FITC-DNA signals in the cell nucleus Lower left -enhanced image of FITC-DNA signals in the cell nucleus Upper right - image of FITC- DNA signals overlaid on the phase image of nucleus Lower right - phase image of the center of the cell nucleus showing nucleoli Note all images except lower right were photographed at the same focus level (focus unchanged between these photos)
Figs 2A, 2B, 2C, 2D, 2E, 2F, 2G, 2H, 21, 2J, 2K, and 2L RecA protein-mediated native FISH in metabolically active cell nuclei Hep-2 cell nuclei from cells encapsulated in agarose were incubated with RecA-coated biotinylated p53 DNA (A-l) or RecA-coated biotinylated chromosome 1 satellite III DNA probes (K-L) Panels B-l show FISH signals in digital images from serial CLSM optical sections of FITC-labeled p53 probe DNA incubated in metabolically active Hep-2 nuclei The phase image of a representative nucleous in shown in Panel A and was sectioned by CLSM Digital images in Panels B-H were serially overlaid upon one another to produce the composite digital image shown in Panel I containing all three FITC labeled p53 FISH signals The effect of cssDNA probe concentration and RecA protein on efficiency of native dsDNA hybridization in metabolically active nuclei is shown in Panel J The percentage of labeled RecA coated or uncoated p53 cssDNA is shown as a function of the amount of p53 DNA probe per hybridization reaction Closed circles show hybridization reactions with RecA-coated p53 cssDNA probe, open triangles show control reactions without RecA protein coating of p53 cssDNA probe Panel K shows the FISH digital image in Panel L overlaid onto the phase image
Fig 3 Genetic map of mammalian expression lacZ plasmid pMCIIacXpA with an 11 base insertion in the Xba linker site Fig 4 Genetic map of the mammalian expression lacZ plasmid pMCIIacpA, with an insertion mutation
Fig 5 PCR products and primers from the lacZ (β-galactosidase) gene sequence The location of the 11 bp Xba linker is shown
Fig 6 Tests for alteration of an insertion mutation in the lacZ gene of a eukaryotic expression vector NIH 3T3 cells were needle microinjected with five types of plasmids Two plasmids contained a wild-type β-galactosidase gene (pMCI Iacpa or pSV-β-gal [Promega]), a plasmid with a mutant β-gal gene (pMCIIacXpa), pMCI IacXpa plasmid incubated with a protein uncoated wild-type 276-mer DNA, or pMCIIacXpa plasmid reacted and D-looped with RecA-coated wild-type 276-mer DNA The wild-type 276-mer DNA was heat denatured and either coated or not coated with RecA protein in a standard RecA protein coating reaction protocol (Sena and Zarling, supra) Following a 10-mιn RecA coating reaction, the RecA-coated complementary single-stranded 276-mers were incubated at 37°C for 60 mm with the mutant target plasmid to allow hybrid formation A 60 mm incubation of the mutant target plasmid DNA with uncoated complementary single-stranded normal wild-type 276-mers was carried out as a control and hybrids were not formed The β-galactosidase activity in needle microinjected cells using the wild-type plasmids is shown for comparison On average, about 50% of the total microinjected cells survived The numbers of surviving cells scoring blue with the mutant plasmid hybridized with RecA-coated CSS DNA and reacted with non-RecA-coated CSS DNA samples (3, 4 and 5) were compared with fourfold P2 tests The frequency of corrected blue cells in the RecA-coated CSS DNA samples (Sample 5, 6 out of 168) is significantly higher than that of either Sample 3 or Sample 4 The frequency of corrected RecA-coated CSS DNA probe target hybrids blue cells in Sample 5 is significantly higher than that of Sample 4 at the 5% significance level (P2 = 3 76 > P2ooδ) The frequency of corrected blue cells in Sample 5 containing RecA-coated CSS DNA probe target hybrids is significantly higher than that of Sample 3 at the 1 % significance level (P2 = 6 28 > P2 001) When Samples 3 and 4 are combined and compared with Sample 5, the frequency of corrected blue cells in Sample 5 is significantly higher than that of the combined sample at the 0 1% signficance level (P2 = 9 99 > P2 00oι)
Fig 7A Southern hybridization analysis of the 687-bp fragment amplified from genomic DNA Electrophoretic migration of a 687-bp DNA fragment generated with primers CF1 and CF6 from genomic DNA of 3CFTE29o-cells which were capillary needle-micromjected with the 491 -nucleotide DNA fragment in the presence of recA protein (lane 2) or transfected as a protem-DNA-lipid complex where the 491 -nucleotide fragments were coated with recA protein (+, lane 3) The control DNA was amplified from nontransfected 3CFTE29o-cultures (lane 1 ) Fig 7B Autoradiographic analysis of DNA transferred to Gene Screen Plus filters and hybridized with a 32P-labeled oligonucleotide specific for normal exon 10 sequences in the region of the ΔF508 mutation Cells transfected by micro-injection or protein-lipid-DNA complexes both were positive for homologous targeting, whereas control cells were not
Fig 8A Analysis of DNA from cells electroporated or transfected with CSS DNA encapsulated in a protein- pid complex Allele-specific PCR amplification of the 687/684 bp DNA fragment amplified in the first round with primers CF1 and oligo N (N) or oligo ΔF (ΔF) Ethidium bromide-stained 300 bp DNA fragment separated by electrophoresis in a 1% agarose gel The DNA in each lane is as follows lane 1 , 100-bp marker DNA, lane 2, control 16HBE14o-cell DNA amplified with the CF1/N primer pair, lane 3, nontransfected ECFTE29o-cell DNA amplified with CF1/N primers, lane 4, nontransfected
ECFTE29o-cell DNA amplified with CF1/ΔF primers, lane 5, DNA from ECFTE29o-cells electroporated with recA-coated 49l-nucleotιde fragments and amplified with CF1/N primers, lane 6, DNA from ECFTE29o-cells transfected with recA-coated 491 -nucleotide fragment encapsulated in a protein-hpid complex and amplified with CF1/N primers
Fig 8B Autoradiographic analysis of the DNA in Fig HA transferred to Gene Screen Plus filters and hybridized with 32P-labeled oligo N probe Samples in lanes 1-5 for the autoradiographic analysis are equivalent to samples in lanes 2-6 in Fig IIA
Fig 9 PCR analysis of 3CFTE29o-genomιc DNA reconstructed with the addition of 2 x 105 copies of recA-coated 491 -nucleotide CSS DNA fragments per microgram of genomic DNA This number of CSS DNA fragments represents the total number of DNA copies microinjected into cells and tests whether the 491 -nucleotide fragment can act as a primer for the 687/684-bp fragment amplification DNA was amplified as described in Fig 8A When the second round of amplification was conducted with CF1 and oligo N primers (lane 2), the 300-bp DNA band was not detected when aliquots of the amplification reaction were separated electrophoretically Amplification of the ECFTE29o/491 bp DNA fragment with the CF1 /oligo ΔF primer pair produced a 299-bp DNA product (lane I) Marker DNA is in lane 3
Figure 10 depicts the scheme for the recombination assay used in Example 4
Fig 11 shows RecA mediated cssDNA targeting to dsDNA with deletions produces a mixed population of probe target hybrids The biotinylated cssDNA probes were denatured and coated with RecA at 37°C as described in Material The reaction mixture was incubated for 60 minutes at 37°C All reactions were stopped by deprotemization with 1 2% SDS and separated by electrophoresis on a 20 cm X 25 cm 1% agarose gel The gel was run overnight at 30V then blotted onto a positively charged Tropilon Plus (TROPIX) membrane The DNA was monitored for the presence of unhybridized probe or probe target hybrids using an alkaline phosphatase based chemiluminescent detection of biotin When the membranes were exposed to X-ray film and developed, it is evident that cssDNA probes will hybridize to dsDNA targets which are completely homologous, as well as dsDNA targets which contain a deletion (lanes 3 and 6, respectively) RecA mediated cssDNA targeting to completely homologous dsDNA (pRD 0) forms a probe target hybrid whose electrophoretic mobility is comparable to the electrophoretic mobility of completely relaxed Form I DNA, which is similar to the mobility of Form II DNA (lanes 3, 8, and 10), referred to as the rl* hybrid RecA mediated hybridization of cssDNA to dsDNA containing a 59 base pair deletion (pRD 59), a probe target hybrid that migrates to a position similar to Form I DNA (lane 6), is referred to as the I* hybrid
Fig 12 shows data for the enhanced homologous recombination (EHR) of cssDNA probe target hybrids in E coli, as per Example 4 The homologously targeted probe target hybrids have enhanced homologous recombination frequencies in recombination proficient cells cssDNA probe target hybrids were formed as described in the legend of Figure 11 and were introduced into RecA+ and RecA-E coli as in described Figure 12 The molar ratio of cssDNA probe target in the in vitro targeting reaction varied from 1 1 to 1 5 6 The % recombinant/total colonies is the percentage of blue colonies in the total population of ampicillin-resistant colonies Groups with 0% recombinants did not produce any blue colonies in at least 105 plated colonies Plasmid DNA was isolated from blue colonies that were serially propagated for three generations to determine if homologous recombination stably occurred in the lacZ gene
Fig 13 shows double D-loop hybrids with internal homology clamps A) Duplex target DNA (thin line) is completely homologous to the cssDNA probe (thick) and each probe strand can pair with its complementary strand in the target B) Duplex target has a deletion with respect to the cssDNA probe The deleted region is indicated with a dashed line The region of the cssDNA probes homologous to the deleted region in the target can re-pair with each other forming a stable hybrid complex C)
Duplex target has an insertion (dashed line) with respect to the cssDNA probe Structures on the left show the re-annealing of cssDNA probe or target strands to form internal homology clamps Structures on the right show the presence of unpaired regions in comparable single D-loop hybrids
Figs 14A and 14B Figure 14A depicts the Maps of Plasmids pRD 0 and pRD 59 Relative positions of cssDNA probes IP290 and CP443, PCR primers 1 A and 4B, restriction endonuclease sites EcoRI, Seal, and Dral are indicated The alpha peptide sequence of the LacZ gene is indicated Note the deletion ()) in pRD 59 is approximately equidistant from the ends of primers 1 A and 4B Figure 14B) Time course for cssDNA probe target hybrid formation with linear dsDNA targets Biotinylated, RecA coated cssDNA probe IP290 was hybridized as described to Seal-digested plasmids pRD 0 and pRD 59 carrying 0 or 59 bp deletion, respectively at the EcoRI site in pRD 0 Probe IP290 is completely homologous to pRD 0, but has a 59 bp insertion with respect to pRD 59
Fig 15 depicts the formation of cssDNA probe target hybrids formed with linear dsDNA targets containing small deletions A) Plasmid constructs and probes used in this study A series of plasmids with defined deletions were constructed from the EcoRI site of pRD 0 (pbluescπptllSK+ (Stratagene) as described in Example 5 Each plasmid is named for the size of the deletion, as indicated on the left A series of cssDNA probes were labelled and constructed by PCR from various primers which flank the deleted region Probes were made from either pRD 0 or the deleted plasmids and named for the size of the probe when made from pRD 0 (2960 bp) For example, p527 is 527 bp long When the cssDNA probes are produced from pRD 0 and targeted to plasmids containing deletions, the probe is called IP527 to indicate that the insertion probe (IP) has an insertion with respect to the target When the probe is made from one of the targets with a deletion and then, targeted to pRD 0, the probe is called DP527 to indicate that the deletion probe (DP) has a deletion with respect to pRD 0 Control probe CP443 is made from a region of pRD 0 that does not contain any insertions or deletions The limits of the deleted regions in the plasmid DNA target are indicated by dashed lines and the size limits of cssDNA probes are indicated by solid lines B) Biotinylated cssDNA probes IP527, IP407, and CP443 were coated with RecA protein and hybridized at 37°C to a series of linear duplex DNA targets containing deletions ranging in size from 0 to 447 bp The products of the targeting reaction were deprotemized and separated on a 1 % TAE-agarose gel and then transferred to nylon membranes as described in Example 5 Biotinylated DNA was detected with a chemiluminescent substrate as described The extent of hybrid product formation of Form III DNA targets was determined by densitometry of the autoradiographs The relative amount of hybrid formed between RecA coated cssDNA probes IP527 and IP407 is shown in (B) Error bars are indicated The amount of probe target hybrids formed with each target DNA was normalized by the amount of probe.target hybrids formed with control probe CP443 which hybridizes to the target located in a region which is a significant distance away from the deletion site Examples of the cssDNA probe target hybrid formed with linear targets are shown in the autoradiogram (C) In Fig 15(D) the difference in the percent hybrid formation between cssDNA probes IP527 and IP407 are plotted from the data shown in (B)
Fig 16 depicts that insertions and deletions have the same effect on the relative efficiency of probe target hybrid formation RecA-coated cssDNA probes IP215 made from pRD 0 was targeted to Seal-digests of plasmids pRD 0, pRD 8, pRD 25, and pRD 59 and compared to similar reactions of DP215 cssDNA probes made from pRD 0, pRD 8, pRD 25, and pRD 59 and targeted to pRD 0 The effect of insertions in the cssDNA probe (dark line) is compared with deletions in the cssDNA probe (shaded line) of the same size The relative level of hybrid formation for each cssDNA probe with a heterologous target is normalized by the level of hybridization with the homologous target, respectively The data represents an average of three experiments Error bars are indicated
Fιgs 17A, 17B and 17C Figure 17A depict the formation of stable double-D-Loop hybrids in linear dsDNA targets containing large deletions Biotinylated cssDNA probe IP1246 was coated with RecA protein and targeted to Seal digests of the indicated plasmids as described herein The relative amount of hybrid formation formed between RecA-coated cssDNA probes and plasmids with deletions ranging from 0-967 bp was normalized to the amount of probe target hybrids formed with control probe CP443 Autoradiograph (17A) shows the biotinylated cssDNA probes or probe target hybrids The position of the untargeted Seal-digested (Formlll) marker for each of the plasmids are indicated on the right The relative level of hybrid formation (B) of each of the bands in (A) was normalized to the level of hybrid formation with control cssDNA probe CP443, as described herein The relative position of the cssDNA probes with respect to the position of the deletion in the target DNA is shown in (C)
Figs 18A, 18B, 18C and 18D depict the formation of restriction endonuclease sites in probe target hybrids The probe target hybrids formed between probe IP290 and pRD 0 and pRD 59 targets were deprotemized by extraction with chloroform phenol isoamyl alcohol and chloroform Restriction enzyme treated DNA samples were incubated with EcoRI for three hours before separation on a 1% agarose gel and transferred onto a nylon membrane The ethidium bromide stained DNA of the products of the targeting reactions formed between cssDNA probe IP290 and circular plasmid targets pRD 0 or pRD 59 (A and B) and autoradiographs showing the positions of biotinylated cssDNA probe target hybrids (C and D) are shown The positions of form I and form III markers of pRD 0 are shown on the right The positions of the pRD59 hybrids I* (form I) and rl* (relaxed) are shown on the left
Fig 19 depicts the thermal stability of relaxed and non-relaxed probe target hybrids The RecA mediated cssDNA targeting reaction was performed with the cssDNA probe IP290 and the dsDNA target pRD 59, as described herein The probe target hybrids were deprotemized with 1 2% SDS and then incubated for 5 minutes at the indicated temperatures The thermally melted products were then separated on a 1% agarose gel and blotted onto a positively charged Tropilon membrane Autoradiograph shows the position of biotinylated cssDNA probe target hybrids I* (forml) and rl* (relaxed) as shown on the left
Figs 20A and 20B The organization of the mouse OTC gene Sequence of cssDNA probes and PCR primers used in this study are indicated Sizes of the exons in base pairs are indicated The relative position of PCR primers M9, M8 and M11 are shown B) Map of plasmid pTAOTCI A 250 bp fragment containing the normal OTC exon 4 sequence and surrounding introns were cloned into the EcoRV site of pbluescπpt SK (+) (Stratagene)
Fig 21 Sequence analysis of exon 4 of the mouse OTC gene in founder mice PCR amplification of genomic DNA from tail biopsies of a pool of all of the homozygous (spf-ash/spf-ash) females used as egg donors and each indicated individual founder mice were sequenced using cycle sequencing with the M11 primer (Cyclist kit, Stratagene) The DNA sequence surrounding the spf-ash locus (arrow) in the OTC gene is shown
Fig 22 Germ ne transmission of OTC+ allele corrected by EHR The inheritance patterns of the OTC alleles are depicted Legend indicates the genotype and/or phenotype of the F0, F1 , and F2 mice produced from microinjected zygotes obtained from the cross of homozygous (spf-ash/spf-ash) mutant females and normal males (top) The genotype of F0 and F1 animals were determined by DNA sequencing and the typing of F2 animals as deduced by phenotype Control cross A of (hemizygous spf-ash/Y) mutant F0 male with normal (+/+) females and control cross B of heterozygous (spf-ash/+) F1 females with a normal male are indicated The number below the boxes or circles indicate the total number of mice of each type produced from each cross Total numbers of mice counted are representative of 2-4 litters Mouse #213 and #1014 (noted by arrow) are F1 animals that carry a germhne transmitted gene corrected allele from mosaic HR gene corrected male mouse #16
Fig 23 Germhne transmission of corrected allele of F0 male #16 Pictures of F1 progeny from the cross of mouse #16 with homozygous (spf-ash/spf-ash) females (top) This cross produced several pups with spf-ash mutant phenotypes (middle) and one F1 pup (#1014) with a normal phenotype Three views of mouse #1014 are shown (bottom) All of the F1 animals were two weeks old at the time of photography
DEFINITIONS Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are described For purposes of the present invention, the following terms are defined below
As used herein, the twenty conventional ammo acids and their abbreviations follow conventional usage (Immunology -A Synthesis. 2nd Edition, E S Golub and D R Green, Eds , Sinauer Associates, Sunderland, Massachusetts (1991 ), which is incorporated herein by reference) By Anucleic acιd@, Aoiιgonucleotιde@, and Apolynucleotιde@ or grammatical equivalents herein means at least two nucleotides covalently linked together A nucleic acid of the present invention will generally contain phosphodiester bonds, although in some cases nucleic acid analogs are included that may have alternate backbones, comprising, for example, phosphoramide (Beaucage et al , Tetrahedron 49(10) 1925 (1993) and references therein, Letsmger, J Org Chem 35 3800 (1970), Spπnzl et al , Eur J Biochem 81 579 (1977), Letsmger et al , Nucl Acids Res 14 3487 (1986), Sawai et al, Chem Lett 805 (1984), Letsmger et al , J Am Chem Soc 110 4470 (1988), and Pauwels et al , Chemica Scπpta 26 141 91986)), phosphorothioate, phosphorodithioate, O-methylphophoroamidite linkages (see Eckstein, Oligonucleotides and Analogues A Practical Approach, Oxford University Press), and peptide nucleic acid backbones and linkages (see Egholm, J Am Chem Soc 114 1895 (1992), Meier et al , Chem Int Ed Engl 31 1008 (1992), Nielsen, Nature, 365 566 (1993), Carlsson et al , Nature 380 207 (1996), all of which are incorporated by reference) These modifications of the πbose-phosphate backbone or bases may be done to facilitate the addition of other moieties such as chemical constituents, including 2' O-methyl and 5' modified substituents, as discussed below, or to increase the stability and half-life of such molecules in physiological environments
The nucleic acids may be single stranded or double stranded, as specified, or contain portions of both double stranded or single stranded sequence The nucleic acid may be DNA, both genomic and cDNA, RNA or a hybrid, where the nucleic acid contains any combination of deoxyπbo-and ribonucleotides, and any combination of bases, including uracil, adenme, thymine, cytosme, guanme, inosine, xathanme and hypoxathanme, etc Thus, for example, chimeπc DNA-RNA molecules may be used such as described in Cole-Strauss et al , Science 273 1386 (1996) and Yoon et al , PNAS USA 93 2071 (1996), both of which are hereby incorporated by reference
In general, the targeting polynucleotides may comprise any number of structures, as long as the changes do not substantially effect the functional ability of the targeting polynucleotide to result in homologous recombination For example, recombinase coating of alternate structures should still be able to occur
As used herein, the terms Apredeterm ed endogenous DNA sequence" and "predetermined target sequence" refer to polynucleotide sequences contained in a target cell Such sequences include, for example, chromosomal sequences (e g , structural genes, regulatory sequences including promoters and enhancers, recombinatoπal hotspots, repeat sequences, integrated proviral sequences, hairpins, palindromes), episomal or extrachromosomal sequences (e g , rephcable plasmids or viral or parasitic replication intermediates) including chloroplast and mitochondπal DNA sequences By "predetermined" or Apre-selected@ it is meant that the target sequence may be selected at the discretion of the practitioner on the basis of known or predicted sequence information, and is not constrained to specific sites recognized by certain site-specific recombinases (e g , FLP recombinase or CRE recombinase) In some embodiments, the predetermined endogenous DNA target sequence will be other than a naturally occurring germhne DNA sequence (e g , a transgene, parasitic, mycoplasmal or viral sequence) An exogenous polynucleotide is a polynucleotide which is transferred into a target cell but which has not been replicated in that host cell, for example, a virus genome polynucleotide that enters a cell by fusion of a viπon to the cell is an exogenous polynucleotide, however, replicated copies of the viral polynucleotide subsequently made in the infected cell are endogenous sequences (and may, for example, become integrated into a cell chromosome) Similarly, transgenes which are microinjected or transfected into a cell are exogenous polynucleotides, however integrated and replicated copies of the transgene(s) are endogenous sequences
The term "corresponds to" is used herein to mean that a polynucleotide sequence is homologous (i e , may be similar or identical, not strictly evolutionaπly related) to all or a portion of a reference polynucleotide sequence, or that a polypeptide sequence is identical to a reference polypeptide sequence In contradistinction, the term "complementary to" is used herein to mean that the complementary sequence is homologous to all or a portion of a reference polynucleotide sequence As outlined below, preferably, the homology is at least 50-70%, preferably 85%, and more preferably 95% identical Thus, the complementarity between two single-stranded targeting polynucleotides need not be perfect For illustration, the nucleotide sequence "TATAC" corresponds to a reference sequence "TATAC@ and is perfectly complementary to a reference sequence "GTATA"
The terms "substantially corresponds to" or "substantial identity" or Ahomologous@ as used herein denotes a characteristic of a nucleic acid sequence, wherein a nucleic acid sequence has at least about 60 percent sequence identity as compared to a reference sequence, typically at least about 75 percent sequence identity, and preferably at least about 95 percent sequence identity as compared to a reference sequence The percentage of sequence identity is calculated excluding small deletions or additions which total less than 25 percent of the reference sequence The reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome However, the reference sequence is at least 12-18 nucleotides long, typically at least about 30 nucleotides long, and preferably at least about 50 to 100 nucleotides long ASubstantially complementary" as used herein refers to a sequence that is complementary to a sequence that substantially corresponds to a reference sequence In general, targeting efficiency increases with the length of the targeting polynucleotide portion that is substantially complementary to a reference sequence present in the target DNA "Specific hybridization© is defined herein as the formation of hybrids between a targeting polynucleotide (e g , a polynucleotide of the invention which may include substitutions, deletion, and/or additions as compared to the predetermined target DNA sequence) and a predetermined target DNA, wherein the targeting polynucleotide preferentially hybridizes to the predetermined target DNA such that, for example, at least one discrete band can be identified on a Southern blot of DNA prepared from target cells that contain the target DNA sequence, and/or a targeting polynucleotide in an intact nucleus localizes to a discrete chromosomal location characteristic of a unique or repetitive sequence In some instances, a target sequence may be present in more than one target polynucleotide species (e g , a particular target sequence may occur in multiple members of a gene family or in a known repetitive sequence) It is evident that optimal hybridization conditions will vary depending upon the sequence composition and length(s) of the targeting polynucleotιde(s) and target(s), and the experimental method selected by the practitioner Various guidelines may be used to select appropriate hybridization conditions (see. Maniatis et al , Molecular Cloning A Laboratory Manual (1989), 2nd Ed , Cold Spring Harbor, N Y and Berger and Kimmel, Methods in Enzvmology. Volume 152. Guide to Molecular Cloning Techniques (1987), Academic Press, Inc , San Diego, CA , which are incorporated herein by reference Methods for hybridizing a targeting polynucleotide to a discrete chromosomal location in intact nuclei are provided herein in the Detailed Description
The term "naturally-occurring® as used herein as applied to an object refers to the fact that an object can be found in nature For example, a polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring
A metabolically-active cell is a cell, comprising an intact nucleoid or nucleus, which, when provided nutrients and incubated in an appropriate medium carries out DNA synthesis and RNA for extended periods (e g , at least 12-24 hours) Such metabolically-active cells are typically undifferentiated or differentiated cells capable or incapable of further cell division (although non-dividing cells many undergo nuclear division and chromosomal replication), although stem cells and progenitor cells are also metabolically-active cells
As used herein, the term "disease allele@ refers to an allele of a gene which is capable of producing a recognizable disease A disease allele may be dominant or recessive and may produce disease directly or when present in combination with a specific genetic background or pre-existing pathological condition A disease allele may be present in the gene pool or may be generated de novo in an individual by somatic mutation For example and not limitation, disease to alleles include activated oncogenes, a sickle cell anemia allele, a Tay-Sachs allele, a cystic fibrosis allele, a Lesch-Nyhan allele, a retinoblastoma-susceptibihty allele, a Fabry's disease allele, and a Huntington's chorea allele As used herein, a disease allele encompasses both alleles associated with human diseases and alleles associated with recognized veterinary diseases For example, the ΔF508 CFTR allele in a human disease allele which is associated with cystic fibrosis in North Americans
As used herein, the term "cell-uptake component® refers to an agent which, when bound, either directly or indirectly, to a targeting polynucleotide, enhances the intracellular uptake of the targeting polynucleotide into at least one cell type (e g , hepatocytes) A cell-uptake component may include, but is not limited to, the following specific cell surface receptors such as a galactose-terminal (asialo-) glycoprotem capable of being internalized into hepatocytes via a hepatocyte asialoglycoprotein receptor, a polycation (e g , poly-L-lysme), and/or a protein-hpid complex formed with the targeting polynucleotide Various combinations of the above, as well as alternative cell-uptake components will be apparent to those of skill in the art and are provided in the published literature
DETAILED DESCRIPTION
Generally, the nomenclature used hereafter and the laboratory procedures in cell culture, molecular genetics, and nucleic acid chemistry and hybridization described below are those well known and commonly employed in the art Standard techniques are used for recombinant nucleic acid methods, polynucleotide synthesis, cell culture, and transgenesis Generally enzymatic reactions, oligonucleotide synthesis, oligonucleotide modification, and purification steps are performed according to the manufacturer's specifications The techniques and procedures are generally performed according to conventional methods in the art and various general references which are provided throughout this document The procedures therein are believed to be well known in the art and are provided for the convenience of the reader All the information contained therein is incorporated herein by reference
Transgenic mice are derived according to Hogan, et al , "Manipulating the Mouse Embryo A Laboratory Manual®, Cold Spring Harbor Laboratory (1988) which is incorporated herein by reference
Embryonic stem cells are manipulated according to published procedures (Teratocarcinomas and embryonic stem cells a practical approach, E J Robertson, ed , IRL Press, Washington, D C , 1987, Zjilstra et al , Nature 342 435-438 (1989), and Schwartzberg et al , Science 246 799-803 (1989), each of which is incorporated herein by reference) Zygotes are manipulated according to known procedures, for example see U S Patent No 4,873,191 , Bπnster et al , PNAS 86 7007 (1989), Susuhc et al , J Biol Chem 49 29483 (1995), and Cavard et al , Nucleic Acids Res 16 2099 (1988), hereby incorporated by reference
Oligonucleotides can be synthesized on an Applied Bio Systems oligonucleotide synthesizer according to specifications provided by the manufacturer Modified oligonucleotides and peptide nucleic acids are made as is generally known in the art
The present invention provides methods for targeting and altering, by homologous recombination, a pre-selected target nucleic acid sequence in a target cell, to make targeted sequence modifications The methods comprise introducing into the target cells a recombinase and at least two single-stranded targeting polynucleotides which are substantially complementary to each other The targeting polynucleotides each comprise at least one homology clamp that substantially corresponds to or is substantially complementary to the preselected target nucleic acid sequence The target cells are then screened to identify target cells containing the targeted sequence modification
Targeting Polynucleotides Targeting polynucleotides may be produced by chemical synthesis of oligonucleotides, nick-translation of a double-stranded DNA template, polymerase chain-reaction amplification of a sequence (or ligase chain reaction amplification), purification of prokaryotic or target cloning vectors harboring a sequence of interest (e g , a cloned cDNA or genomic clone, or portion thereof) such as plasmids, phagemids, YACs, cosmids, bacteπophage DNA, other viral DNA or replication intermediates, or purified restriction fragments thereof, as well as other sources of single and double-stranded polynucleotides having a desired nucleotide sequence Targeting polynucleotides are generally ssDNA or dsDNA, most preferably two complementary single-stranded DNAs
Targeting polynucleotides are generally at least about 2 to 100 nucleotides long, preferably at least about 5-to 100 nucleotides long, at least about 250 to 500 nucleotides long, more preferably at least about 500 to 2000 nucleotides long, or longer, however, as the length of a targeting polynucleotide increases beyond about 20,000 to 50,000 to 400,000 nucleotides, the efficiency or transferring an intact targeting polynucleotide into the cell decreases The length of homology may be selected at the discretion of the practitioner on the basis of the sequence composition and complexity of the predetermined endogenous target DNA sequence(s) and guidance provided in the art, which generally indicates that 1 3 to 6 8 kilobase segments of homology are preferred (Hasty et al (1991 ) Molec Cell Biol 11 5586, Shulman et al (1990) Molec Cell Biol 10 4466, which are incorporated herein by reference) Targeting polynucleotides have at least one sequence that substantially corresponds to, or is substantially complementary to, a predetermined endogenous DNA sequence (i e , a DNA sequence of a polynucleotide located in a target cell, such as a chromosomal, mitochondπal, chloroplast, viral, episomal, or mycoplasmal polynucleotide) Such targeting polynucleotide sequences serve as templates for homologous pairing with the predetermined endogenous sequence(s), and are also referred to herein as homology clamps In targeting polynucleotides, such homology clamps are typically located at or near the 5' or 3' end, preferably homology clamps are internally or located at each end of the polynucleotide (Beπnstein et al (1992) Molec. Cell Biol 12 360, which is incorporated herein by reference) Without wishing to be bound by any particular theory, it is believed that the addition of recombinases permits efficient gene targeting with targeting polynucleotides having short (i e , about 50 to 1000 basepair long) segments of homology, as well as with targeting polynucleotides having longer segments of homology
Therefore, it is preferred that targeting polynucleotides of the invention have homology clamps that are highly homologous to the predetermined target endogenous DNA sequence(s), most preferably isogenic Typically, targeting polynucleotides of the invention have at least one homology clamp that is at least about 18 to 35 nucleotides long, and it is preferable that homology clamps are at least about 20 to 100 nucleotides long, and more preferably at least about 100-500 nucleotides long, although the degree of sequence homology between the homology clamp and the targeted sequence and the base composition of the targeted sequence will determine the optimal and minimal clamp lengths (e g , G-C rich sequences are typically more thermodynamically stable and will generally require shorter clamp length) Therefore, both homology clamp length and the degree of sequence homology can only be determined with reference to a particular predetermined sequence, but homology clamps generally must be at least about 12 nucleotides long and must also substantially correspond or be substantially complementary to a predetermined target sequence Preferably, a homology clamp is at least about 12, and preferably at least about 50 nucleotides long and is identical to or complementary to a predetermined target sequence Without wishing to be bound by a particular theory, it is believed that the addition of recombinases to a targeting polynucleotide enhances the efficiency of homologous recombination between homologous, nonisogenic sequences (e g , between an exon 2 sequence of a albumin gene of a Balb/c mouse and a homologous albumin gene exon 2 sequence of a C57/BL6 mouse), as well as between isogenic sequences
The formation of heteroduplex joints is not a stringent process, genetic evidence supports the view that the classical phenomena of meiotic gene conversion and aberrant meiotic segregation result in part from the inclusion of mismatched base pairs in heteroduplex joints, and the subsequent correction of some of these mismatched base pairs before replication Observations on recA protein have provided information on parameters that affect the discrimination of relatedness from perfect or near- perfect homology and that affect the inclusion of mismatched base pairs in heteroduplex joints The ability of recA protein to drive strand exchange past all single base-pair mismatches and to form extensively mismatched joints in superhehcal DNA reflect its role in recombination and gene conversion This error-prone process may also be related to its role in mutagenesis RecA-mediated pairing reactions involving DNA of NX174 and G4, which are about 70 percent homologous, have yielded homologous recombinants (Cunningham et al (1981 ) Cell 24 213), although recA preferentially forms homologous joints between highly homologous sequences, and is implicated as mediating a homology search process between an invading DNA strand and a recipient DNA strand, producing relatively stable heteroduplexes at regions of high homology Accordingly, it is the fact that recombinases can drive the homologous recombination reaction between strands which are significantly, but not perfectly, homologous, which allows gene conversion and the modification of target sequences Thus, targeting polynucleotides may be used to introduce nucleotide substitutions, insertions and deletions into an endogeneous DNA sequence, and thus the corresponding ammo acid substitutions, insertions and deletions in proteins expressed from the endogeneous DNA sequence
In a preferred embodiment, two substantially complementary targeting polynucleotides are used in one embodiment, the targeting polynucleotides form a double stranded hybrid, which may be coated with recombinase, although when the recombinase is recA, the loading conditions may be somewhat different from those used for single stranded nucleic acids
In a prefered embodiment, two substantially complementary single-stranded targeting polynucleotides are used The two complementary single-stranded targeting polynucleotides are usually of equal length, although this is not required However, as noted below, the stability of the four strand hybrids of the invention is putatively related, in part, to the lack of significant unhybridized single-stranded nucleic acid, and thus significant unpaired sequences are not preferred Furthermore, as noted above, the complementarity between the two targeting polynucleotides need not be perfect The two complementary single-stranded targeting polynucleotides are simultaneously or contemporaneously introduced into a target cell harboring a predetermined endogenous target sequence, generally with at lease one recombinase protein (e g , recA) Under most circumstances, it is preferred that the targeting polynucleotides are incubated with recA or other recombinase prior to introduction into a target cell, so that the recombinase proteιn(s) may be "loaded" onto the targeting polynucleotιde(s), to coat the nucleic acid, as is described below Incubation conditions for such recombinase loading are described infra, and also in U S S N 07/755,462, filed 4 September 1991 , U S S N 07/910,791 , filed 9 July 1992, and U S S N 07/520,321 , filed 7 May 1990, each of which is incorporated herein by reference A targeting polynucleotide may contain a sequence that enhances the loading process of a recombinase, for example a recA loading sequence is the recombinogenic and recombinase nucleation sequence poly[d(A-C)] and its complement, poly[d(G-T)] The duplex sequence ohgo[d(A- C)n 'd(G-T)n], where n is from 4 to 35, is a middle repetitive element in target DNA There appears to be a fundamental difference in the stability of RecA-protein-mediated D-loops formed between one single-stranded DNA (ssDNA) probe hybridized to negatively supercoiled DNA targets in comparison to relaxed or linear duplex DNA targets Internally located dsDNA target sequences on relaxed linear DNA targets hybridized by ssDNA probes produce single D-loops, which are unstable after removal of RecA protein (Adzuma, Genes Devel 6 1679 (1992), Hsieh et al, PNAS USA 89 6492 (1992), Chiu et al , Biochemistry 32 13146 (1993)) This probe DNA instability of hybrids formed with linear duplex DNA targets is most probably due to the incoming ssDNA probe W-C base pairing with the complementary DNA strand of the duplex target and disrupting the base pairing in the other DNA strand The required high free-energy of maintaining a disrupted DNA strand in an unpaired ssDNA conformation in a protein-free smgle-D-loop apparently can only be compensated for either by the stored free energy inherent in negatively supercoiled DNA targets or by base pairing initiated at the distal ends of the joint DNA molecule, allowing the exchanged strands to freely intertwine
However, the addition of a second complementary ssDNA to the three-strand-containing smgle-D-loop stabilizes the deprotemized hybrid joint molecules by allowing W-C base pairing of the probe with the displaced target DNA strand The addition of a second RecA-coated complementary ssDNA
(cssDNA) strand to the three-strand containing single D-loop stabilizes deprotemized hybrid joints located away from the free ends of the duplex target DNA (Sena & Zarhng, Nature Genetics 3 365 (1993), Revet et al J Mol Biol 232 779 (1993), Jayasena and Johnston, J Mol Bio 230 1015 (1993)) The resulting four-stranded structure, named a double D-loop by analogy with the three- stranded single D-loop hybrid has been shown to be stable in the absence of RecA protein This stability likely occurs because the restoration of W-C basepaiπng in the parental duplex would require disruption of two W-C basepairs in the double-D-loop (one W-C pair in each heteroduplex D-loop) Since each base-pairing in the reverse transition (double-D-loop to duplex) is less favorable by the energy of one W-C basepair, the pair of cssDNA probes are thus k etically trapped in duplex DNA targets in stable hybrid structures The stability of the double-D loop joint molecule within internally located probe target hybrids is an intermediate stage prior to the progression of the homologous recombination reaction to the strand exchange phase The double D-loop permits isolation of stable multistranded DNA recombination intermediates
In addition, when the targeting polynucleotides are used to generate insertions or deletions in an endogeneous nucleic acid sequence, the use of two complementary single-stranded targeting polynucleotides allows the use of internal homology clamps as depicted in Figure 13 The use of internal homology clamps allows the formation of stable deprotemized cssDNA probe target hybrids with homologous DNA sequences containing either relatively small or large insertions and deletions within a homologous DNA target Without being bound by theory, it appears that these probe target hybrids, with heterologous inserts in the cssDNA probe, are stabilized by the re-anneahng of cssDNA probes to each other within the double-D-loop hybrid, forming a novel DNA structure with an internal homology clamp Similarly stable double-D-loop hybrids formed at internal sites with heterologous inserts in the linear DNA targets (with respect to the cssDNA probe) are equally stable Because cssDNA probes are kmetically trapped within the duplex target, the multi-stranded DNA intermediates of homologous DNA pairing are stabilized and strand exchange is facilitated
In a preferred embodiment, the length of the internal homology clamp (i e the length of the insertion or deletion) is from about 1 to 50% of the total length of the targeting polynucleotide, with from about 1 to about 20% being preferred and from about 1 to about 10% being especially preferred, although in some cases the length of the deletion or insertion may be significantly larger As for the targeting homology clamps, the complementarity within the internal homology clamp need not be perfect
The invention may also be practiced with individual targeting polynucleotides which do not comprise part of a complementary pair In each case, a targeting polynucleotide is introduced into a target cell simultaneously or contemporaneously with a recombinase protein, typically in the form of a recombinase coated targeting polynucleotide as outlined herein (i e , a polynucleotide pre-mcubated with recombinase wherein the recombinase is noncovalently bound to the polynucleotide, generally referred to in the art as a nucleoprotein filament)
A targeting polynucleotide used in a method of the invention typically is a single-stranded nucleic acid, usually a DNA strand, or derived by denaturation of a duplex DNA, which is complementary to one (or both) strand(s) of the target duplex nucleic acid Thus, one of the complementary single stranded targeting polynucleotides is complementary to one strand of the endogeneous target sequence (i e Watson) and the other complementary single stranded targeting polynucleotide is complementary to the other strand of the endogeneous target sequence (i e Crick) The homology clamp sequence preferably contains at least 90-95% sequence homology with the target sequence, to insure sequence-specific targeting of the targeting polynucleotide to the endogenous DNA target Each single-stranded targeting polynucleotide is typically about 50-600 bases long, although a shorter or longer polynucleotide may also be employed Alternatively, targeting polynucleotides may be prepared in single-stranded form by oligonucleotide synthesis methods, which may first require, especially with larger targeting polynucleotides, formation of subfragments of the targeting polynucleotide, typically followed by splicing of the subfragments together, typically by enzymatic ligation
Recombinase Proteins Recombinases are proteins that, when included with an exogenous targeting polynucleotide, provide a measurable increase in the recombination frequency and/or localization frequency between the targeting polynucleotide and an endogenous predetermined DNA sequence Thus, in a preferred embodiment, increases in recombination frequency from the normal range of 10"8 to 10"4, to 10"4 to 101, preferably 103 to 101, and most preferably 102 to 101, may be acheived
In the present invention, recombinase refers to a family of RecA-hke recombination proteins all having essentially all or most of the same functions, particularly (i) the recombinase protein's ability to properly bind to and position targeting polynucleotides on their homologous targets and (n) the ability of recombinase protein/targeting polynucleotide complexes to efficiently find and bind to complementary endogenous sequences The best characterized recA protein is from E coli, in addition to the wild-type protein a number of mutant recA-hke proteins have been identified (e g , recA803, see Madiraju et al , PNAS USA 85(18) 6592 (1988), Madiraju et al, Biochem 31 10529 (1992), Lavery et al , J Biol Chem 267 20648 (1992)) Further, many organisms have recA-hke recombinases with strand-transfer activities (e g , Fugisawa et al , (1985) Nucl Acids Res 13 7473, Hsieh et al , (1986) CeJl 44 885. Hsieh et al . (1989) J Biol Chem 264 5089, Fishel et al , (1988) Proc Natl Acad Sci (USA) 85 3683, Cassuto et al , (1987) Mol Gen Genet 208 10, Ganea et al , (1987) Mol Cell Biol 7 3124. Moore et al . (1990) J Biol Chem 19 11108, Keene et al , (1984) NjjcL Acids Res 12 3057, Kimeic, (1984) Cold Spring Harbor Svmp 48 675, Kmeic, (1986) CeJI 44 545, Kolodner et al , (1987) Proc Natl Acad Sci USA 84 5560, Sugmo et al , (1985) Proc Natl Acad Sci USA 85 3683. Halbrook et al . (1989) J Biol Chem 264 21403. Eisen et al . (1988) Proc Natl Acad Sci USA 85 7481 , McCarthy et al , (1988) Proc Natl Acad Sci USA 85 5854, Lowenhaupt et al , (1989) J Biol Chem 264 20568, which are incorporated herein by reference Examples of such recombinase proteins include, for example but not limitation recA, recA803, uvsX, and other recA mutants and recA-hke recombinases (Roca, A i (1990) Cπt Rev Biochem Molec Biol 25 415), seo1 (Kolodner et al (1987) Proc Natl Acad Sci (U S A ) 84 5560. Tishkoff et al Molec Cell Biol 112593), RuvC (Dunderdale et al (1991) Nature 354 506), DST2, KEM1. XRN1 (Dykstra et al
(1991) Molec Cell Biol 11 2583). STP DST1 (Clark et al (1991) Molec Cell Biol 11 2576). HPP-1 (Moore et al (1991) Proc Natl Acad Sci (U S A ) 88 9067). other target recombinases (Bishop et al
(1992) CeJ 69 439, Sh ohara et al (1992) CeH 69 457), incorporated herein by reference RecA may be purified from E coli strains, such as E coli strains JC12772 and JC15369 (available from A J
Clark and M Madiraju, University of California-Berkeley, or purchased commercially) These strains contain the recA coding sequences on a "runaway" replicating plasmid vector present at a high copy numbers per cell The recA803 protein is a high-activity mutant of wild-type recA The art teaches several examples of recombinase proteins, for example, from Drosophila, yeast, plant, human, and non-human mammalian cells, including proteins with biological properties similar to recA (i e , recA-hke recombinases), such as Rad51 from mammals and yeast, and Pk-rec (see Rashid et al , Nucleic Acid Res 25(4) 719 (1997), hereby incorporated by reference) In addition, the recombinase may actually be a complex of proteins, i e a Arecombιnosome@ In addition, included within the definition of a recombinase are portions or fragments of recombinases which retain recombinase biological activity, as well as variants or mutants of wild-type recombinases which retain biological activity, such as the E coli recA803 mutant with enhanced recombinase activity
In a preferred embodiment, recA or rad51 is used For example, recA protein is typically obtained from bacterial strains that overproduce the protein wild-type E coli recA protein and mutant recA803 protein may be purified from such strains Alternatively, recA protein can also be purchased from, for example, Pharmacia (Piscataway, NJ)
RecA proteins, and its homologs, form a nucleoprotein filament when it coats a single-stranded DNA In this nucleoprotein filament, one monomer of recA protein is bound to about 3 nucleotides This property of recA to coat single-stranded DNA is essentially sequence independent, although particular sequences favor initial loading of recA onto a polynucleotide (e g , nucleation sequences) The nucleoprotein filament(s) can be formed on essentially any DNA molecule and can be formed in cells (e g , mammalian cells), forming complexes with both single-stranded and double-stranded DNA, although the loading conditions for dsDNA are somewhat different than for ssDNA
Recombinase Coating of Targeting Polynucleotides
The conditions used to coat targeting polynucleotides with recombinases such as recA protein and ATP(S have been described in commonly assigned U S S N 07/910,791 , filed 9 July 1992, U S S N 07/755,462, filed 4 September 1991 , and U S S N 07/520,321, filed 7 May 1990, each incorporated herein by reference The procedures below are directed to the use of E coli recA, although as will be appreciated by those in the art, other recombinases may be used as well Targeting polynucleotides can be coated using GTP(S, mixes of ATP(S with rATP, rGTP and/or dATP, or dATP or rATP alone in the presence of an rATP generating system (Boehnnger Mannheim) Various mixtures of GTP(S, ATP(S, ATP, ADP, dATP and/or rATP or other nucleosides may be used, particularly preferred are mixes of ATP(S and ATP or ATP(S and ADP
RecA protein coating of targeting polynucleotides is typically carried out as described in U S S N 07/910,791 , filed 9 July 1992 and U S S N 07/755,462, filed 4 September 1991 , which are incorporated herein by reference Briefly, the targeting polynucleotide, whether double-stranded or single-stranded, is denatured by heating in an aqueous solution at 95-100°C for five minutes, then placed in an ice bath for 20 seconds to about one minute followed by centrifugation at 0°C for approximately 20 sec, before use When denatured targeting polynucleotides are not placed in a freezer at -20°C they are usually immediately added to standard recA coating reaction buffer containing ATP(S, at room temperature, and to this is added the recA protein Alternatively, recA protein may be included with the buffer components and ATP(S before the polynucleotides are added
RecA coating of targeting polynucleotιde(s) is initiated by incubating polynucleotide-recA mixtures at 37°C for 10-15 mm RecA protein concentration tested during reaction with polynucleotide varies depending upon polynucleotide size and the amount of added polynucleotide, and the ratio of recA molecule nucleotide preferably ranges between about 3 1 and 1 3 When single-stranded polynucleotides are recA coated independently of their homologous polynucleotide strands, the mM and μM concentrations of ATP(S and recA, respectively, can be reduced to one-half those used with double-stranded targeting polynucleotides (i e , recA and ATP(S concentration ratios are usually kept constant at a specific concentration of individual polynucleotide strand, depending on whether a smgle- or double-stranded polynucleotide is used)
RecA protein coating of targeting polynucleotides is normally carried out in a standard 1X RecA coating reaction buffer 10X RecA reaction buffer (i e , 10x AC buffer) consists of 100 mM Tπs acetate (pH 7 5 at 37°C), 20 mM magnesium acetate, 500 mM sodium acetate, 10 mM DTT, and 50% glycerol) All of the targeting polynucleotides, whether double-stranded or single-stranded, typically are denatured before use by heating to 95-100°C for five minutes, placed on ice for one minute, and subjected to centrifugation (10,000 rpm) at 0°C for approximately 20 seconds (e g , in a Tomy centrifuge) Denatured targeting polynucleotides usually are added immediately to room temperature RecA coating reaction buffer mixed with ATP(S and diluted with buffer or double-distilled H20 as necessary
A reaction mixture typically contains the following components (i) 0 2-4 8 mM ATP(S, and (n) between 1-100 ng/μl of targeting polynucleotide To this mixture is added about 1-20 μl of recA protein per 10- 100 μl of reaction mixture, usually at about 2-10 mg/ml (purchased from Pharmacia or purified), and is rapidly added and mixed The final reaction volume-for RecA coating of targeting polynucleotide is usually in the range of about 10-500 μl RecA coating of targeting polynucleotide is usually initiated by incubating targeting polynucleotide-RecA mixtures at 37°C for about 10-15 mm
RecA protein concentrations in coating reactions varies depending upon targeting polynucleotide size and the amount of added targeting polynucleotide recA protein concentrations are typically in the range of 5 to 50 μM When single-stranded targeting polynucleotides are coated with recA, independently of their complementary strands, the concentrations of ATP(S and recA protein may optionally be reduced to about one-half of the concentrations used with double-stranded targeting polynucleotides of the same length that is, the recA protein and ATP(S concentration ratios are generally kept constant for a given concentration of individual polynucleotide strands
The coating of targeting polynucleotides with recA protein can be evaluated in a number of ways First, protein binding to DNA can be examined using band-shift gel assays (McEntee et al , (1981 ) J_ Biol Chem 256 8835) Labeled polynucleotides can be coated with recA protein in the presence of ATP(S and the products of the coating reactions may be separated by agarose gel electrophoresis Following incubation of recA protein with denatured duplex DNAs the recA protein effectively coats single-stranded targeting polynucleotides derived from denaturing a duplex DNA As the ratio of recA protein monomers to nucleotides in the targeting polynucleotide increases from 0, 1 27, 1 2 7 to 3 7 1 for 121-mer and 0, 1 22, 1 2 2 to 4 5 1 for 159-mer, targeting polynucleotide's electrophoretic mobility decreases, i e , is retarded, due to recA-b ding to the targeting polynucleotide Retardation of the coated polynucleotide's mobility reflects the saturation of targeting polynucleotide with recA protein An excess of recA monomers to DNA nucleotides is required for efficient recA coating of short targeting polynucleotides (Leahy et al , (1986) J Biol Chem 261 954)
A second method for evaluating protein binding to DNA is in the use of nitrocellulose filter binding assays (Leahy et al , (1986) J Biol Chem 261 6954. Woodbury, et al , (1983) Biochemistry 22(20) 4730-4737 The nitrocellulose filter binding method is particularly useful in determining the dissociation-rates for protein DNA complexes using labeled DNA In the filter binding assay, DNA protein complexes are retained on a filter while free DNA passes through the filter This assay method is more quantitative for dissociation-rate determinations because the separation of DNA protein complexes from free targeting polynucleotide is very rapid
Alternatively, recombinase proteιn(s) (prokaryotic, eukaryotic or endogeneous to the target cell) may be exogenously induced or administered to a target cell simultaneously or contemporaneously (i e , within about a few hours) with the targeting polynucleotιde(s) Such administration is typically done by micro-injection, although electroporation, lipofection, and other transfection methods known in the art may also be used Alternatively, recombinase-proteins may be produced m vivo For example, they may be produced from a homologous or heterologous expression cassette in a transfected cell or transgenic cell, such as a transgenic totipotent cell (e g a fertilized zygote) or an embryonal stem cell (e g , a muπne ES cell such as AB-1 ) used to generate a transgenic non-human animal line or a somatic cell or a pluπpotent hematopoietic stem cell for reconstituting all or part of a particular stem cell population (e g hematopoietic) of an individual Conveniently, a heterologous expression cassette includes a modulatable promoter, such as an ecdysone-mducible promoter-enhancer combination, an estrogen-induced promoter-enhancer combination, a CMV promoter-enhancer, an insulin gene promoter, or other cell-type specific, developmental stage-specific, hormone-inducible, or other modulatable promoter construct so that expression of at least one species of recombinase protein from the cassette can by modulated for transiently producing recombιnase(s) in vivo simultaneous or contemporaneous with introduction of a targeting polynucleotide into the cell When a hormone-inducible promoter-enhancer combination is used, the cell must have the required hormone receptor present, either naturally or as a consequence of expression a co-transfected expression vector encoding such receptor Alternatively, the recombinase may be endogeneous and produced in high levels In this embodiment, preferably in eukaryotic target cells such as tumor cells, the target cells produce an elevated level of recombinase In other embodiments the level of recombinase may be induced by DNA damaging agents, such as mitomycm C, UV or (-irradiation Alternatively, recombinase levels may also be elevated by transfection of a virus or plasmid encoding the recombinase gene into the cell
Cell-Uptake Components
A targeting polynucleotide of the invention may optionally be conjugated, typically by covalently or preferably noncovalent binding, to a cell-uptake component Various methods have been described in the art for targeting DNA to specific cell types A targeting polynucleotide of the invention can be conjugated to essentially any of several cell-uptake components known in the art For targeting to hepatocytes, a targeting polynucleotide can be conjugated to an asialoorosomucoid (ASOR)-poly-L- lysme conjugate by methods described in the art and incorporated herein by reference (Wu GY and Wu CH (1987) J Biol Chem 262 4429, Wu GY and Wu CH (1988) Biochemistry 27 887, Wu GY and Wu CH (1988) J Biol Chem 263 14621. Wu GY and Wu CH (1992) J Biol Chem 267 12436, Wu et al (1991) J Biol Chem 266 14338, and Wilson et al (1992) J Biol Chem 267 963, WO92/06180, WO92/05250, and W091/17761 , which are incorporated herein by reference)
Alternatively, a cell-uptake component may be formed by incubating the targeting polynucleotide with at least one lipid species and at least one protein species to form protein-hpid-polynucleotide complexes consisting essentially of the targeting polynucleotide and the lipid-protem cell-uptake component Lipid vesicles made according to Feigner (W091/17424, incorporated herein by reference) and/or cationic hpidization (WO91/16024, incorporated herein by reference) or other forms for polynucleotide administration (EP 465,529, incorporated herein by reference) may also be employed as cell-uptake components Nucleases may also be used
In addition to cell-uptake components, targeting components such as nuclear localization signals may be used, as is known in the art
Homologous Pairing of Targeting Polynucleotides Having Chemical Substituents In addition to recombinase and cellular uptake components, the targeting polynucleotides may include chemical substituents Exogenous targeting polynucleotides that have been modified with appended chemical substituents may be introduced along with recombinase (e g , recA) into a metabolically active target cell to homologously pair with a predetermined endogenous DNA target sequence in the cell In a preferred embodiment, the exogenous targeting polynucleotides are derivatized, and additional chemical substituents are attached, either during or after polynucleotide synthesis, respectively, and are thus localized to a specific endogenous target sequence where they produce an alteration or chemical modification to a local DNA sequence Preferred attached chemical substituents include, but are not limited to cross-linking agents (see Podyminogm et al , Biochem 34 13098 (1995) and 35 7267 (1996), both of which are hereby incorporated by reference), nucleic acid cleavage agents, metal chelates (e g , iron/EDTA chelate for iron catalyzed cleavage), topoisomerases, endonucleases, exonucleases, hgases, phosphodiesterases, photodynamic porphyπns, chemotherapeutic drugs (e g , adriamycin, doxirubicm), intercalating agents, labels, base- modification agents, agents which normally bind to nucleic acids such as labels, etc (see for example Afonina et al , PNAS USA 93 3199 (1996), incorporated herein by reference) immunoglobulm chains, and oligonucleotides Iron/EDTA chelates are particularly preferred chemical substituents where local cleavage of a DNA sequence is desired (Hertzberg et al (1982) J Am Chem Soc 104 313, Hertzberg and Dervan (1984) Biochemistry 23 3934, Taylor et al (1984) Tetrahedron 40 457, Dervan, PB ( 1986) Science 232 464, which are incorporated herein by reference) Further preferred are groups that prevent hybridization of the complementary single stranded nucleic acids to each other but not to unmodified nucleic acids, see for example Kutryavin et al , Biochem 35 11170 (1996) and Woo et al , Nucleic Acid Res 24(13) 2470 (1996), both of which are incorporated by reference 2'-0 methyl groups are also preferred, see Cole-Strauss et al , Science 273 1386 (1996), Yoon et al , PNAS 93 2071 (1996)) Additional preferred chemical substitutents include labeling moieties, including fluorescent labels Preferred attachment chemistries include direct linkage, e g , via an appended reactive ammo group (Corey and Schultz (1988) Science 238 1401 , which is incorporated herein by reference) and other direct linkage chemistries, although streptavidin/biotin and digoxigenm/antidigoxigenin antibody linkage methods may also be used Methods for linking chemical substituents are provided in U S Patents 5,135,720, 5,093,245, and 5,055,556, which are incorporated herein by reference Other linkage chemistries may be used at the discretion of the practitioner
Typically, a targeting polynucleotide of the invention is coated with at least one recombinase and is conjugated to a cell-uptake component, and the resulting cell targeting complex is contacted with a target cell under uptake conditions (e g , physiological conditions) so that the targeting polynucleotide and the recombιnase(s) are internalized in the target cell A targeting polynucleotide may be contacted simultaneously or sequentially with a cell-uptake component and also with a recombinase, preferably the targeting polynucleotide is contacted first with a recombinase, or with a mixture comprising both a cell-uptake component and a recombinase under conditions whereby, on average, at least about one molecule of recombinase is noncovalently attached per targeting polynucleotide molecule and at least about one cell-uptake component also is noncovalently attached Most preferably, coating of both recombinase and cell-uptake component saturates essentially all of the available binding sites on the targeting polynucleotide A targeting polynucleotide may be preferentially coated with a cell-uptake component so that the resultant targeting complex comprises, on a molar basis, more cell-uptake component than recombιnase(s) Alternatively, a targeting polynucleotide may be preferentially coated with recombιnase(s) so that the resultant targeting complex comprises, on a molar basis, more recombιnase(s) than cell-uptake component
Cell-uptake components are included with recombmase-coated targeting polynucleotides of the invention to enhance the uptake of the recombmase-coated targeting polynucleotιde(s) into cells, particularly for in vivo gene targeting applications, such as gene therapy to treat genetic diseases, including neoplasia, and targeted homologous recombination to treat viral infections wherein a viral sequence (e g , an integrated hepatitis B virus (HBV) genome or genome fragment) may be targeted by homologous sequence targeting and inactivated Alternatively, a targeting polynucleotide may be coated with the cell-uptake component and targeted to cells with a contemporaneous or simultaneous administration of a recombinase (e g , liposomes or immunohposomes containing a recombinase, a viral-based vector encoding and expressing a recombinase)
Once the recombinase-targeting polynucleotide compositions are formulated, they are introduced or administered into target cells The administration is typically done as is known for the administration of nucleic acids into cells, and, as those skilled in the art will appreciate, the methods may depend on the choice of the target cell Suitable methods include, but are not limited to, microinjection, electroporation, lipofection, etc By Atarget cells® herein is meant prokaryotic or eukaryotic cells Suitable prokaryotic cells include, but are not limited to, bacteria such as E coli, Bacillus species, and the extremophile bacteria such as thermophiles, etc Preferably, the procaryotic target cells are recombination competent Suitable eukaryotic cells include, but are not limited to, fungi such as yeast and filamentous fungi, including species of Aspergillus, Tnchoderma, and Neurospora, plant cells including those of corn, sorghum, tobacco, canola, soybean, cotton, tomato, potato, alfalfa, sunflower, etc , and animal cells, including fish, birds and mammals Suitable fish cells include, but are not limited to, those from species of salmon, trout, tulapia, tuna, carp, flounder, halibut, swordfish, cod and zebrafish Suitable bird cells include, but are not limited to, those of chickens, ducks, quail, pheasants and turkeys, and other jungle fowl or game birds Suitable mammalian cells include, but are not limited to, cells from horses, cattle, buffalo, deer, sheep, rabbits, rodents such as mice, rats, hamsters, gerbils, and guinea pigs, minks, goats, pigs, primates, marsupials, marine mammals including dolphins and whales, as well as cell lines, such as human cell lines of any tissue or stem cell type, and stem cells, including pluπpotent and non-pluπpotent, and non-human zygotes
In a preferred embodiment, procaryotic cells are used In this embodiment, a pre-selected target DNA sequence is chosen for alteration Preferably, the pre-selected target DNA sequence is contained within an extrachromosomal sequence By Aextrachromosomal sequence® herein is meant a sequence separate from the chromosomal or genomic sequences Preferred extrachromosomal sequences include plasmids (particularly procaryotic plasmids such as bacterial plasmids), P1 vectors, viral genomes, yeast, bacterial and mammalian artificial chromosomes (YAC, BAC and MAC, respectively), and other autonomously self-rephcating sequences, although this is not required As described herein, a recombinase and at least two single stranded targeting polynucleotides which are substantially complementary to each other, each of which contain a homology clamp to the target sequence contained on the extrachromosomal sequence, are added to the extrachromosomal sequence, preferably in vitro The two single stranded targeting polynucleotides are preferably coated with recombinase, and at least one of the targeting polynucleotides contain at least one nucleotide substitution, insertion or deletion The targeting polynucleotides then bind to the target sequence in the extrachromosomal sequence to effect homologous recombination and form an altered extrachromosomal sequence which contains the substitution, insertion or deletion The altered extrachromosomal sequence is then introduced into the procaryotic cell using techniques known in the art Preferably, the recombinase is removed prior to introduction into the target cell, using techniques known in the art For example, the reaction may be treated with proteases such as proteinase K, detergents such as SDS, and phenol extraction (including phenol chloroform isoamyl alcohol extraction) These methods may also be used for eukaryotic cells
Alternatively, the pre-selected target DNA sequence is a chromosomal sequence In this embodiment, the recombinase with the targeting polynucleotides are introduced into the target cell, preferably eukaryotic target cells In this embodiment, it may be desirable to bind (generally non-covalently) a nuclear localization signal to the targeting polynucleotides to facilitate localization of the complexes in the nucleus See for example Kido et al , Exper Cell Res 198 107-114 (1992), hereby expressly incorporated by reference The targeting polynucleotides and the recombinase function to effect homologous recombination, resulting in altered chromosomal or genomic sequences
In a preferred embodiment, eukaryotic cells are used For making transgenic non-human animals (which include homologously targeted non-human animals) embryonal stem cells (ES cells) and fertilized zygotes are preferred In a preferred embodiment, embryonal stem cells are used Muπne ES cells, such as AB-1 line grown on mitotically inactive SNL76/7 cell feeder layers (McMahon and Bradley, Cell 62 1073-1085 (1990)) essentially as described (Robertson, E J (1987) in Teratocarcinomas and Embryonic Stem Cells A Practical Approach E J Robertson, ed (oxford IRL Press), p 71-112) may be used for homologous gene targeting Other suitable ES lines include, but are not limited to, the E14 line (Hooper et al (1987) Nature 326 292-295), the D3 line (Doetschman et al (1985) J Embrvol Exp Morph 87 21-45), and the CCE line (Robertson et al (1986) Nature 323 445-448) The success of generating a mouse line from ES cells bearing a specific targeted mutation depends on the plunpotence of the ES cells (i e , their ability, once injected into a host blastocyst, to participate in embryogenesis and contribute to the germ cells of the resulting animal)
The plunpotence of any given ES cell line can vary with time in culture and the care with which it has been handled The only definitive assay for plunpotence is to determine whether the specific population of ES cells to be used for targeting can give rise to chimeras capable of germhne transmission of the ES genome For this reason, prior to gene targeting, a portion of the parental population of AB-1 cells is injected into C57B1/6J blastocysts to ascertain whether the cells are capable of generating chimeπc mice with extensive ES cell contribution and whether the majority of these chimeras can transmit the ES genome to progeny
In a preferred embodiment, non-human zygotes are used, for example to make transgenic animals, using techniques known in the art (see U S Patent No 4,873,191 ) Preferred zygotes include, but are not limited to, animal zygotes, including fish, avian and mammalian zygotes Suitable fish zygotes include, but are not limited to, those from species of salmon, trout, tuna, carp, flounder, halibut, swordfish, cod, tulapia and zebrafish Suitable bird zygotes include, but are not limited to, those of chickens, ducks, quail, pheasant, turkeys, and other jungle fowl and game birds Suitable mammalian zygotes include, but are not limited to, cells from horses, cattle, buffalo, deer, sheep, rabbits, rodents such as mice, rats, hamsters and guinea pigs, goats, pigs, primates, and marine mammals including dolphins and whales See Hogan et al , Manipulating the Mouse Embryo (A Laboratory Manual), 2nd Ed Cold Spring Harbor Press, 1994, incorporated by reference
Once made and administered to a target host cell, the compositions of the invention find use in a number of applications, including the creation of transgenic plants and animals Such transgenic animals can be any of the animals, fish and birds outlined above as suitable for zygotes Preferably the transgenic animals are mammals, including, but not limited to, farm animals such as cattle, buffalo, goats, including BELE® goats, sheep, and pigs or other transgenic animals such as mice, rabbits, monkeys, etc In a preferred embodiment, the animals or mammals are non-human
In general, transgenic animals are made with any number of changes Exogeneous sequences, or extra copies of endogeneous sequences, including structural genes and regulatory sequences, may be added to the animal, as outlined below Endogeneous sequences (again, either genes or regulatory sequences) may be disrupted, i e via insertion, deletion or substitution, to prevent expression of endogeneous proteins Alternatively, endogeneous sequences may be modified to alter their biological function, for example via mutation of the endogeneous sequence by insertion, deletion or substitution
Accordingly, tThe methods of the present invention are useful to add exogenous DNA sequences, such as exogenous genes or regulatory sequences, extra copies of endogenous genes or regulatory sequences, or exogeneous genes or regulatory sequences, to a transgenic plant or animal This may be done for a number of reasons for example, adding one or more copies of a wild-type gene can increase the production of a desirable gene product, adding or deleting one or more copies of a therapeutic gene can alleviate a disease state, or to create an animal model of disease Adding one or more copies of a modified wild type gene may be done for the same reasons Adding therapeutic genes or proteins may yield superior transgenic animals, for example for the production of therapeutic or nutπceutical proteins Adding human genes to non-human mammals may facilitate production of human proteins and adding regulatory sequences derived from human or non-human mammals may be useful to increase or decrease the expression of endogenous or exogenous genes Such inserted genes may be under the control of endogenous or exogenous regulatory sequences, as described herein
The methods of the invention are also useful to modify endogeneous gene sequences, as outlined below Suitable endogenous gene targets include, but are not limited to, genes which encode peptides or proteins including enzymes, structural or soluble proteins, as well as endogeneous regulatory sequences including, but not limited to, promoters, transcriptional or translational sequences, repetitive sequencs including olιgo[d(A-C)n «d(G-T)n], ohgo[d(A-T)]n, olιgo[d(C-T)]n, etc Examples of such endogenous gene targets include, but are not limited to, genes which encode lactoglobuhns including both α-lactoglobuhn and $-lactoglobuhn, casein, including both α-casem, β-casem and κ-caseιn, albumins, including serum albumin, particularly human and bovine, immunoglobuhns, including IgE, IgM, IgG and IgD and monoclonal antibodies, globm, integπn, hormones, growth factors, particularly bovine and human growth factors, including transforming growth factor, epidermal growth factor, nerve growth factors, etc , collagen, mterleukins, including IL-1 to IL- 17, a major histocompatibihty antigen (MHC), G-protem coupled receptors (GPCR), nuclear receptors, ion channels, multidrug resistance genes, amyloid proteins, enzymes, including esterases, proteases (including tissue plasminogen activator (tPA)), hpases, carbohydrases, etc , APRT, HPRT, leptm, tumor suppressor genes, provirus, pπons, OTC, CFTR, sugar transferases such as alpha-galactosyl transferase (galT) or fucosyl transferase, a milk or urine protein gene including the caseins, lactoferπn and whey proteins, oncogenes, cytok es, particularly human, transcription factors, and other pharmaceuticals Any or all of these may also be suitable exogeneous genes to add to a genome using the methods outlined herein
Endogeneous genes (or regulatory sequences, as outlined herein) may be modified in several ways, including disruptions and alterations
The endogenous target gene may be disrupted in a variety of ways The term Adisrupt® as used herein comprises a change in the coding or non-coding sequence of an endogenous nucleic acid that alters the transcription or translation of an endogenous gene In a preferred embodiment, a disrupted gene will no longer produce a functional gene product Generally, disruption may occur by either the insertion, deletion or frame shifting of nucleotides
The term Ainsertion sequence® as used herein means one or more nucleotides which are inserted into an endogenous gene to disrupt it In general, insertion sequences can be as short as 1 nucleotide or as long as a gene, as outlined below For non-gene insertion sequences, the sequences are at least 1 nucleotide, with from about 1 to about 50 nucleotides being preferred, and from about 10 to 25 nucleotides being particularly preferred An insertion sequence may comprise a polylinker sequence, with from about 1 to about 50 nucleotides being preferred, and from about 10 to 25 nucleotides being particularly preferred
In a preferred embodiment, an insertion sequence comprises a gene which not only disrupts the endogenous gene, thus preventing its expression, but also can result in the expression of a new gene product Thus, in a preferred embodiment, the disruption of an endogenous gene by an insertion sequence gene is done in such a manner to allow the transcription and translation of the insertion gene An insertion sequence that encodes a gene may range from about 50 bp to 5000 bp of cDNA or about 5000 bp to 50000 bp of genomic DNA As will be appreciated by those in the art, this can be done in a variety of ways In a preferred embodiment, the insertion gene is targeted to the endogenous gene in such a manner as to utilize endogenous regulatory sequences, including promoters, enhancers or a regulatory sequence In an alternate embodiment, the insertion sequence gene includes its own regulatory sequences, such as a promoter, enhancer or other regulatory sequence etc
Particularly preferred insertion sequence genes include, but are not limited to, genes which encode therapeutic and nutπceutical proteins, and reporter genes Suitable insertion sequence genes which may be inserted into endogenous genes include, but are not limited to, nucleic acids which encode those genes listed as suitable endogeneous genes for alterations, above, particularly mammalian enzymes, mammalian antibodies, mammalian proteins including serum albumin as well as mammalian therapeutic genes In a preferred embodiment, the inserted mammalian gene is a human gene Suitable reporter genes are those genes which encode detectable proteins, such as the genes encoding luciferase, β-galactosidase (both of which require the addition of reporter substrates), and the fluorescent proteins, including green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), and red fluorescent protein (RFP)
Thus, in a preferred embodiment, the targeted sequence modification creates a sequence that has a biological activity or encodes a polypeptide having a biological activity In a preferred embodiment, the polypeptide is an enzyme with enzymatic activity In another preferred embodiment, the polypeptide is an antibody In a third preferred embodiment, the polypeptide is a structural protein
In addition, the insertion sequence genes may be modified or variant genes, i e they contain a mutation from the wild-type sequence Thus, for example, modified genes including, but not limited to, improved therapeutic genes, modified "-lactalbumm genes that do not encode any phenylalanme residues, or human enzyme or human antibody genes that do not encode any phenylalanme residues
The term Adeletion® as used herein comprises removal of a portion of the nucleic acid sequence of an endogenous gene Deletions range from about 1 to about 100 nucleotides, with from about 1 to 50 nucleotides being preferred and from about 1 to about 25 nucleotides being particularly preferred, although in some cases deletions may be much larger, and may effectively comprise the removal of the entire endogenous gene and/or its regulatory sequences Deletions may occur in combination with substitutions or modifications to arrive at a final modified endogenous gene
In a preferred embodiment, endogenous genes may be disrupted simultaneously by an insertion and a deletion For example, some or all of an endogenous gene, with or without its regulatory sequences, may be removed and replaced with an insertion sequence gene Thus, for example, all but the regulatory sequences of an endogenous gene may be removed, and replaced with an insertion sequence gene, which is now under the control of the endogenous gene's regulatory elements
The term Aregulatory element® is used herein to describe a non-coding sequence which affects the transcription or translation of a gene including, but are not limited to, promoter sequences, πbosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, enhancer or activator sequences, or dimeπzing sequences In a preferred embodiment, the regulatory sequences include a promoter and transcriptional start and stop sequence Promoter sequences encode either constitutive or inducible promoters The promoters may be either naturally occurring promoters or hybrid promoters Hybrid promoters, which combine elements of more than one promoter, are also known in the art, and are useful in the present invention
In addition to disrupting endogeneous genes, the endogeneous genes may be altered by substitutions, insertions or deletions of nucleotides that do not completely eliminate the biological function of the sequence, but rather alter it That is, targeted gene modifications may be made to alter gene function For example, defective genes may be fixed, or the activity of a gene may be modulated, either increasing or decreasing the activity of the sequence (either the nucleic acid sequence, for example in the case of regulatory nucleic acid, or of the gene product, i e the ammo acid sequence of the protein may be altered)
The methods of the present invention are useful to provide methods for fully or partially modifying endogenous regulatory sequences Suitable targets for such fully or partially modified regulatory sequences include, but are not limited to, regulatory sequences that regulate any of the suitable endogeneous genes listed above, with preferred embodiments altering the endogeneous regulatory sequences that control the genes which encode "-lactoglobulm, $-lactoglobuhn, casein, α-casem, β- casein, κ-caseιn, serum albumin, globin, IgG, integπn, lactoferπn, a refroviral provirus, a pπon, alpha-galactosyl transferase (galT), a sugar transferase or a milk or urine production gene Examples of such fully or partially modified endogenous regulatory sequences include, but are not limited to, a modified regulatory element for an endogenous gene, a modified transcriptional regulation cassette or start site for an endogenous gene, a modified promoter, transcription initiation site, or enhancer sequences
When the modification of the endogeneous gene is to alter a structural gene, generally ammo acid changes will be made as is known in the art Substitutions, deletions, insertions or any combination thereof may be used to arrive at a final derivative Generally these changes are done on a few ammo acids to minimize the alteration of the molecule However, larger changes may be tolerated in certain circumstances or for certain purposes When small alterations in the characteristics of the endogeneous protein are desired, substitutions are generally made in accordance with the following chart
Chart I Original Residue Exemplary Substitutions
Ala Ser
Arg Lys
Asn Gin, His
Asp Glu Cys Ser
Gin Asn Glu Asp
Gly Pro
His Asn, Gin
He Leu, Val
Leu lie, Val
Lys Arg, Gin, Glu
Met Leu, lie
Phe Met, Leu, Tyr
Ser Thr
Thr Ser
Trp Tyr
Tyr Trp, Phe
Val lie, Leu
Substantial changes in function or immunological identity are made by selecting substitutions that are less conservative than those shown in Chart I For example, substitutions may be made which more significantly affect the structure of the polypeptide backbone in the area of the alteration, for example the α-hehcal or β-sheet structure, the charge or hydrophobicity of the molecule at the target site, or the bulk of the side chain The substitutions which in general are expected to produce the greatest changes in the polypeptide's properties are those in which (a) a hydrophilic residue, e g seryl or threonyl, is substituted for (or by) a hydrophobic residue, e g leucyl, isoleucyl, phenylalanyl, valyl or alanyl, (b) a cysteme or prolme is substituted for (or by) any other residue, (c) a residue having an electropositive side chain, e g lysyl, argmyl, or histidyl, is substituted for (or by) an electronegative residue, e g glutamyl or aspartyl, or (d) a residue having a bulky side chain, e g phenylalanme, is substituted for (or by) one not having a side chain, e g glycine
Preferred embodiments of the present invention include, but are not limited to (1 ) a farm animal including cattle, sheep, pigs, horses and goats with a 1-25 base pair deletion, or a 10-25 base pair insertion of a poly nker sequence, or insertion of a reporter gene such as a luciferase gene, a β- galactosidase gene or a green fluorescent (GFP) protein gene in an endogenous gene or sequence encoding ornithme transcarbamylase (OTC), lactoglobulm, casein, β-casein, α-casein, κ-caseιn, albumin, globin, immunoglobulm, IgG, mterleukin, a sugar transferase, integπn, a milk protein, a urine protein, a retroviral provirus, an endogenous virus, a pπon, a leptm, or cystic fibrosis transmembrane regulator (CFTR), (2) a farm animal including cattle, sheep, pigs, horses and goats with an exogenous gene such as a gene encoding human lysozyme, human growth hormone, human serum albumin, human globin, a human antibody (human IgG), a tissue plasminogen activator, a human therapeutic protein, human lactase, a human hpase, a hormone receptor gene, a viral receptor gene, a G-protein coupled receptor gene, a drug or a human enzyme gene, including for example the human lysozyme gene, the human α-1 anti-trypsin gene, the human anti-thrombin III gene, (4) a farm animal including cattle, sheep, pigs, horses and goats with a modified endogenous repeated (A-C)n sequence, a modified repeated (A-G)n sequence, a modified repeated (A-T)π sequence, a modified endogenous CFTR gene or a modified endogenous OTC gene, (5) a farm animal including cattle, sheep, pigs, horses and goats with a modified "-lactoglobulm gene or $-lactoglobulιn gene does not encode any phenylalanme residues, (6) a farm animal including cattle, sheep, pigs, horses and goats with a human monoclonal antibody gene, or a gene for a human antibody that does not encode any phenylalanme residues, for example inserted (or replacing) in the endogenous gene or sequence encoding an immunoglobulm, or IgG, and (7) a farm animal including cattle, sheep, pigs, horses and goats with a human gene under control of its endogenous promoter, a modified endogenous regulatory element for an endogenous gene which may or may not be disrupted by an insertion sequence, a transcriptional regulation cassette ord a dimeπzing sequence Specific preferred embodiments also include, a farm animal including cattle, sheep, pigs, horses and goats with an endogenous regulatory element which is disrupted by, deletion of at least one nucleotide
Additional preferred embodiments comprise a pig, monkey or cow with a 1-25 to 1-50 base pair insertion, examples of which include a hormone receptor gene, a viral receptor gene or a G-protein coupled receptor gene, or a 1-25 to 1-50 bp deletion in a sugar transferase gene including the α- galactosyl transferase gene (galT) or the fucosyl transferase gene, a BELE® goat with a human gene, and a pig, goat, sheep or cow with a 1-25 base pair insertion or a 1-25 base pair deletion in a endogenous refroviral provirus gene such as deletion of the sequence for proviral KC Further specific preferred embodiments include, a cow with a modified milk production gene such as, a cow with a lactase gene insertion in a milk promoter, a cow with the human lactoferπn gene replacing the bovine lactoferπn gene, a monkey with a human therapeutic gene, or a human antibody gene, a cow with the human hpase gene in a milk promoter, a cow with a human gene placed in a transcription initiation site of a milk gene under the control of its endogenous promoter, a cow with a human gene placed in a transcription initiation site of a globin gene under the control of its endogenous globin gene promoter, a cow and goat with a modified urine protein gene, a mammal with a modified endogenous leptm gene, a modified endogenous OTC gene, a modified endogenous CFTR gene or a modified interleukm gene Additional preferred embodiments include an animal such as a mouse, rabbit or goat with a transcriptional regulation cassette inserted in the transcriptional start site of an integπn gene, and a mouse with a modification in the integπn gene or G-protem coupled receptor gene
The vectors containing the DNA segments of interest can be transferred into the host cell by well- known methods, depending on the type of cellular host For example, micro-injection is commonly utilized for target cells, although calcium phosphate treatment, electroporation, lipofection, biolistics or viral-based transfection also may be used Other methods used to transform mammalian cells include the use of Polybrene, protoplast fusion, and others (see, generally, Sambrook et al Molecular Cloning A Laboratory Manual, 2d ed , 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N Y , which is incorporated herein by reference) Direct injection of DNA and/or recombmase-coated targeting polynucleotides into target cells, such as skeletal or muscle cells also may be used (Wolff et al (1990) Science 247 1465, which is incorporated herein by reference)
Targeting of Endogenous DNA Sequences
Once made and administered to a target host cell, the compositions of the invention find use in a number of applications, including the site directed modification of endogeneous sequences within any target cell, the creation of transgenic plants and animals, and the use of the compositions to do site-directed mutagenesis or modifications of target sequences
Generally, any predetermined endogenous DNA sequence, such as a gene sequence, can be altered by homologous recombination (which includes gene conversion) with an exogenous targeting polynucleotides (such as a complementary pair of single-stranded targeting polynucleotides) The target polynucleotides have at least one homology clamp which substantially corresponds to or is substantially complementary to a predetermined endogenous DNA target sequence and are introduced with a recombinase (e g , recA) into a target cell having the predetermined endogenous DNA sequence Typically, a targeting polynucleotide (or complementary polynucleotide pair) has a portion or region having a sequence that is not present in the preselected endogenous targeted sequence(s) (i e , a nonhomologous portion or mismatch) which may be as small as a single mismatched nucleotide, several mismatches, or may span up to about several kilobases or more of nonhomologous sequence Generally, such nonhomologous portions are flanked on each side by homology clamps, although a single flanking homology clamp may be used Nonhomologous portions are used to make insertions, deletions, and/or replacements in a predetermined endogenous targeted DNA sequence, and/or to make single or multiple nucleotide substitutions in a predetermined endogenous target DNA sequence so that the resultant recombined sequence (i e , a targeted recombinant endogenous sequence) incorporates some or all of the sequence information of the nonhomologous portion of the targeting polynucleotιde(s) Thus, the nonhomologous regions are used to make variant sequences, i e targeted sequence modifications Additions and deletions may be as small as 1 nucleotide or may range up to about 2 to 4 kilobases or more In this way, site directed modifications may be done in a variety of systems for a variety of purposes
In a preferred application, a targeting polynucleotide is used to repair a mutated sequence of a structural gene by replacing it or converting it to a wild-type sequence (e g , a sequence encoding a protein with a wild-type biological activity) For example, such applications could be used to convert a sickle cell trait allele of a hemoglobin gene to an allele which encodes a hemoglobin molecule that is not susceptible to sickhng, by altering the nucleotide sequence encoding the $-subunιt of hemoglobin, so that the codon at position 6 of the $-subunιt is converted fromVal$6->Glu$6 (Shesely et al (1991) op cit ) Other genetic diseases can be corrected, either partially or totally, by replacing, inserting, and/or deleting sequence information in a disease allele using appropriately selected exogenous targeting polynucleotides For example but not for limitation, the )F508 deletion in the human CFTR gene can be corrected by targeted homologous recombination employing a recA-coated targeting polynucleotide of the invention
For many types of in vivo gene therapy to be effective, a significant number of cells must be correctly targeted, with a minimum number of cells having an incorrectly targeted recombination event To accomplish this objective, the combination of (I) a targeting polynucleotιde(s), (2) a recombinase (to provide enhanced efficiency and specificity of correct homologous sequence targeting), and (3) a cell- uptake component (to provide enhanced cellular uptake of the targeting polynucleotide), provides a means for the efficient and specific targeting of cells m vivo, making in vivo homologous sequence targeting, and gene therapy, practicable
Several disease states may be amenable to treatment or prophylaxis by targeted alteration of heptocytes m vivo by homologous gene targeting For example and not for limitation, the following diseases, among others not listed, are expected to be amenable to targeted gene therapy hepatocellular carcinoma, HBV infection, familial hypercholesterolemia (LDL receptor defect), alcohol sensitivity (alcohol dehydrogenase and/or aldehyde dehydrogenase insufficiency), hepatoblastoma, Wilson's disease, congenital hepatic porphyπas, inherited disorders of hepatic metabolism, ornithine transcarbamylase (OTC) alleles, HPRT alleles associated with Lesch Nyhan syndrome, etc Where targeting of hepatic cells in vivo is desired, a cell-uptake component consisting essentially of an asialoglycoprotem-poly-L- lysme conjugate is preferred The targeting complexes of the invention which may be used to target hepatocytes jn vivo take advantage of the significantly increased targeting efficiency produced by association of a targeting polynucleotide with a recombinase which, when combined with a cell-targeting method such as that of WO92/05250 and/or Wilson et al (1992) J Biol Chem 267 963, provide a highly efficient method for performing in vivo homologous sequence targeting in cells, such as hepatocytes
In a preferred embodiment, the methods and compositions of the invention are used for gene inactivation That is, in addition to correcting disease alleles, exogenous targeting polynucleotides can be used to inactivate, decrease or alter the biological activity of one or more genes in a cell (or transgenic nonhuman animal) This finds particular use in the generation of animal models of disease states, or in the elucidation of gene function and activity, similar to Aknock out® experiments These techniques may be used to eliminate a biological function, for example, a galT gene (alpha galactosyl transferase genes) associated with the xenoreactivity of animal tissues in humans may be disrupted to form transgenic animals (e g pigs) to serve as organ transplantation sources without associated hyperacute rejection responses Alternatively, the biological activity of the wild-type gene may be either decreased, or the wild-type activity altered to mimic disease states This includes genetic manipulation of non-coding gene sequences that affect the transcription of genes, including, promoters, repressors, enhancers and transcriptional activating sequences
Once the specific target genes to be modified are selected, their sequences may be scanned for possible disruption sites (convenient restriction sites, for example) Plasmids are engineered to contain an appropriately sized gene sequence with a deletion or insertion in the gene of interest and at least one flanking homology clamp which substantially corresponds or is substantially complementary to an endogenous target DNA sequence Vectors containing a targeting polynucleotide sequence are typically grown in E coli and then isolated using standard molecular biology methods, or may be synthesized as oligonucleotides Direct targeted inactivation which does not require vectors may also be done When using microinjection procedures it may be preferable to use a transfection technique with linearized sequences containing only modified target gene sequence and without vector or selectable sequences The modified gene site is such that a homologous recombinant between the exogenous targeting polynucleotide and the endogenous DNA target sequence can be identified by using carefully chosen primers and PCR, followed by analysis to detect if PCR products specific to the desired targeted event are present (Erhch et al , (1991 ) Science 252 1643, which is incorporated herein by reference) Several studies have already used PCR to successfully identify and then clone the desired transfected cell lines (Zimmer and Gruss, (1989) Nature 338 150, Mouellic et al , (1990) Proc Natl Acad Sci USA 87 4712. Sheselv et al . (1991) Proc Natl Acad Sci USA 88 4294, which are incorporated herein by reference) This approach is very effective when the number of cells receiving exogenous targeting polynucleotιde(s) is high (i e , with microinjection, or with liposomes) and the treated cell populations are allowed to expand to cell groups of approximately 1 x 104 cells (Capecchi, (1989) Science 244 1288) When the target gene is not on a sex chromosome, or the cells are derived from a female, both alleles of a gene can be targeted by sequential inactivation (Mortensen et al . (1991 ) Proc Natl Acad Sci USA 88 7036)
In addition, the methods of the present invention are useful to add exogeneous DNA sequences, such as exogeneous genes or extra copies of endogeneous genes, to an organism As for the above techniques, this may be done for a number of reasons, including to alleviate disease states, for example by adding one or more copies of a wild-type gene or add one or more copies of a therapeutic gene, to create disease models, by adding disease genes such as oncogenes or mutated genes or even just extra copies of a wild-type gene, to add therapeutic genes and proteins, for example by adding tumor suppressor genes such as p53, Rb1 , Wt1 , NF1 , NF2, and APC, or other therapeutic genes, to make superior transgenic animals, for example superior livestock, or to produce gene products such as proteins, for example for protein production, in any number of host cells Suitable gene products include, but are not limited to, Rad51 , alpha-antitrypsin, casein, hormones, antithrombin III, alpha glucosidase, collagen, proteases, viral vaccines, tissue plaminogen activator, monoclonal antibodies, Factors VIII, IX, and X, glutamic acid decarboxylase, hemoglobin, prostaglandm receptor, lactoferπn, calf intestine alkaline phosphatase, CFTR, human protein C, porcine liver esterase, urokmase, and human serum albumin
Thus, in a preferred embodiment, the targeted sequence modification creates a sequence that has a biological activity or encodes a polypeptide having a biological activity In a preferred embodiment, the polypeptide is an enzyme with enzymatic activity
In addition to fixing or creating mutations involved in disease states, a preferred embodiment utilizes the methods of the present invention to create novel genes and gene products Thus, fully or partially random alterations can be incorporated into genes to form novel genes and gene products, to produce rapidly and efficiently a number of new products which may then be screened, as will be appreciated by those in the art
In a preferred embodiment, the compositions and methods of the invention are useful in site-directed mutagenesis techniques to create any number of specific or random changes at any number of sites or regions within a target sequence (either nucleic acid or protein sequence), similar to traditional site-directed mutagenesis techniques such as cassette mutagenesis and PCR mutagenesis Thus, for example, the techniques and compositions of the invention may be used to generate site specific variants in any number of systems, including E coli, Bacillus, Archebacteπa, Thermus, yeast (Sacchromyces and Pichia), insect cells (Spodoptera, Tnchoplusia, Drosophila), Xenopus, rodent cell lines including CHO, NIH 3T3 and primate cell lines including COS, or human cells, including HT1080 and BT474, which are traditionally used to make variants The techniques can be used to make specific changes, or random changes, at a particular site or sites, within a particular region or regions of the sequence, or over the entire sequence
In this and other embodiments, suitable target sequences include nucleic acid sequences encoding therapeutically or commercially relevant proteins, including, but not limited to, enzymes (proteases, recombinases, lipases, kinases, carbohyd rases, isomerases, peptides tautomerases, nucleases etc ), hormones, receptors, transcription factors, growth factors, antibodies, cytokmes, globin genes, immunosupppressive genes, tumor suppressors, oncogenes, complement-activating genes, milk proteins (casein, "-lactalbumin, β-lactoglobuhn, whey proteins, serum albumin), immunoglobuhns, urine proteins, milk proteins, esterases, pharmaceutical proteins and vaccines
In a preferred embodiment, the methods of the invention are used to generate pools or libraries of variant nucleic acid sequences, and cellular libraries containing the variant libraries Thus, in this embodiment, a plurality of targeting polynucleotides are used The targeting polynucleotides each have at least one homology clamp that substantially corresponds to or is substantially complementary to the target sequence Generally, the targeting polynucleotides are generated in pairs, that is, pairs are made of two single stranded targeting polynucleotides that are substantially complementary to each other (i e a Watson strand and a Crick strand) However, as will be appreciated by those in the art, less than a one to one ratio of Watson to Crick strands may be used, for example, an excess of one of the single stranded target polynucleotides (i e Watson) may be used Preferably, sufficient numbers of each of Watson and Crick strands are used to allow the majority of the targeting polynucleotides to form double D-loops, which are preferred over single D-loops, as outlined above In addition, the pairs need not have perfect complementarity, for example, an excess of one of the single stranded target polynucleotides (i e Watson), which may or may not contain mismatches, may be paired to a large number of variant Crick strands, etc Due to the random nature of the pairing, one or both of any particular pair of single-stranded targeting polynucleotides may not contain any mismatches However, generally, at least one of the strands will contain at least one mismatch
The plurality of pairs preferably comprise a pool or library of mismatches The size of the library will depend on the number of residues to be mutagenized, as will be appreciated by those in the art Generally, a library in this instance preferably comprises at least 40% different mismatches, with at least 30% mismatches being preferred and at least 10% being particularly preferred That is, the plurality of pairs comprise a pool of random and preferably degenerate mismatches over some regions or all of the entire targeting sequence As outlined herein, Amismatches® include substitutions, insertions and deletions Thus, for example, a pool of degenerate variant targeting polynucleotides covering some, or preferably all, possible mismatches over some region are generated, as outlined above, using techniques well known in the art Preferably, but not required, the variant targeting polynucleotides each comprise only one or a few mismatches (less than 10), to allow complete multiple randomization, as outlined below
As will be appreciated by those in the art, the introduction of a pool of variant targeting polynucleotides (in combination with recombinase) to a target sequence, either in vitro to an extrachromosomal sequence or in vivo to a chromosomal or extrachromosomal sequence, can result in a large number of homologous recombination reactions occuπng over time That is, any number of homologous recombination reactions can occur on a single target sequence, to generate a wide variety of single and multiple mismatches within a single target sequence, and a library of such variant target sequences, most of which will contain mismatches and be different from other members of the library This thus works to generate a library of mismatches In a preferred embodiment, the variant targeting polynucleotides are made to a particular region or domain of a sequence (i e a nucleotide sequence that encodes a particular protein domain) For example, it may be desirable to generate a library of all possible variants of a binding domain of a protein, without affecting a different biologically functional domain, etc Thus, the methods of the present invention find particular use in generating a large number of different variants within a particular region of a sequence, similar to cassette mutagenesis but not limited by sequence length In addition, two or more regions may also be altered simultaneously using these techniques Suitable domains include, but are not limited to, kmase domains, nucleotide-binding sites, DNA binding sites, signaling domains, structural domains, receptor binding domains, transcriptional activating regions, promoters, origins, active enzyme domains, dimeπzing domains, leader sequences, terminators, localization signal domains, and, in immunoglobulm genes, the complementaity determining regions (CDR), Fc, VH and VL
In a preferred embodiment, the variant targeting polynucleotides are made to the entire target sequence In this way, a large number of single and multiple mismatches may be made in an entire sequence
Thus for example, the methods of the invention may be used to create superior recombinant reporter genes such as lacZ, luiciferase and green fluorescent protein (GFP), superior antibiotic and drug resistance genes, superior recombinase genes, superior recombinant vectors, and other superior recombinant genes and proteins, including peptides, immunoglobuhns, vaccines or other proteins with therapeutic value For example, targeting polynucleotides containing any number of alterations may be made to one or more functional or structural domains of a protein, and then the products of homologous recombination evaluated
Once made and administered to target cells, the target cells may be screened to identify a cell that contains the targeted sequence modification This will be done in any number of ways, and will depend on the target gene and targeting polynucleotides, as will be appreciated by those in the art The screen may be based on phenotypic, biochemical, genotypic, or other functional changes, depending on the target sequence In an additional embodiment, as will be appreciated by those in the art, selectable markers or marker sequences may be included in the targeting polynucleotides to facilitate later identification
In a preferred embodiment, kits containing the compositions of the invention are provided The kits include the compositions, particularly those of libraries or pools of degenerate cssDNA probes, along with any number of reagents or buffers, including recombinases, buffers, ATP, etc The broad scope of this invention is best understood with reference to the following examples, which are not intended to limit the invention in any manner All references cited herein are expressly incorporated by reference
EXPERIMENTAL EXAMPLES
EXAMPLE 1
Homologous Targeting of recA-Coated Chemically-Modified Polynucleotides in Cells
Homologously targeted exogenous targeting polynucleotides specifically target human DNA sequences in intact nuclei of metabolically active cells RecA-coated complementary exogenous targeting polynucleotides were introduced into metabolically active human cells encapsulated in agarose microbeads and permeabihzed to permit entry of DNA/protem complexes using the Jackson- Cook method (Cook, P R (1984) EMBO J 3 1837, Jackson and Cook (I985) EMBO J 4 919, Jackson and Cook (1985) EMBO J 4 913, Jackson and Cook (1986) J Mol Biol 192 65, Jackson et al (1988) J Cell Sci 90 365, which are incorporated herein by reference) These experiments were designed to specifically target homologous DNA sequences with recA protein in intact nuclei of metabolically active human HEp-2 cells
Jackson and Cook previously demonstrated that the nuclear membranes of human or other cells may be permeabihzed without loss of metabolic function when the cells are first encapsulated in a gel of agarose microbeads The agarose microbead coat contains the cell constituents and preserves native conformation of chromosomal DNA, while permitting diffusion of macromolecules into and out of the cell compartment Wittig et al (1991) Proc Natl Acad Sci (U S A ). 88 2259, which is incorporated herein by reference, demonstrated that monoclonal antibodies directed against left-handed Z-DNA could be diffused into these agarose-embedded cells, and that the antibodies were specifically targeted to chromosomal sequences and conformations In a similar manner, we incubated biotin- or FITC-labeled complementary DNA targeting polynucleotides coated with recA with agarose-coated cell nuclei and verified the correct homologous targeting of the exogenous targeting polynucleotides to specific predetermined human DNA sequences in cell nuclei of metabolically active cells
RecA-mediated homologous gene targeting with complementary oligonucleotides in intact human cell nuclei was verified directly by homologous targeting using targeting polynucleotides that were biotinylated These were subsequently labeled with a fluorescent reporter compound to verify homologous pairing at specific locations having the predetermined sequence(s) RecA-coated targeting polynucleotides for human chromosome 1 peπcentrometπc alpha-satellite DNA sequences were specifically targeted to chromosome 1 centromere sequences in living human cell nuclei that were permeabihzed and suspended in agarose
In these experiments, recA-coated biotinylated exogenous targeting polynucleotides containing homologous sequences to human chromosome 1 alpha satellite DNA were incubated with human HEp-2 cells The cells were embedded in agarose, then treated with standard buffers (according to Jackson and Cook, op cit ) to remove the cytoplasmic membrane and cytoplasm immediately before the addition of targeting polynucleotide coated with recA protein
The experiments were performed with the following results
First, in order to test protocols to be used in nuclear encapsulation, freshly trypsmized growing human HEp-2 tumor cells were suspended in complete DMEM encapsulated in a mixture of agarose (2 5%, Fisher-Biotech) and complete DMEM media adapting the protocols of Nilsson et al , 1983, so that the final agarose concentration was 0 5% (4 volumes cells in suspension with 1 volume 2 5% agarose), and the final cell concentration range was approximately 2 4 x 107 to 8 x 105 The encapsulated cells in agarose "beads® were placed in petri dishes to which DMEM complete media was added and were allowed to grow for 24 hr in an incubator at 37°C , 7% C02 At 24 hr, the cells were clearly growing and multiplying and thus were alive and metabolically active
An aliquot of agarose containing cells (in beads in DMEM medium) was treated to remove the cytoplasmic membrane and cytoplasm by addition of ice-cold sterile PBS, New Buffer (Jackson et al (1988) op cit . 130 mM KC1 , 10 mM Na2HP04, 1 mM MgC12, 1 mM Na2ATP, and 1 mM dithithreitol, pH 7 4 ), New Buffer with 0 5% Tπton-X 100, New Buffer with 0 2% BSA, then was centπfuged at low speed using protocols developed by Jackson and Cook, 1985 and 1986 op cit . Wittig et al (1989) J_ Cell Biol 108 755, Wittig et al (1991 ) op cit ) who have shown that this treatment allows the nuclear membrane to remain morphologically intact The nuclei are metabolically active as shown by a DNA synthesis rate of 85 to 90% compared with that of untreated control cells
Cytoplasm was effectively removed by the above treatment, and the encapsulated nuclei were intact as demonstrated by their morphology and exclusion of 0 4% trypan blue Nuclei in agarose were returned to the humidified C02 incubator at 37°C for 24 hr and remained metabolically active We observed that sterile mineral oil used in the emulsification process was difficult to remove entirely and interfered with the microscopic visualization of suspended nuclei Therefore, the cell-agarose suspension process was simplified In subsequent experiments cells were gently vortexed with melted (39°C) agarose, then the agarose-celi mixture was steπlely minced before New Buffer treatments This simpler process, eliminating the oil step, makes it easier to visualize the cells and chromosomes at the completion of reactions
After mincing of the agar and New Buffer treatments of the cells, the above protocols were used to homologously target endogenous DNA sequences in encapsulated nuclei as follows 16 5 μl recA- coated (or non-recA-coated control) nick-translated DNA (labeled with bιotιn-14-dATP) targeting polynucleotide was prepared and bound under standard native recA protocols (see U S S N 07/755,462 and 07/910,791) Minced agarose fragments were centπfuged and New Buffer supernatant removed The fragments were resuspended in 1 X AC buffer in a 1 5-ml Eppendorf tube, then centπfuged for removal of the buffer (leaving an estimated 50 to 75 μl of buffer), and prepared targeting polynucleotide was mixed with the fragments of agarose-contaming nuclei Reactions were incubated in a 37°C water bath for 2 to 4 hr, then washed, incubated in standard preblock solution, then in preblock supplement with 10 μg/ml FITC-avidm (Vector, DCS grade), and again washed Experimental results were analyzed by placing a minute amount of a reaction with 3 to 4 μl antifade on a slide with a slide cover and viewing it by using the Zeiss CLSM-10 confocal laser scanning microscope (CLSM) Completed reactions were also stored refrigerated for later examination
In the first jn vivo experiment, metabolically active HEp-2 cells suspended in 1 x PBS were encapsulated in agarose by gentle vortexing, treated using New Buffer protocols, then incubated for 3 hr 15 mm with 100 ng of recA-coated targeting polynucleotide specific for Chromosome 1 alpha- satellite DNA biotinylated with bιo-14-dATP by nick translation (BRL, Nick Translation System) using pUC 1 77 plasmid DNA (a 1 77 kb long EcoRI fragment of human DNA in the vector pUC9, Cooke et al (1979) Nucleic Acids Res 6 3177, Emmerich et al (1989) Exp Cell Res 181 126) We observed specific targeting by the alpha-satellite targeting polynucleotide to peπcentromeπc chromosome 1 targets in intact nuclei of metabolically active cells The signals were essentially identical to those using the same targeting polynucleotide with methanol (or ethanol) fixed HEp-2 cell targets in suspension Figure 1 shows specific targeting signals in several metabolically active cells from this experiment
In the second m vivo experiment, cells suspended in incomplete DMEM media instead of 1 x PBS were encapsulated in agarose and treated with 62 5 ng of the same targeting polynucleotide used in the first experiment described above and 62 5 ng of a freshly biotinylated targeting polynucleotide prepared under the same protocols In this experiment, the minced agarose fragments were not resuspended in 1 x AC buffer before addition of targeting polynucleotide and some nuclei disintegrated, especially with subsequent centrifugation The results show that in the nuclei that remained intact, the targeting polynucleotides coated with recA specifically targeted predetermined human DNA targets In contrast, targeting polynucleotides in control reactions without recA did not target the human DNA sequences
Thus, the recA-coated targeting polynucleoπdes were targeted to the repetitive alpha satellite sequences of chromosome 1 This result showed DNA targeting in intact nuclei to specific human chromosome 1 sequences (data not shown)
In the third experiment, cells were suspended in 1 x PBS or in incomplete DMEM media before vortexing with agarose and were tested using 62 5 ng of targeting polynucleotide in reactions with and wtthout recA protein in addition, the reactions were divided in half and washed and FITC-avidm treated in either buffer adjusted to pH 7 or pH 7 4 Cells were incubated with the recA coated targeting polynucleotide for 3 hr 25 mm Live nuclei treated with targeting polynucleotide alone without recA showed no signals In the recA-treated reactions, relatively weaker signals were observed in nuclei incubated in 1 x PBS, whereas very strong specific signals were present in nuclei that had been incubated in incomplete DMEM There was clearly significantly more signal present in nuclei that were washed and treated with FITC-avidm at pH 7 4 compared with nuclei incubated at pH 7 0 Figure 4 shows nuclei that were treated with recA coated targeting polynucleotides and incubated at both pH 7 4 and 7 0
In a fourth experiment, HEp-2 cells were embedded in agarose prepared with I x PBS, New Buffer treated, then treated with 100 ng of biotinylated targeting polynucleotide complementary to chromosome 1 alpha-satellite DNA Controls in this experiment also included reactions without recA protein and additional control reactions supplemented with an identical amount of BSA protein to replace the recA protein Additionally, cells were also embedded in agarose prepared with I x AC buffer Examples of specific targeting to endogenous target sequences were recorded
In a fourth experiment, we directly determined if the embedded nuclei under the conditions used above were metabolically active The nuclei in agarose were incubated with bιo-21-rUTP in complete medium, then incubated for 2 days in the humidified C02 atmosphere After 2 days at 37°C, the cells were examined Bιo-21-rUTP was incorporated in RNA and incubated with FITC-streptavidm FITC was specifically associated with nucleo indicative of πbosomal RNA biosynthesis, thus directly showing metabolic activity in these human cells Similar results were obtained using DNA precursors to measure DNA synthesis In this experiment it was clear that the majority of nuclei in the PBS agarose reaction had condensed chromosomes There was nuclear activity in a number of these nuclei also, indicative of full metabolic viability, which was also shown in the AC buffer-treated cells
A fifth experiment was performed using, again, HEp-2 cells embedded in agarose Final concentration of the cells in agarose was 3 7 x l06/ml The cells were suspended in 1 x PBS prior to combining with agarose The final agarose concentration was 0 5% There were two reactions, one in which recA was used to coat targeting polynucleotide, the second in which recA protein was replaced by BSA at the same protein concentration followed by New Buffer treatments to remove the cytoplasm The nuclei in agarose were incubated for 3 hr with targeting polynucleotide, then processed for detection of correctly targeted polynucleotide using the protocols describe previously FITC-avidm was used to visualize the biotinylated targeting polynucleotide at a concentration of 20 μg/ml Results showed that cells with the recA-coated complementary targeting polynucleotide displayed specific signals in 25% or more of the intact nuclei In contrast, the BSA-treated controls (without RecA) did not show any signal
Cells in agarose from this experiment were further incubated at 37°C in the C02 incubator in complete medium At 22 hr, these cells were metabolically active Chromosomes were condensed, and a number of nuclei were in the process of dividing In these experiments, a significant number of the cells incubated with recA-coated complementary targeting polynucleotides showed specific signal, whereas 0% of the cells incubated with targeting polynucleotide alone showed specific signal
In summary, recA-coated biotinylated targeting polynucleotides for human chromosome 1 alpha- satellite DNA were specifically targeted to human HEp-2 epithelial carcinoma chromosomal DNA in intact cell nuclei of metabolically active cells that had been suspended in agarose, then treated with buffers and recA-coated targeting polynucleotides under suitable reaction conditions (supra and U S S N 07/755,462, U S S N 07/755,462, and U S S N 07/520,321 , incorporated herein by reference) Specific binding by the recA-coated targeting polynucleocide to chromatin alpha-satellite DNA was observed only in the agarose embedded nuclei which were incubated with recA-coated targeting polynucleotides Control nuclei incubated with targeting polynucleotides in the absence of recA and/or with nonspecific protein exhibited no signal
Targeting of Human p53 Gene
We performed recA-mediated homologous targeting of biotinylated targeting polynucleotides that were homologous to the human p53 tumor suppressor gene, and compared the results to targeting of alpha satellite DNA sequences in human chromosome 1 In these experiments, exponentially growing cells were trypsmized, washed, suspended in incomplete medium and encapsulated in agarose The agarose was minced into pieces with a razor blade and the encapsulated cells were treated with New Buffer A sample from each group was removed to verify that nuclei were intact
Nuclei were washed in 1 x AC buffer and incubated with recA-coated complementary single-stranded DNA oligonucleotides (i e , exogenous targeting polynucleotides) for 3 5 hours at 37°C The alpha satellite DNA targeting polynucleotides for chromosome 1 were previously described and were nick- translated with biotinylated deoxyribonucleotides (bιo-14-dATP) The p53 tumor suppressor gene polynucleotide was obtained from Oncor (209 Perry Parkway, Gaithersburg, MD 20877) and is a 1 2 kilobase cDNA fragment from a wild-type human p53 gene (Fields and Jang, (1990) Science 242 1046, Miller et al (1986) Nature 319 783, Zakut-Houre et al (1985) EMBO J 4 1251 ) The 1 2 kilobase human p53 DNA was nick-translated with biotinylated deoxyribonucleotides and yielded a population of biotinylated targeting polynucleotides having a size range (about 100 to 600 nucleotides) similar to that obtained for the human chromosome 1 alpha satellite targeting polynucleotides The targeting polynucleotides were separately incubated with encapsulated cells Following incubation 3 washes of 1 75 x SSC were done, and sampled nuclei were verified as intact after the washing step After washing, the targeted encapsulated cell nuclei were incubated in preblock and FITC-avidm was added to preblock buffer to a final concentration of 20 μg/ml for 15 minutes in the dark The targeted encapsulated cell nuclei were washed sequentially in 4 x SSC, 4 x SSC with 0 1% Triton X-100, and then 4 x SSC Samples of nuclei were again taken and used to verify that the targeted nuclei were metabolically active Microscopic examination showed that metabolically active cells contained specific FITC-targetmg polynucleotide targeted endogenous sequence complexes (shown in Figure 2) The p53 targeting polynucleotides were specifically targeted to human chromosome 17, the location of the endogenous human p53 gene sequences, indicating specific pairing of a targeting polynucleotide to a unique endogenous DNA target sequence The human chromosome 1 alpha satellite DNA was also specifically targeted to the chromosome 1 peπcentromeπc satellite sequences
The experiments validated a highly specific DNA targeting technique for human or other cells as evidenced by homologous sequence targeting techniques in metabolically active cells The targeting technique employs the unique properties of recA-mediated DNA sequence targeting with single- stranded (complementary) short targeting polynucleotides Native intact nuclei were incubated with labeled, heat-denatured targeting polynucleotides coated with recA protein The DNA hybridized to the predetermined targeted homologous sequences In these experiments, the targeting polynucleotides formed paired complexes with specific gene sequences within metabolically active cell nuclei This in vivo targeting by recA-mediated homologous targeting polynucleotides shows the targeting specificity and therapeutic potential for this new in vivo methodology Application of recA or other recombmase-mediated targeting of (complementary) ssDNA or denatured dsDNA targeting polynucleotides to predetermined endogenous DNA targets is important for gene eatry, gene knockout, gene replacement, and gene mutation or correction
EXAMPLE 2 Correcting a Mutant Gene to Produce a Functional Gene Product Homologously targeted complementary DNA oligonucleotides were used to correct 11 bp insertion mutations in vector genes and restore vector gene expression and vector protein function in microinjeαte mammalian cells
Expeπmentswere designed to testwhether homologously targeted complementary 276-bp oligonucleote! targeting polynucleotides could correct an 11-bp insertion mutation in the lacZ gene of a mammahalMA vector, which encoded a nonfunctional $-galactosιdase, so that a corrected lacZ gene encoded and expressed a functional enzyme Functional enzyme ($-galactosιdase) was detected by an X-gal assay that turns cells expressing a revertant (i e , corrected) lacZ gene a blue color
NIH3T3 cells microinjected with the mutant test vector bearing an 11 basepair insertion mdflacZ coding sequence do not produce any detectable functional $-galactosιdase enzyme In contrast, cells microinjected with the wild type test vector do produce functional enzyme
We obtained the functional lac plasmid pMCIIacpA for use as a positive control for expression of $- galactosidase pMCI IacXpA is the target test mutant plasmid (shown in Figure 3) It is identical to pMCI IacpA (shown in Fιgure4) but has a 11-bp Xbal linker insertional mutation This plasmid does not express $-galactosιdase activity in rouse NIH3T3 cells when introduced by electroporation It does not produce blue color in the presence of X-gal indicative of $-galactosιdase production following vector micro-injection Negative controls with mock or noninjected cells we also done Using these conditions and NIH3T3 cells have no detectable background blue staining
The plasmid pMC1 lacpA (8 4 kb) contains the strong polyoma virus promoter of transcription plus ATG placed in front of the lacZ gene The polyadenylation signal from SV40 virus was placed in back of the lacZ gene The plasmid vector was plB130 from IBI (New Haven, CT) The mutant vector pMCIIacpA has a 11-bp inserton in the Xbal site consisting of the inserted sequence CTCTAGACGCG (see Figure
5)
In several control micro-injection experiments using pMC1 lacXpA weonsistently failed to detect any ble microinjected cells In contrast, in various experiments monitored early after microinjection aptpjimately
9 to 13% of the NIH3T3 cells injected with pMCIIacpA HA expressed $-galactosιdase as evidenced by their blue color No cells microinjected with injection buffer alone omock injected were observed as blue
We synthesized two 20-bp primers (PCR" and PCR$) for producing a 276-bp PCR product (see Figure 5) from the wild-type lacZ sequence for use as targeting polynucleotides We chose this 276-bp fnarapnt to span the 11 bp insertion mutation as a nonhomologous sequence The 276-bp DNA oligonucleotide was separated by gel electrophoresis and electroeluted from agarose, ethanol precipitated, and its concentration determined by absorbance at 260 nm The 276-bp fragment was 5' end-labeled with 32P and specifically D-looped with the pMd lacXpA or pMCIIacpA plasmid DNA using recA as shown by agarose gel electrophoresis
Experiments were designed to test for $-galactosιde production in cells microinjected with pMd lacXpA vectors with targeting polynucleotide-target complexes using complementary 276-bp oligonucleotide targeting polynucleotide treated with recA The 276-mer targetg polynucleotides in 1 X TE buffer were denatured by heating at I00°C for 5 mm and immediately quenched in an ice bath for 1 m The DN A solution was collected at 4°C by centrifugation RecA-mediated targeting polynucleotide reactions containing a final volumeof 10 μl were assembled using 1 0 μl 10 x AC buffer, 1 5 μl 16 mM ATP(S, 3 8 μl dd H,0, 1 2 μl recA protein solution (13 μg/μl), and 2 5 μl of a 30 μg/ml stock of healenatured 276-bp targeting polynucleotide The recA protein was allowedctcoat the DNA for 10 mm at 37°C Next, 1 0 μl of 10 x AC buffer, 10 μl of 0 2 M magnesium acetate, 1 3 μl of pMCIIacXpA (1 0 μg/μl), and 6 7 μl of dd H20 was added to a final volume of 20 μl Control reactions werperformed without added recA protein
NIH3T3 cells were capillary needle microinjected with targeting polynucleotide-target DNA mixtures ade in glass pipettes freshly pulled into microneedles using a Sutter instruments microprocessor controlled apparatus An ECET Eppendorf microinjection pump ad computerized micromanipulator were used for computer-assisted microinjection using an Olympus IMT-2 inverted microscope Cells were carefully microinjected under controlled pressure and time NIH3T3 cells injected with pMCIIacpA showed approximately 9% of the injected cells were blue None (0%) of the celfeijected with pMCIIacXpA DNA in reactions containing the 271 bp oligonucleotide but without recA protein showed a blue color In marfeie contrast, approximately 3 6% of the cells microinjected with the recA-coated 271-bp targeting polynucleotide targeted to-the pMCIIacXpA target hybd were blue (Figure 6), indicating that the mutant pMCIIacXpA gene can be targeted and corrected by the 271-bp oligonucleotide, whichas been targeted with recA-coated targeting polynuα&otides In summary, these measurements show that the 11 bp Xba I insertion mutation can be corrected with the recA-mediated targeted corrected m vivo, but not with the 271-bp oligonucleotide alone Notethat the m situ identification of 3T3 cells expressing $-galactosιdase was performed following incubation with X-gal (5-bromo-4-chloro-3-ιndolyl-$- galactopyranoside) (Sigrηa) as described by Fischσ et al (1988) Nature 332 853, Price et al (1987) Proc Natl Acad Sci (U S A ) 84 156. Lim and Chae (1989) BioTechniques 7 576
EXAMPLE 3 Correcting a Human CFTR Disease Allele
Homologously targeted complementary DNA hgonucleotides were used to correct a naturally occurring 3 bp deletion mutation in a human CFTR allele and restore expression of a functional CFTR protein in targeted mammalian cells A major goal of cystic fibrosis (CF) gene therapy is the correction of mutant portions of the CF transmembrane conductance regulator (CFTR) gene by replacement with wild-type DNA sequences to restore the normal CFTR protein and lortransport function Targeting polynucleotides that were coated with recA protein were introduced into transformed CF airway epithelial cells, homozygous for bothers ΔF508 CFTR gene mutation, by either intranuclear microinjection, eleαtporation, or by transfection with a protein-DNA-hpid complex
Isolation and characterization of the CFTR gene (Rommens et al (1989) Science 245 1059, Riordan et al (1989) Science 245 1066, incorporated herein by reference) has been crucial for understanding the biochemical mechanιsm(s) underlying CF pathology The most common mutation associated with CF , a 3-base-paιr, in-frame deletion eliminating a phenylalanme at ammo acidqsition 508 (ΔF508) of CFTR, has been found in about 70% of all CF chrcnosomes (Kerem et al (1989) Science 245 1073, Kerem et al (1990) Proc Natl Acad Sci (U S A )87 8447) Correction ofΔF508 and other CFTR DNA nutations lies at the basis of DNA gene therapy for CF disease Elimination of the cAMP-dependent C1 ion transport defect associate! with CFTR gene mutations has been accomplished through the introduction of the transcribed portion of wild-type CFTR cDSIA into CF epithelial cells (Rich et al (1990) Nature 347 358, Drumm et al (1990) CeH 62 1227)
An immortalized CF tracheobronchial epithelial humaπcell line, ECFTE290-, is homozygous for theΔF508 mutation (Kunzelmann et al (1993) Am J Respir Cell Mol Biol . 8 522) These cells are useful as targets for homologous recombination analysis, because they contain the same 3 basepair deletion in CFTR allele on all copies of chomosome 7 Replacement of the ΔF508 allele with wild-type CFTR DNA in indicated only when homologous recombination has occurred The 491 bp region of the CFTR gene spanning exon 11 and containing 3' and 5' flanking mtron sequences was selected from sequence data published previously (Zielenski et al (1991) Genomics 10 214, incorporated herein by reference) and used as a targeting polynucleotide TheDNA fragment was PCR amplified in preparative quantities and then denatured for introduction into cells as recA-coated complementary ssDNA (or dsDNA) Exponentially growing eels were transfected by intranuclear microinjection and were propagated on the same petri dishes in which they wee microinjected Cells outside the microinjected area were removed by scraping with a rubber policeman Exponentially growing cells were typsmized and washed before electroporation Cells transfected with protein-DNA-hpic omplexes were grown to approximately 70-8ΘO confluence before transfection
The 491 bp fragment was generated by PCR amplification from the T6/20 plasmid (Rommene-t al (1989) op cit . incorporated herein by reference) ad verified by restriction enzyme mapping and propagated as described previously After digestion with EcoRI and Hindlll, a 860 bp insert was isolated following electrophoresis in 0 8% SeaPlaque agarose gel The 860 bp fragmti contained CFTR exon 10, as well as 5' and 3' mtron sequences, as defined by the restriction enzyme cleavagates (Zielenski et al (1991 ) op cit ) A 50 ng aliquot of the fragment was amplified by PCR using primers CF1 and CF5 (Table 1 ) to generate a 491 bp fragment The conditions for amplification were denaturation, 94°C for 1 annealing, 53°C for 30 sec, extension, 72°C for 30 sec witha 4 sec/cycle increase in the extension time for 40 cycles The fragment size was confirmed by electrophoresis on a 1% agarose gel, then amplified in bulk in 20 separate PCR amplifications, each containing 50 ng of target DNA The 491 bp PCR products were extracted with phenol chloroform isoamyl alcohol (25 24 1) extraction and precipted with ethanol DNA precipitates were collected by centπfugabn in an Eppendorf microcentπfuge and resuspended at a final concentration of 1 mg/ml The 491 bp fragment contained exon 10 (193 bp), as well as 5' (163 bp) and 3' (135 bp) flanking mtron sequences, as defined by primers CF1 and CF5
The 491 nucleotide fragments were coaid with recA protein using the reaction buffer of Cheng (Cheng, et al (1988) J Biol Chem 263 15110, incorporated herein by reference) Typically, the 491 bp DNA fragment (5μg) was denatured at 95°C for 10 mn, then added to a 63 μl of coating buffer containing 200 μg of recA protein, 4 8 mM ATP(S, and 1 7 μl reaction buffer (100 mM Tπs-Ac, pH 7 5 at 37°C, 10 m M dithiothreitol, 500 mM NaOAc, 20 mM MgOAc, 50 percent glycerol) and incubated for 10 mm at 37°C Next, the MgOAc concentration was increased to a final concentration of about 22 mM by addition of 7 μl of 200 mM MgOAc Under these conditions, the 491 nucleotide fragment was coated witbcrA protein at a molar ratio of 3 bases per 1 recA molecule After coating the-£fgments were immediately placed on ice at 4°C until transfection (10 mm to 1 hr)
Microinjection, when used, was performed with an Eppendorf 5242 microinjection pump fitted to an Eppendorf 5170 micromanipulator using borosihcate pipete (Brunswick, 1 2 OD x 1 9ID) fabricated into a microneedle with a Sutter instruments (P-87) micropipette puller The micropipettes were filled by capillary force from the opposite side of the needle Approximately 100 pipettes were used for injecting 4000 cells Cells were injected with approximately 1,000-10,000 fragments per cell by intranuclear injection witi 120 hPa for 0 1-0 3 s at a volume of 1-10 fl/nucleus Microinjected cells were viewed with an Olympus IMT-2 inverted microscope during the injection The area of the petri dish containing icjed cells was marked with 2 to 5 mm diameter rings Needlsnicromjection was performed in cells grown on 10 separate 60 mm petri dishes Cells were injected at room temperature in culture medium after two washes in phosphate buffered saline (PBS) After microinjection, nonmjected cells in the culture were removed by scraping Injected cells were grown at 37°C in a humidified incubator at 7 days and then harvested for DNA and RNA
Electroporation experiments were performed using recA-coated 491 -mer ssDNA as described above Approximately 1 x 10? exponentially growing cells were supended in 400μl of coating buffer with 5 μg of recA coated-DNA The cell suspension wa pre-mcubated on ice for 10 mm and electroporated at room temperature with 400 V and 400μF in a BTX 300 electroporator (BTX Corporation, San Diego, CA) Afte electroporation, cells were incub ed on ice for an additional 10 mm, diluted in Eagle's minimal essential medium (MEM) supplemented with 10% fetal bovine serum (FBS) and HDμg/ml streptomycin, 100 U/ml penicillin (Cozens et al (1992Proc Natl Acad Sci (U S A )89 5171 , Gruenert et al (1988) Proc Natl Acad Sci (U S A ) 85 5951 , Kunzelmann, (1992) op cit ). and then seeded in T75 flasks Under these conditions of elecroporation, approximately 30-50% of the cells survive Cells we cultured for 507 days at 37°C and then harvested for DNA and RNA
Protein DNA-lipid complexes (liposomes) were prepared Briefly, dioleoylphosphatidyl-ethanolamine (PtdEtn, DOPE) was used for preparing liposomes by ng 4 μM solutions of the lipid under nitrogen at room temperature The lipid film was rehydrated with 4 ml of 30 mM Tπs-HC1 buffer (pH 9), then sonicated for 15 minutes under an atmosphere or argon The protem-DNA complex was prepared in polystyrene tubes by diluting 20 μg of recA-coated 491-base DNA in 30 mM Tπs-HC1 , (pH 9) buffer Gramicidin S protein (GmS) was also diluted with 30 mM Tπs HC1 (pH 9) to a final concentration of 2 mg/ml from a 20 mg/ml stock solution prepared in dimethyl sulfoxide The protein (40 μg) was added to the DNA and rapidly mixed Next, 175 μl of the hposome solution (175 nmoles αpld) were added to the peptide DNA mixture
Genomic DNA was isolated and purified from cells as described in Maniatisp cit to test for homologous DNA recombination Cellular DNA was firsPCR-amphfied with primers CF1 and CF6 (Table 1 ) CF1 is within the region of homologydefined at the 5' end of the 491 bp CFTR fragment CF6 is outside the regro of homology at the 3' end of this fragment
The conditions for the PCR amplification were as follows CF1/CF6, 684/687 bp fragment, primers, 0 5 μM, DNA, 1-2 μg, denaturation, 94°C for 1 mm, annealing, 53°C for 45 s, extension, 72°C for 90 s wit h a 4-s/cycle increase in extension time for 40 cycles, Mg+2 1 5 mM DNA fragments were separated by agarose electrophoresis and visualized by staining with ethidium bromide, then transferred to Gene Screen Plus filters (DuPont) The DNA was then hybridized with the allele-specific normal CFTW>-end- labeled DNA probe defined by oligo N as described by Cozens et al (1992) op cit . Kunzelmann (1992) op cit , incorporated herein by reference The presence of wild-type (WT) sequences was determined autoradiographically by hybridization with the radiolabeled DNA probe
Homologous recombination was verified ma second round of PCR DNA amplification using the 687/684 bp fragment as a DNA template for amplification The primers used in this allele-specific reaction were CFI and the oligo N or ohgoΔF The size of the DNA fragments was 300 bp (oligo N) or 299 bp (ohgΔF) The conditions for the reaction wereas follows CF1/olιgo N/ΔF, 300/299 bp fragment, primers, 0 5 μM, DNA, 1 -2 μg, denaturation, 95°C for 45s, annealing, 51 °C for 30s, extension, 72°C for 30 s with a 3-s/cy£l increase in extension time for 40 cycles, Mg+2, 1 5 mM In DNA from transfected ECFTE29o- cells, amplified with the CF1/ohgo N primers, a PCR product was detected only if the wild-type CFTR sequenoe were present Amplification with the CFI/ohgcΔF gives a PCR DNA productof DNA targets purified from transfected and nontransfected 3CFTE29o- cells but not for DNA targetssolated from control normal celt (16HBE140-) The presence of wild-type CFTR sequences in the amplified DNA fragments was also determined autoradiographically after hybridization with 32P-5'-end-labeled oligo N as probe
Cytoplasmic RNA was isolated and denatured at 95°C for 2 mm, then reveestranscnbed using the DNA polymerase provided in a PCR RNA Gene Amp kit according to manufacturer's instructions (Perkin- Elmer/Cetus) First strand cDNA was amplified by using primer CF17 at the 5' end of exon 9 and the allele-specific oligo N or ohgoΔF primers The length of the PCR fragments is 322 bp (CF17/ohgo J tand 321 bp (CF17/ohgo ΔF)
The conditions for PCR amplification are CF17/ohgo N/ΔF, 322/321 bp fragment, primers, 1 μM, denaturation, 94°C forl mm, annealing, 51 °C for 30s, extension, 72°C for 20s with a 4-s/cycle increase in extension time for 40 cycles, Mg+2, 0 8 mM DNA fragments were visualized after electrophoresis on ethidium bromide-stained 1 % agarose gels In addition to the allele-specific PCR amplification of first-strand cDNA, Southern hybridization was performed as described above Fragments were transferred to Gene Screen Plus filtes then hybridized with allele-specific oligo N probe under the same conditions used for the Southern analysis of the genomic DNA (Kuretmann et al (1992) op cit . Cozens et al (1992) op cit ) The presence of wild-type CFTR RNA was confirmed by hybridization and autoradiography of RNA extracted from normal (16HBE140-) control DNA and in DNA of transfected 3CFTE29o-cells
Hybridization was performed as described previously (Cozens et al (1992jp cit ) DNA fragments were separated by agarose gel electrophoresis DNA was denatured with 0 4 N NaOHnd 0 6 M NaC1 for 30 m , then washed oncewith 1 5 M NaC1 and 0 5 M Tπs-HC1 for 30 m DNA was transferred to Gene Screen Plus membrane (NEN-DuPont) by capillary blot, again denatured witβ 4 N NaOH for 1 mm, and then neutralized with 0 2 M Tπs-HC1 (pH 7 0) DNA on membranaewas prehybridized for 1 h at 37°C in 6 x SSC, 5 x Denhardt's solution, 1 % SDS, containing 100 μg/ml of denatured salmon sperm DNA (Sigma) Oligonucleotide probes (oligcN or oligo ΔF, 10 ng) were 32P-5'-end-labeled with 20 units of T4 kmase and 40 μCi 32P-(-ATP for 30 mm at 37°C Unincorporated nucleotides were removed by centπfugatDn of the reaction mix through a minispin column (Worthmgton Biochemical Corp , Freehold, NJ) Hybridization was performed overnight at 37°C Membranesvere washed twice for 5 mm each tine in 2 x SSC at room temperature, twice foι30 mm in 2 x SSC, 0 1 % SDS at 45°C, and once in 0 1 x SSC for 30 mm at room temperature After washing, hybrids on membranes were analyzed autoradiographically by exposure to x-ray film
Analysis of 3CFTE29o- DNA shows replacement of the endogenous mutar(ΔF508) sequences with the exogenous normal fragment as evidenced by PCR amplification of genomic DNA and allele-specific Southern blot hybridization PCR primers, one inside (CF1), and one outside (CF6) the region of homology (491 bp), were used to test whether the Hiphfied DNA band was possibly due to amplification of any residual DNA fragment remaining in the cell after the transfection or by possible random DNA integration A 687 bp fragmentcontams normal CFTR sequences while the 684 bp fragment is generate from ΔF508 CFTR DNA To determine whether endogenous ΔF508 sequences were replaced with exogenous normal CFTR sequences, we analyzd aliquots of the 687 or 684 bp amplification fragments by Southern hybridization using 32P-end-labeled DNA probes specific for the ΔF508 or wild-type sequences (Table 1) h addition, the 687 bp fragment was PCR amplified by using the CF6 primer and a primer specific for either ΔF508 (oligo ΔF) or normal sequences (oligo N) The second round of DNA amplification with the CF1/ohgo N or ΔF primer pair combination yields 300/299 bp fragments, respectively With the CF1/ohgo N primer pair combination, a fragment will be detteri only if the mutant DNA has been replaced by normal sequences Furter confirmation of homologous DNA recombination was tested by allele-specific Southern blot hybridization of the 300/299 bp fragments
Analysis of cytoplasmic RNA to detect normal exon 10 sequences in CFTR mRNA, verify that the homologous DNA recombination was legitimate and that normal CFTR mRNA is expressed in the cytoplasm To test whether the PCR geerated DNA fragments were exclusively CFTR mRNA-derived, primers in exon 9 (CF17) and allele-specific (normal, oligo N oΔF508, oligo ΔF) primers in exon 10 Thε amplification with pπmers CF17/N yields a 322 bp normal fragment only if transcription of homologously recombined DNA has occurred A 321 bp DNA fragment would be generated if th6F508 mutation were present Furthermore, Southern hybridization analysis with allele-specific 32P-end-labeled probes differentiated between normal and ΔF508 mutant sequences and were also used to confirm expression of wild-type CFTR mRNA in the cytoplasm
Homologous recombination between the targeting polynucleotide coprismg WT CFTR DNA and ΔF508 mutant cellular DNA allehc targets was evaluated by analysis or cellular DNA and RNA isolated from transfected and nontransfected 3CFTE29o-cell cultures Nuclear genomic DNA and cytoplasmic RNA were isolated 6 days after transfection, CFTR exon I sequences were amphffeby PCR Oligonucleotide primers (Table 1 ) were used to amplify the region of CFTR DNA spanning exon 10 One PCR primer fC I) was within the region of homology defined by the 491 bp DNA fragment (sense primer), and the other (CF 6) was outside the homologous region in the 3' mtron (antisense primer) This DNA amplification reaction produces a 687 bp fragment with normal human CFTR DNA or a 684 bp fragment if the DNA contains the ΔF508 mutation, as shown in Fig 7A Southern hybridization waεarπed out on the 687/681 bp DNA fragments generated from amplification of genomic DNA from cell cultures by microinjection or by transfection with the protem-DNA-lipid complex, shown in Fig 7B A probe consisting of 32P-end- labeled oligonucleotide DNA that hybridized only to DNA sequences generated from a normal exon 1 0 was used DNA from all microinjected and transfected cells produced specific hybrids as evidenced by autoradiographic hybridization For cells microinjected with the 491 nucleotide fragmerffig 7B, lane 2), the present of normal exon 10 sequences indicated homologous replacement at least a frequency of $ 2 5 x 10"4 This result indicates at least one correctly targeted homologous DNA replacement in about 4000 microinjected nuclei Other similar experiments using either electroporation or protem-DNA-lipid transfection to transfer the recA-coated 491 nucleotide CFTR DNA fragments also showed homologous recombination with the normal CFTR sequence in transfected CF cells No hybridization was observed in control nontransfected (or mock-injected 3CFTE29o- cells) In each cell transfectedith normal CFTR DNA, analysis of the genomic DNA in a second round of allele-specific amplification of the 681/684 bp fragments with primers CFI/ohgo N (Table 1 ) clearly showed the 300 bp fragment expecteβHien wild-type CFTR sequences are present, as shown inFig 8A Fragments were detected for control 16HBE14o- ceϋ (Fig 8A, lane 2) and cells transfected with recA-coated DNA (Fig 8A, lanes 5 and) 6 A 299 bp fragment (ΔF508-specιfic primer ends one base closer to the CF1 than the oligo N) was detected in DNA from nontransfected 3CFTE29o- cells amplified with CF1/ohg<ΔF primers (Fig 8A, lane 4) No fragment was detected in DNA from nontransfected 3CFTE29o- cells reamphfied with thelCI/ohgo N primers (Fig 8A, lane 3) Allele-specific Southern blot hybridization of these fragments with the 32P-end-labeled oligo N probe resulted in autoradiographic hybridization signals from control normal and transfected CF cells (Fig 8B, lanes 1 , 4, and 5) but not from DNA of nontransfected CF cells amplified tøii CF1 and ohgo-N or -ΔF (Fig 8B lanes 2 and 3) We tested whether any residual 491 nucleotide DNA fragments, which might remain in the cell after 6 days could act as a primer for the PCReaction, genomic 3CFTE29o- DNA was mcubatedwith an equivalent number of recA-coated DNA'ragments (10 04) introduced by micromjectiσi (Fig 9) One antisense primer contains the wild-type normal (N) sequence while the other contains the ΔF508 (ΔF) mutation Amplification wbh the CFI/ΔF primer combination gives a 299 bp fragments when the ΔF508 mutation is present No DNA fragment product was detected when the CF1/N primer combination we used with control nontransfected 3CFTE29o- DNA (Fig 9, lane 2) However, when the CF1/ΔF primer combination was used for DNA amplification in nontransfected 3CFTE29o- cells, a DNA product of the expected size (299 bp) was produced (Fig 9, lane 1 ) These results indicate that all residual 491 nucleotide DNA fragments which might remain in the cells after 6 days of culture were incapable of competing with the CF1 PCR primers in the PCR arrlrϋcation of the 687/684 bp fragments
Table 1 PCR Primers and Oligonucleotides OliQonuclectide DNA Strand DNA Seguence
CF1 S 5'-GCAGAGTACCTGAAACAGGA
CF5 A 5'-CATTCACAGTAGCTTACCCA
CF6 A 5'-CCACATATCACTATATGCATGC
PCR Primers and Oligonucleotides
Ohgonuclectide DNA Strand DNA Sequence
CF17 S 5'-GAGGGATTTGGGGAATTATTTG
OLITGO N A 5'-CACCAAAGATGATATTTTC
OLIGO aF A 5'-AACACCAAGATATTTTCTT
Notes
(1) CF1 and CF5 PCR primers were used to synthesize the 491 bp fragment used for the- targetmg polynucleotide
(2) CF1 and CF6 PCR primers were used to amplify the 687/684 bp CFTR fragment
(3) The CF17 primer is located at the 5' end of exon 9 and was used for amplification of first strand cDNA derived from CFTR mRNA
(4) Oligo N and Oligo ΔF are allele-specific probes and can also be used as allele-specific PCR primers for amplifying the 300/299 bp fragments (DNA analysis) and the 322/321 bp fragments (RNA analysis)
(5) Sense (S) and antisense (A) primers are designated under DNA Strand and indicate the sense of the strand relative to the transcribed direction (i e , the CFTR mRNA)
The corrected CFTR DNA must also be expressed at the mRNA level for normal function to be restored Therefore, cytoplasmic CFTR mRNA was analyzed for the presence of a normal CFTR RNA sequence in the ΔF508 region of exon 10 Cytoplasmic RNA was isolated from the cells, reverse- transcribed with DNA polymerase and PCR-amphfied as first-strand cDNA This amplification was performed with a PCR primer located in exon 9 (CF17, sense) and CFTR allele-specific PCR primer in exon 10 (oligo N or ΔF, antisense) The exon 10 primer contains the CF mutation site, and the resulting fragment is 322 bp in normal DNA or 321 bp in DNA containing the ΔF508 mutation Amplification of genomic DNA is eliminated by using primers that require amplification across mtron/exon boundaries Amplified cDNA generated from normal control 16HBE140- cells and experimentally transfected cells yielded DNA product fragments with the CF17/ohgo N, whereas nontransfected 3CFTE29o- cells only showed a DNA fragment after amplification with the CF17/ohgo ΔF primers but not with the CF17/ohgo N primers Cells electroporated with wild-type 491 -mer CFTR DNA showed the presence of wild-type CFTR mRNA In addition, protein-DNA-hpid-transfected 3CFTE29o- cell cultures also showed the presence of wild-type CFTR mRNA in cells transfected with the recA-coated 491 nucleotide fragment Southern hybridization of the 322/321 bp cDNA fragments with the 32P-end-labeled N oligonucleotide DNA probe showed the specificity of the PCR amplification and produced specific autoradiographic hybridization signals from all cell cultures transfected with recA-coated 491 nucleotide targeting polynucleotide No autoradiographic hybridization signals were detected in nontransfected 3CFTE29o- cells amplified with the CF17/ohgo N or oligo ΔF primers These analyses verify that the genomic DNA homologously recombined with the WT 49l-mer DNA at the ΔF508 CFTR DNA locus resulting in RNA expressed and transported to the cytoplasm as wild- type CFTR mRNA
This evidence demonstrates that human CF ΔF508 epithelial cells CFTR DNA can homologously recombme with targeting polynucleotides comprising small fragments of WT CFTR DNA resulting in a corrected genomic CFTR allele, and that a recA-coated targeting polynucleotide can be used in transfection reactions in cultured human cells, and that cystic fibrosis ΔF508 mutations can be corrected in genome DNA resulting in the production of normal CFTR cytoplasmic mRNA
Taken together, the data provided indicates that 491 -mer ssDNA fragments can find their genomic homologues when coated with recA protein and efficiently produce homologously targeted intact cells having a corrected gene sequence Analysis of CFTR in cytoplasmic RNA and genomic DNA by allele-specific polymerase chain reaction (PCR) amplification and Southern hybridization indicated wild-type CFTR DNA sequences were introduced at the appropriate nuclear genomic DNA locus and was expressed as CFTR mRNA in transfected cell cultures Thus, in human CF airway epithelial cells, 491 nucleotide cytoplasmic DNA fragments can target and replace the homologous region of CFTR DNA containing a 3 bp ΔF508 deletion
Correctly targeted homologous recombination was detected in one out of one microinjection experiment with recA-coated targeting polynucleotide, two of two different electroporation experiments with recA-coated targeting polynucleotide, and one of one pid-DNA-protein complex transfection experiment with recA-coated targeting polynucleotide Taken together, these 4 separate experiments strongly indicate that homologous recombination with recA-coated targeting polynucleotides (491 -mer CFTR DNA) is feasible for treatment of human genetic diseases, and can be performed successfully by using various methods for delivering the targeting polynucleotide-recombinase complex
EXAMPLE 4 Homologous recombination in procaryotic cells
In order to study the biological consequences of the cssDNA probe target hybrid DNA structures in cells, we developed a simple and elegant assay to rapidly screen for in vivo homologous recombination events in Eschenchia coli The principle of this assay is to screen for the recombinogenocity of hybrid structures formed between a dsDNA plasmid target carrying a 59 bp deletion in the lacZ gene (pRD 59) and cssDNA probes from the wild type lacZ (IP290) gene by introducing these pre-formed protein-free hybrids into E coli by electroporation (Figure 10) Homologous recombination frequencies are scored by plating transformed cultures in the presence of a chromogenic substrate (X-gal) so that recombinant bacterial cells (carrying plasmids that encode a wild type lacZ gene resulting from homologous recombination) appear blue
DNA plasmids and DNA probes The plasmid pRD 59 was made from the 2 9 kb cloning vector pBluescπpt IISK(-) (pRD 0) (Stratagene) The pRD 0 DNA was linearized at a unique EcoRI site in the poiyhnker region of the lacZ gene and digested with mung bean nuclease (Boehπnger-Mannheim) The plasmids were then ligated and transformed into the RecA(-) E coli host XL1-Blue (Stratagene) The resulting alpha peptide mutant clones were screened for lack of alpha-complementation of β- galactosidase activity, which results in white colonies when grown on plates containing X-gal and IPTG (Sambrook et al , 1989) Plasmid DNAs recovered from white colonies by a mini-prep procedure (Qiagen) lacked the unique EcoRI site, as well as the Xhol and Xbal sites These mutant clones were then sequenced using Sanger dideoxy sequencing methods (Sequenase Kit version 2, USB) to determine the length of the deletion Several clones containing deletions ranging from 4 bp to 967 bp were sequenced and named pRD for plasmids with an EcoRI deletion The cloning vector pBluescπpt IISK(-) was named pRD 0 because it does not contain any deletions
All samples of the plasmid DNA were then prepared by the Qiagen Maxi-Prep (Qiagen) procedure from strain of XL1-Blue (Stratagene) containing the plasmids The cultures were grown on Luna-Broth (LB) media (Sambrook, et al , 1989) containing 100 μg/ml ampicilhn Recovered plasmids were more than 90% negatively supercoiled Form I DNA as judged by agarose gel electrophoresis
Biotinylated cssDNA probes were made from a fragment of the normal pBluescπpt IISK(-) plasmid The plasmid DNA was linearized with Bgll and run on a 1% agarose gel in 1X TAE After ethidium bromide staining, the 1 6 kB fragment band was excised from the gel and purified using the Qiaex II gel purification method (Qiagen) This 1 6 kb fragment was diluted 1 20 and then used as a template for PCR The PCR reaction mixture contained bιotιn-14-dATP (GIBCO-BRL) in order to synthesize IP290, a 290 bp biotinylated cssDNA probe homologous to the LacZ region of pRD 0 In addition, pRD 59 was linearized with Bgll and the 1 55 kb fragment was purified in the same manner as the pRD 0 1 6 kb fragment Using the same primers that were used to synthesize IP290, the pRD 59 1 55 kb fragment was used as a template for PCR to synthesize DP231 , a 231 bp biotinylated cssDNA probe homologous to the LacZ region of pRD 59 It is missing the 59 base pair sequence that flanks the EcoRI site Biotinylated cssDNA probe CP443 was made in the same manner except that pRD 0 was linearized with Dral and different primers were used CP443 is completely homologous to pRD 0 and pRD 59 in a region outside of the LacZ gene
RecA mediated cssDNA targeting reactions and purification of probe target DNA hybrids Before targeting, biotinylated cssDNA probes (70 ng) were denatured by heat at 98°C for 10 minutes, cooled immediately in an ice-water bath, and then centπfuged at 4°C for 10 seconds to recover all liquids Reactions without cssDNA probe contained equivalent volumes of water The denatured cssDNA probes were then coated with RecA protein (Boehπnger-Mannheim) in Tπs-acetate reaction buffer (Cheng et al , 1988, 10 mM Tπs-acetate (pH 7 5), 1 mM dithiothreitol, 50 mM sodium acetate, 2 mM magnesium acetate, 5% (v/v) glycerol) with 2 43 mM ATPS for 15 minutes at 37°C in a 10 μl volume Reactions without the RecA protein contained equivalent volumes of RecA storage buffer (20 mM Tris- HCI, pH 7 5, 0 1 mM EDTA, 1 mM DTT, and 20% glycerol)
The RecA mediated targeting reactions were performed by adding 1- 4 μg of the appropriate plasmid DNA in an aqueous solution containing 22 mM magnesium acetate, bringing the final magnesium concentration to 11 mM and the final reaction volume to 20 μl The reaction was incubated for another 60 minutes at 37°C
At the end of the targeting reaction, SDS was added to a final concentration of 1 2% to deprotemize the complexes If further enzymatic treatments were necessary on the targeted complexes, 3 volumes of phenol choloform isoamyl alcohol (Sigma), shaken on a Multi-Tube Vortexer (VWR) for 4 minutes at 4°C, and centπfuged for 5 minutes at 4°C The supernatant was recovered, placed in a new tube, and extracted with 1 volume of chloroform The mixture was shaken for 2 minutes at 4°C, and centπfuged for 5 minutes at 4°C The supernatant was recovered, containing the purified targeted complexes
Detection of probe target DNA hybrids After deprotemization, the complexes were run for 20 hours at 30 V on a 20 cm by 25 cm 1% agarose TAE gel (GIBCO-BRL) at room temperature The gels were visualized by staining in 1 μg/ml ethidium bromide and then cut down to 11 cm by 14 cm before they were soaked in 10X SSC and transferred to positively charged Tropilon membranes (Tropix) by Southern blotting method under non-denaturing conditions Blots were then UV cross-linked (Stratalmker)
Biotinylated cssDNA probes and probe target hybrids were detected using the Southern-Light System (Tropix) The nylon bound DNA blots were treated with avidin conjugated to alkaline phosphatase, followed by the chemiluminescent substrate, CDP-Star (Tropix), in conditions described by the manufacturer Blots were exposed to X-ray film (Kodak) for varying times (1 minute to 8 minutes) and developed Electroporation of probe target DNA hybrids into metabolically active E coli cells After purification of targeted complexes, 40 μl of electro-competent RecA(+) and/or RecA(-) E coli (Dower et al , 1988) was added to 30-200 ng of the targeted complexes in a chilled microfuge tube The RecA(+) cells were BB4 (Stratagene) and the RecA(-) cells were XL1-Blue (Stratagene) The mixture was incubated on ice for 1 minute This mixture was then transferred to a chilled 0 1 cm gap electroporation cuvette (Bio-Rad) and electroporated under the following conditions 1 3 V, 200 ohms, 25 μF on a Bio-Rad Gene Pulser The time constant ranged from 4 5 - 4 7 msec Immediately afterwards, 1 mL of SOC media (Sambrook, et al , 1989) was added and the mixture was transferred into a 10 mL culture tube After all the electroporation groups were finished, the tubes were shaken at 225 rpm at 37°C for 1 hour Appropriate amounts were plated onto LB agar plates which already contained 100 μg/ml ampicillin (Sigma), 20 μg/ml X-gal (GIBCO-BRL), and 48 μg/ml IPTG (GIBCO-BRL), and incubated at 37°C overnight
Screening for homologous DNA recombination in LacZ After overnight incubation (approximately 16 hrs ), colonies were counted to determine electroporation efficiency and scored for any blue colonies in plates Blue colonies were scored if they resembled blue colonies displayed by the control plasmid pBluescπpt II SK(-), which is able to undergo alpha-complementation and produce blue colonies Blue colonies were serially propagated on AIX plates at least twice to confirm recombinant stability as monitored by consistency of color When the colonial streaks displayed a homogeneous color, plasmids were isolated by a mini-prep and digested with EcoRI, Xhol, and Pvull to confirm homologous recombination of the plasmid at the DNA level EcoRI and Xhol sites are restored if homologous recombination has occurred Pvull restriction sites which flank the LacZ region contains the 59 base pair deletion, if recombination has occurred, this fragment will be significantly larger than fragments lacking the 59 base pairs after digestion with Pvull
RecA mediated cssDNA targeting to negatively supercoiled dsDNA substrates containing deletions Stable probe target hybrids formed in the RecA mediated targeting reaction between the biotinylated RecA coated cssDNA probes IP290 and the negatively supercoiled Form I dsDNA targets pRD 59, which contain a 59 base pair deletion respective to the cssDNA probe, were monitored by chemiluminescent detection of biotinylated hybrids (Figure 11) The presence of a sizable region of non-homologous nucleotide sequences (59 bp) in the cssDNA probe IP290 does not significantly affect the ability of the RecA coated cssDNA probe IP290 to form stable probe target hybrids with pRD 59 in comparison to completely homologous dsDNA pRD 0 (Figure 11 , lane 3 and 6) In each reaction, under these conditions, the presence of the RecA protein was absolutely required for hybrid detection (Figure 11 , lane 2 and 5) Probe target DNA hybrids formed when the RecA coated biotinylated cssDNA probe IP290 is hybridized to the completely homologous dsDNA target pRD 0 differ from probe target hybrids formed when the same cssDNA probe is hybridized to the dsDNA target pRD 59 containing a 59 base pair deletion with respect to IP290 While more than 90% of both the dsDNA targets exist as negatively supercoiled Form I DNA, when hybrids formed between pRD 0 and RecA coated cssDNA probe IP290 are deprotemized, the probe target hybrids migrate to a position that is similar to the migration of Form II, relaxed circular dsDNA, in 1 % agarose gel in 1X TAE buffer (Figure 11 , lane 3 and 6), and there was no evidence of probe target hybrids that co-migrate to Form I DNA on a 1% agarose gel (Figure 11 , lane 3) This probe target hybrid is referred to as a relaxed Form I* hybrid or a rl* hybrid because the hybrid has the same elelctrophoretic mobility as relaxed circular DNA In contrast, when the RecA coated cssDNA probe IP290 was hybridized to the dsDNA target pRD 59, which as a 59 bp deletion with respect to the probe, two different probe target hybrids were apparent One has an electrophoretic mobility comparable to that of Form I supercoiled dsDNA (Figure 11 , lane 6) while the other migrates to the same position as the rl* hybrid These two forms appear to be present in equal amounts as indicated by the signal from chemiluminescent DNA detection This probe target hybrid is referred to as a Form I* hybrid or I* hybrid, differentiating it from Form I DNA because it is targeted with RecA coated cssDNA probe In order to exclude the possibility that it is the structure of the dsDNA target that creates the formation of two major probe target hybrid products, the cssDNA probe DP231 was hybridized to pRD 59 The cssDNA probe DP231 is completely homologous to the mutant region of the LacZ gene in pRD 59 The only probe target hybrid detected has the electrophoretic mobility of Form II dsDNA, the rl* hybrid (Figure 11 , lane 8) In addition, when the cssDNA probe CP443, which is completely homologous to a region outside of the 59 base pair deletion, was hybridized to pRD 59, only the rl* hybrid product was detected (Figure 11 , lane 10) Thus, when the RecA coated cssDNA probes are targeted to homologous sequences, only the rl* hybrid is present, but when it is targeted to homologous sequences with relatively short heterologies, two forms of hybrids, rl* and I* hybrids are formed in apparently equivalent amounts
Recombinogenicity of probe target DNA hybrids To study the biological consequences of the probe target hybrid structures , we assayed for putative homologous recombination events in E coli by the electroporation assay (described in Figure 10)
Figure 12 shows the percentage of potential recombinant blue colonies formed when IP290 probe pRD 59 target hybrids were electroporated into RecA+ and RecA- cells Blue colonies only arose when deprotemized hybrids formed with pRD 59 and cssDNA probe IP290 are introduced into RecA+ E coli cells Control experiments performed with cssDNA probes homologous to the mutant LacZ region of pRD 59 (DP231) and homologous to a region outside of the LacZ gene (CP443) did not yield any blue colonies (Figure 12) In addition, when all of these hybrids were transformed into RecA(-) hosts, no blue colonies were produced from any type of hybrid, indicating the the recombinogenic effect is also dependent on endogenous RecA protein produced in the cell Thus only the cssDNA probe containing the 59 base pair correction produces recombinogenic clones in bacterial host cells that are RecA(+)
When potential homologous recombinant blue colonies were propagated by streaking out on AIX plates, only 50% of the colonies were blue When a blue colony from the first streak was propagated by recombinant streaking, the colonies remained stably blue over several generations If plasmid DNA was isolated from third generation propagations and then transformed into RecA(-) cells, this resulted in blue colonies which remained stably blue on continued propagation Of the potential recombinants that have been rigorously screened by restriction enzyme digestion, at least 67% of the plasmids recovered from blue colonies are true homologous recombinants This was deteπmined by the restoration of EcoRI and Xhol restriction sites, and a Pvull digest of the DNA shows a fragment that migrates at a higher molecular weight than fragments which are missing the 59 base pair region
This is consistent with the view that only one strand is exchanged in these hybrids to form heteroduplex targets and that upon replication one strand will produce a plasmid that contains the 59 base pair correction while the other does produces the mutant pRD59 plasmid
As outlined in Example 5, we show that the recombinogenicity with probe target hybrids of cssDNA probes and dsDNA targets containing deletions is associated with the re-anneahng of regions of cssDNA probe that can not hybridize to dsDNA targets, by creating internal homology clamps (Figure 13)
EXAMPLE 5 Enhanced homologous recombination with targets containing insertions and deletions containing internal homology clamps
An in vitro DNA hybridization reaction that allows the pairing of RecA-coated complementary smgle- stranded (ess) DNA probes to homologous regions in linear duplex target DNA has been used to study the effects of heterologies within the regions of homology between the probes and target DNA In cssDNA targeting reactions catalysed by RecA protein, cssDNA probes are kinetically trapped within the duplex DNA target at homologous sites and form a highly stable four-stranded DNA hybrid structure After removal of RecA protein, this homologous recombination reaction can be trapped at the DNA pairing step The effect of defined heterologous insertions or deletions in linear duplex targets on the pairing of RecA-coated cssDNA probes was determined for heterologies ranging from 4 to 967 bp We demonstrate that small deletions and insertions up to 10% of the total cssDNA probe lengths, ranging from 215 -1246 bp do not significantly affect DNA pairing Furthermore both insertions and deletions of the same size in the cssDNA probe have the same effect on DNA pairing Moreover, large deletions, up to 967 bp, can be tolerated in deprotemized hybrids form with a RecA- coated 1 2 kb cssDNA probe The stability of these hybrids with heterologous sequences within the homologous paired region is due to the re-anneahng of the cssDNA probes to each other within the DNA hybrid producing a novel four-stranded heteroduplex DNA intermediate that contains a novel internal base-paired homology clamp
Preparation of ds target substrates A series of plasmid DNA targets with defined deletions were constructed by linearization of the plasmid vector pBluescnpt IISK(-) (Stratagene) at a unique EcoRI restriction site in the polylinker region following digestion with mung bean exonuclease (Boehπnger- Mannheim), DNA ligation, and subsequent transformation into XL1-Blue E coli (Stratagene) by standard methods The resulting clones were sequenced using Sanger dideoxy sequencing methods (Sequenase Kit version 2, USB) to determine the extent of deletion A series of plasmids with deletions ranging from 4 to 967 bp were prepared and named for the extent of size of the deletion (see Figure 15) The size of the parent plasmid, pBluescnpt IISK(-), referred to as pRD 0 in this study, is 2960 bp Plasmid DNA was prepared by a modified alkaline lysis procedure with anion-exchange purification (Qiagen) The DNA was further purified by phenol-chloroform-isoamyl alcohol extraction (24 25 1) (SIGMA) and ethanol precipitation, and then resuspended in TE (10 mM Tns HCI, pH7 5, 1 mM EDTA) buffer These preparations contained greater than 90% Form I DNA Preparations of linearized Form III DNA were made by digestion of the plasmids at a unique Seal restriction site outside the polylinker, followed by phenol-chloroform-isoamyl alcohol extraction (SIGMA), chloroform extraction, ethanol precipitation, and resuspension in TE buffer
Preparation of cssDNA probes Biotin-labeled probes homologous to pRD 0 were synthesized by PCR with incorporation of bιotιn-14-dATP using previously described methods where the molar ratio of unlabelled dATP to biotm-labelled dATP was 3 1 (Griffin & Griffin, 1995) Primer pairs flanking the polylinker region of pRD 0 or analogous plasmids with a deletion were chosen to produce PCR fragments which span the deletion in the target plasmids In addition a control PCR fragment (CP443) primer pair flanking sequences outside the polylinker was selected for production of a probe homologous to all clones in the plasmid series The oligonucleotide products were purified by membrane ultrafiltration using Microcon 100 filters (Amicon)
Targeting of cssDNA probes to dsDNA targets in solution cssDNA targeting was performed essentially as described in Sena & Zarhng (1993), with the exception that cssDNA probes were synthesized and labeled by PCR in the presence of bιotιn-14-dATP (GIBCO/BRL), as indicated above In each reaction 70 ng of biotm-labelled RecA-coated cssDNA probe was reacted with 1 μg of Sca1- digested target DNA, resulting in cssDNA probe target ratios of 1 1 (for 215 bp cssDNA probes) to 1 5 (for 1246 bp cssDNA probes) The products of the targeting reactions were deprotemized by treatment with SDS (1 2% final concentration) or phenol chloroform isoamyl alcohol (24 25 1) and chloroform extraction and then separated by electrophoresis on 1% agarose gels in TAE buffer The gels were run at 2V/cm at room temperature in the absence of ethidium bromide for 20 hours After electrophoresis, gels were stained in 1 μg/ml ethidium bromide for 15 mm The DNA was transferred under non-denaturing conditions (10X SSC) onto nylon membranes (Tropix) and cross-linked using a Stratahnker (Stratagene) on the auto-crosshnk setting The extents of biotinylated cssDNAprobe target hybrid formation was measured by quantitatmg the amount of biot -labeled probe DNA that co-migrates with dsDNA target DNA following electrophoretic separation of these biotinylated probe target hybrid products from free unhybridized probe DNA The amount of biotinylated probe DNA in probe target complexes was visualized with a chemiluminescent substrate conjugated to streptavidin (CDP-STAR) (Tropix) after exposure to XAR-5 film (Kodak) The levels of exposure were analyzed by densitometry and quantitated using the software package, NIH Image
In each case the relative level of hybrid formation with heterologous targets was expressed as a percentage of the level of hybrid formation of standardized reactions with a completely homologous probe and target These values were normalized to the level of hybrid formation that occured with control probe CP443 which hybridizes to all of the plasmid targets in a region away from the heterology The data generally represent averages of at least three separate measurements from three independent targeting reactions
Nomenclature and Assay for RecA-mediated pairing of cssDNA probes to dsDNA targets To investigate the effects of heterologous insertions and deletions on homologous pairing of cssDNA probes to double-stranded linear plasmid DNA, we employed a modification of an in vitro DNA targeting assay described in Sena and Zarlmg (1993) The target DNAs used in this study are a series of plasmid DNA constructs that contain defined deletions at the unique EcoRI site in pRD 0
(pbluescπptllSK(+), Stratagene Figure 14A) Plasmid targets (pRD 4 - pRD 967) are named for the size of deletion in bp at the EcoRI site CssDNA probes were made and labelled with bιotιn-14-dATP by PCR using primers which symetπcally flank the deleted region of plasmids in the pRD series CssDNA probes made from pRD 0 that were targeted to plasmids containing deletions are called insertion probes and named for the length of the probe in bp For example, IP290 is a 290 bp cssDNA probe that contains an insertion with respect to a target containing a deletion, but is completely homologous to pRD 0 A cssDNA probe made from pRD 59 and targeted to pRD 0 is called DP231 , since it contains a deletion with respect to pRD 0, but is completely homologous to pRD 59 After the hybridization of RecA-coated cssDNA probes with dsDNA targets, the reactions products were separated by agarose gel electrophoresis The extent of formation of stable deprotemized cssDNA probe target hybrid was measured by the quantitation of the amount of biotinylated cssDNA probes that co-migrated with the dsDNA targets In each case the level of probe target formation between a totally homologous probe and target was normalized to 100% Previous studies have shown that efficient cssDNA targeting is completely dependent on RecA protein, the nucleotide co- factor, specific to homologous DNA targets and that formation of deprotemized stable probe target hybrids also requires both cssDNA strands (Sena and Zarlmg, 1993, Revet et al, 1993) Furthermore we targeted Seal-digested pRD 0 with two synthetic RecA-coated 121-mer cssDNA oligonucleotides homologous to the region sy metrically spanning the EcoRI site in pRD 0 and demonstrated that both cssDNA strands are required for stable hybrid formation with linearized pRD 0 targets (data not shown)
Stable cssDNA probe target hybrids are formed in linear dsDNA targets with deletions at internal sites To determine if a target DNA deletion affects the reaction kinetics of RecA-mediated cssDNA pairing to linear DNA targets, we measured the relative amount of deprotemized cssDNA probe target hybrid formation over time in reactions using cssDNA probe IP290 with either a completely homologous linear target, pRD 0 or a target carrying a 59 bp deletion, pRD 59 Probe IP290 symetπcally spans the 59 bp deletion in pRD 59 Figure 15B shows that in steady state hybrid reactions, the maximum level of stable hybrid formation when RecA-coated IP290 is targeted to pRD 59 is 62% of the steady state level obtained with the fully homologous target pRD 0 Furthermore steady state levels of hybrid formation occurs within 45 minutes with fully homologous pRD 0 targets, but requires 2 hours for pRD 59 targets Thus, in all subsequent experiments RecA-coated probes were hybridized for 2 hours at 37°C with the linear target DNAs
The effect of duplex DNA target deletions on the formation of deprotemized cssDNA probe target hybrids was determined by hybridizing RecA coated cssDNA probes which span the deleted regions in pRD 4 - pRD 298 on DNA targets linearized by Seal (Figure 15A) The relative amount of biotinylated probe target hybrids formed with each of these targets was compared with the amount of cssDNA probe target hybrids formed with pRD 0 These values were normalized to the level of hybrid formation obtained with the control probe, CP443, which is homologous to a region away from the deleted regions or pRD 0 and thus, is completely homologous to all target DNA substrates used in this study
Our initial studies tested the effect of small target deletions on targeting efficiency using either cssDNA probes IP527 or IP407 (Figure 15B and 15C) Because the 5'- and 3'-termιnι of both of these cssDNA probes are approximately symmetric with respect to the 4 to 59 bp deletions, the differences in the efficiency of hybrid formation are not due to the effects of the position of the deletion with respect to the probe in relation to probe ends As expected, in experiments using either the IP527 or IP407 we observed a decrease in the level of hybrid formation with an increase deletion size These data also show that relatively small deletions (< 25 bp) in the target do not dramatically affect the overall targeting efficiency of cssDNA probes to linear targets and that the deletions have relatively the same effect on the hybridization on either IP527 and IP407 However when the size of the deletion is increased to 59 bp (11% of the length of IP527), the relative targeting efficiency of probes IP527 and IP407 drops to 61% and 33%, respectively Furthermore the amount of the difference between the targeting efficiency mediated by these probes continues to increase linearly as the size of the deletion increases (Figure 15D) This indicates that when the size of the deletion is >10% of the length of the probe the efficiency of RecA-mediated DNA targeting is governed by the amount of homology between the cssDNA probe and target, while deletions <10% of the length of the probe are well tolerated for any length of cssDNA probe Similar effects are observed with smaller cssDNA probes IP452, IP290 (data not shown) and IP215 (Figure 16)
Heterologous insertions and deletions are similarly tolerated in the hybridization of cssDNA probes to linear dsDNA targets Other studies by Bianchi and Radding (Cell 35 511-520 (1983)) in which RecA- coated circular ssDNA was hybridized to linear duplex targets demonstrated that heterologous inserts in the ssDNA were tolerated somewhat better than when the insert was in the dsDNA, presumably because the inserts in ssDNA could be folded out of the way In contrast, Morel et al (J Biol Chem 269 19830 (1994)) used somewhat similar substrates and demonstrated that RecA-mediated strand exchange could bypass heterologies with equal efficiency whether the insert was in the ssDNA or dsDNA Since the formation of stable cssDNA probe target hybrids with internal sequences in linear dsDNA requires two cssDNA probe strands, we compared the effects of insertions in the cssDNA probe with having the same sized insertion in the dsDNA to determine how these internal heterologies maybe accommodated within a four strand containing double-D-loop DNA structure
In these studies we compared the effects of 4 to 59 bp insertions in either the dsDNA target or cssDNA probe (deletion in target) using cssDNA probes ranging in size from 156 bp to 215 bp We used this smaller cssDNA probe to maximize the effects of the insertion or deletion of these sizes We prepared cssDNA probe IP215 from pRD 0 using PCR and targeted pRD 0, pRD 4, pRD 25, and pRD 59 to measure the effects of insertions in cssDNA probes (target DNA deletion) Then using the same PCR primer set, we prepared cssDNA probes from templates pRD 0, pRD 4, pRD 25, and pRD 59 and then targeted pRD 0 to measure the effects of deletions in cssDNA (target DNA insertion) Figure 16 shows that both deletions and insertions of the same size have exactly the same effect on RecA-mediated cssDNA targeting and are equally tolerated and stable Large deletions in linear DNA are tolerated in cssDNA probe target hybrids with linear dsDNA To further define the extents of heterology that can be tolerated during cssDNA hybridization, we studied the effect of very large deletions, up to 448-967 bp on the targeting efficiency using a 1246 bp cssDNA probe (IP1246) (Figure 17A) With target deletions in the range of 500 bp (approx 50% of the cssDNA probe length) there is only a slight reduction in the targeting efficiency achieved with this probe (80%), surprisingly the IP1246 can hybridize target DNA molecules bearing deletions up to 967 bp at a detectable efficiency (27%) When IP1246 is targeted to pRD 967, there are a total of 279 bp of homology between the cssDNA probe and target, with 147 bp 5' to the 967 bp insert and 132 bp 3' to the insert (Figure 17B) In order to account for such a high level of targeting efficiency with such a large deletion, we predict that the 967 bp insert in the two in the cssDNA probe strands, which are homologous to each other, may interact with each other to stabilize this hybrid
Furthermore when using a large cssDNA probes of 1246 bp we can observe a visible shift the migration of the cssDNA probe target hybrid in comparison to the linear dsDNA target The positions of the migration of the of the 3 0 kb Seal -digested ds DNA marker are shown in Figure 17A Note the cssDNA probe:target hybrids formed with IP1248 migrate slower than each of the Seal -digested targets, but that cssDNA probe target hybrids formed with CP443, a smaller probe migrate closer the positions of the formlll markers The presence of this labelled slower-migrating species provides further evidence for the existence of the multi-stranded DNA hybrids
EcoRI Restriction endonucleases cut duplex DNA in either homologous or heterologous cssDNA probe target hybrids To further characterize cssDNA probe target hybrids formed with heterologous DNA targets, circular plasmids pRD 0 and pRD 59 were hybridized with biotm-labelled probe IP290 and then deprotemized and digested with EcoRI While plasmid pRD 0 contains a unique EcoRI site in the region of homology between IP290 and pRD 0, the EcoRI site is deleted in pRD 59 (Figure 14A) Digestion of cssDNA probe target hybrids with EcoRI indicates the restoration of Watson-Crick pairing to form a fully duplex EcoRI recognition site Figure 18 shows both the ethidium bromide stained gel of the hybrid product of the targeting reaction (Figure 18A and 18B) and the corresponding autoradiograph that shows the electrophoretic migration of the biotm-labelled probes (Figure 18C and 18D) These data show that when RecA-coated IP290 is hybridized to the fully homologous pRD 0 plasmid all of the probe-target hybrids migrate to the position of fully relaxed DNA (Figure 18 A and C, Lane 1) Furthermore, upon digestion with EcoRI cssDNA probe target hybrids can be completely cut, as shown in Figure 18 A and C, Lane 2 When similar reactions are performed with uncut pRD 59 targets, we found that not all of the probe target hybrids are relaxed as with pRD 0 targets, as judged by the appearance of two bands corresponding to a pRD59 I* hybrid, where the hybrids co-migrate with Form I supercoiled DNA and a pRD59 rl* hybrid that migrates with relaxed targets (Figure 18B and D, Lane 3) When these hybrids are digested with EcoRI we find that the pRD59 rl* hybrid is more susceptible to EcoRI cleavage than the pRD59 rl* hybrid (Figure 18B and D, Lane 4) This shows that there is a restoration of the EcoRI site in relaxed targets, but not in the non-relaxed I* hybrid Since pRD59 targets do not contain an EcoRI site, cleavage by EcoRI can only be explained by re-anneahng of cssDNA probe IP290 within the IP290 probe target pRD59 hybrid
To further characterize the structural differences between pRD59 rl* hybrids and pRD59 I* hybrids, cssDNA probe target hybrids were formed between IP290 and pRD59, deprotemized and thermally melted for 5 mms at 37°C, 45°C, 55°C, and 65°C, respectively Figure 19 shows that pRD59 rl* hybrids are more thermostable than pRD59 I* hybrids For both types of hybrids probe target hybrids are completely dissociated after heating to 95°C (data not shown) Taken together these data support the structures of our models for hybrids (Figure 13)
EXAMPLE 6 Homologous recombination targeting in fertilized mouse zygotes
Ornithme transcarbamylase (OTC) is a mitochondnal matrix enzyme that catalyzes the synthesis of citrulline from ornithme and carbamylphosphate in the second step of the mammalian urea cycle OTC deficiency in humans is the most common and severe defect of the urea cycle disorders OTC is an X-lmked gene that is primarily expressed in the liver and to a lesser extent in the small intestine Affected males develop hyperammonemia, acidosis, orotic aciduπa, coma and death occurs in up to 75% of affected males, regardless of intervention Two allehc mutations at the OTC locus are known in mice spf and spf-ash, (sparse fur-abnormal skin and hair) In addition to hyperammonemia and orotic aciduπa, spf-ash mice can be readily identified by the abnormal skin and hair phenotype The spf-ash mutation is a single-base substitution at the end of exon 4 that results in alternative mtron- exon splicing to produce an aberrant non-functional elongated pre-mRNA Because of the clinical importance of OTC defects in humans, there is an intensive effort to develop in vivo methods to correct the enzymatic defect in the spf-ash mouse model
We used the muπne spf-ash model of OTC deficiency to test the ability of RecA-coated complementary single-stranded DNA (ess) OTC probes to target and correct a single-base substitution mutation in fertilized mouse zygotes A 230 bp RecA-coated cssDNA probe amplified from the normal mouse OTC gene was microinjected into embryos derived from matins of B6C3H homozygous spf-ash female with normal B6D2F1 J males After re-implantation of 75 embryos that were microinjected with RecA-coated cssDNA into CD1 foster mothers, 25 developmentally normal pups (17 female and 8 male) were born Sequence analysis of the genomic DNA isolated from tails of the male pups show that 3 out of 8 males were mosaic for a homologous recombination event at the spf-ash site in exon4 of the mouse OTC gene Subsequent breeding of the three founder males with normal females resulted in normal female F, progeny, thus demonstrataing germhne transmission of the homologous recombinant allele as well as phenotypic correction in F, animals These homologoous recombinant changes were stable in F2 and subsequent generations These studies illustrate cssDNA mediated high frequency homologous recombination in fertilized mouse zygotes to create subtle genetic modifications at a desired target site in the chromosome
Preparation of RecA-coated probe A 230 bp fragment from the normal mouse OTC gene was amplified by PCR with primers M9 and M8 from pTAOTC (Figure 20) The PCR fragment was purified on Mιcrocon-100 columns (Amicon) and then extensively dialyzed The M9-M8 amphcon was denatured by heating the fragments to 98°C and then coated with RecA protein (Boehπnger- Mannheim) at a ratio 3 nucleotides/ protein monomer The final concentration of RecA-coated DNA in coating buffer (5 mM TπsOAc, pH 7 5, 0 5 mM DTT, 10 mM MgOAc, 1 22 mM ATP(S, 5 5 μM RecA) was 5 ng/ μL RecA-coated filaments were made on the day of microinjection and then stored on ice until use
Transgenic Mice Five superovulated B6C3H (spf-ash/spf-ash) 5-7 week old females (Jackson Labs) were mated with five B6D2F1 males (Jackson Labs) Approximately 80-100 embryos were isolated from oviducts as described in Hogan et al (1988) The female pronucleus of fertilized embryos was microinjected with 1-2 pi of RecA-coated M9-M8 cssDNA probe (5 ng/μL) Approximately 75 embryos survived the microinjection procedure and were then re-implanted into a total of three CD1 pseudopregnant foster mothers (Charles River) Pseudopregnant females were produced by mating foster mothers with vasectomized CD1 males (Charles River)
DNA Analysis Tail biopsies were taken from all founder mice after weaning at three weeks of age Genomic DNA was isolated from tail biopsies using standard procedures To obtain the sequence of the DNA at the OTC locus, genomic DNA was amplified with PCR using primers M10-M11 or M54- M11 that flank the cssDNA probe sequence to generate a 250 bp or 314 bp amphcon (Figure 20) PCR fragments were sequenced manually using the Cyclist Exo Kit (Stratagene), automatically on an Applied Biosystems Model 373A sequencer, or by a MALDI-TOF mass spectrometry system (GeneTrace Systems, Menlo Park, CA)
Fertilized zygotes microinjected with RecA-coated DNA are viable Plasmid pTAOTCI carries a 250 bp segment of exon4 and surrounding mtron sequences from the normal mouse OTC gene A 230 bp cssDNA probe OTC1 was prepared by PCR amplification of pTAOTCI with primers M9 and M8 cssDNA probe OTC1 was denatured and coated with RecA protein as described herein Homozygous spf-ash/spf-ash female and hemizygous (spf-ash/y) males can be phenotypically identified by the appearance of sparse fur and wrinkled skin early in development A cross between homozygous spf-ash/spf-ash B6C3H females and normal B6D2F1 males yields heterozygous phenotypically normal females and hemizygous males with sparse fur and wrinkled skin The RecA- coated cssDNA OTC probe was microinjected into embryos made from the cross of B6C3H homozygous female spf-ash and normal males The female pronucleus of approximately 80-90 fertilized zygotes was microinjected with 2 pi of a 5ng/μL solution of RecA-coated cssDNA probe OTC1 Of these, 75 embryos survived the microinjection procedure To demonstrate that embryos that have been microinjected with RecA-coated cssDNA are viable, the embryos were re-implanted into three pseudopregnant CD1 foster mothers From this, 25 developmentally normal pups (17 female and 8 male) were produced All of the female mice were phenotypically normal The eight male mice (mouse # 7, 14,16,17,22,23,24, and 25) were all affected with sparse-fur and wrinkled skin to various degrees
RecA-coated cssDNA probe OTC1 recombines with the homologous chromosomal copy of the OTC gene in fertilized mouse zygotes To determine the genotypes of the 25 founder mice produced from microinjected embryos, genomic DNA was isolated from tail biopsies Genomic DNA was amplified with either the primer set M10-M11 or M54-M11 to produce either a 250 bp or 314 bp amphcon By using these primer sets that flank the OTC1 probe, the DNA amphcon represents DNA from the endogenous OTC gene PCR fragments from all of the eight mice and several female mice were sequenced to determine the base sequence at the spf-ash locus to determine if a normal allele (G) or a mutant allele (A) was present in the genomic DNA Figure 21 shows sequencing gels of representative reactions The panel on the left side shows the sequence of the homozygous spf-ash females that donated the eggs to produce the fertilized zygotes where only the mutant base A is present at the spf-ash locus, as expected The sequence of female mouse #8 that should be heterozygous shows the presence of equal amounts of the bases G and A as expected Male mice 7, 14 (shown), 23, 24, and 25 all showed only the mutant base A at the spf-ash locus, however male mice 16, 17, and 22 (shown) displayed both G (normal) and A (mutant) at the spf-ash locus
To eliminate the possibility of PCR artifacts during PCR cycle sequencing the base compositions of the samples was independently confirmed by mass spectrometry sequencing (GeneTrace, Menlo Park) The relative (%) amounts of the A G base composition at the spf-ash locus was also quantified and determined to be 70% 30% for samples from mouse #16 and #17 and 10% 90% for mouse #22 Since OTC is an X-hnked gene the presence of mixed bases in male mice is likely the result of the mosaic animals produced of a mixture of mutant and gene corrected embryonic cells Germhne transmission of the gene corrected OTC allele To determine if the gene corrected allele in the mosaic male founder mice (#'s 16, 17, and 22) could be passed through to the germhne, these mice and a control hemizygous mutant male (#7) were bred with normal B6D2F1 females In this cross, if the male donates a mutant spf-ash X chromosome, then the resulting female progeny will be heterozygous spf-ash mutants However if the male donates a normal (gene corrected) X chromosome the female progeny will be homozygous normal In both cases the resulting F1 females will be phenotypically normal The results of these crosses are summarized in Figure 22 In the control cross of hemizygous mutant male #7 with B6D2F1 females, all 14 female progeny were heterozygous, as expected In test crosses of mosaic male mouse #17 and #22 with normal females, all resulting female progeny (5 and 9, respectively) were heterozygous However in the cross with mosaic male mouse #16, one out nine total female progeny was a homozygous normal female (mouse # 213), as determined mass spectrometry DNA sequencing (GeneTrace, Menlo Park), demonstrating the gene corrected allele in founder mouse #16 was tranmitted through the germhne
To further verify that the F1 mouse #213 was, in fact, a germhne-transmitted gene corrected homozygous normal female, this mouse and a control heterozygous spf-ash/+ mouse were bred with normal males In the control cross B with the heterozygous female, 50% of the resulting male F2 progeny should be mutant spf-ash/y hemizygotes that can be easily determined by the visualization of the sparse-fur and wrinkled skin phenotype Of the 38 progeny produced in this control cross B, 14 were male, and of these, 8 were phenotypically normal and 6 were mutant, as determined by the presence of wrinkled skin and abnormal fur In the test cross with F1 mouse #213, of the 35 progeny produced in this cross, all eleven of the male progeny were phenotypically normal, clearly showing the genotypmg of F1 mouse #213 as a germhne transmitted gene corrected homozygous normal female
As another independent test to determine if the normal gene corrected allele in mouse #16 could be transmitted through the germhne, mouse #16 was mated with homozygous (spf-ash/spf-ash) mutant females In this cross if mouse #16 does not transmit a normal allele, the resultant progeny will either be hemizygous (spf-ash/Y) mutant males or homozygous (spf-ash/spf-ash) mutant females, both of which are phenotypically mutant However if the mouse allele is transmitted through the germhne, heterozygous (spf-ash/+) females that are phenotypically normal will be produced When mouse #16 was bred with homozygous (spf-ash/spf-ash) mutant females, two litters were produced that consisted of a total of 5 hemizygous (spf-ash/Y) mutant males, 7 homozygous (spf-ash/spf-ash) mutant females, and 1 phenotypically normal female (mouse #1014) Pictures of representative mice from these crosses are shown in Figure 23 The production of the phenotypically normal female mouse provides direct genetic evidence that mouse #16 contains a normal gene corrected OTC allele that is germhne transmissable Although the present invention has been described in some detail by way of illustration for purposes of clarity of understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the claims

Claims

CLAIMS We claim
1 A non-human mammal comprising a modified endogenous gene, wherein said endogenous gene is selected from the group consisting of a gene or sequence encoding an ion-channel, a G- protein coupled receptor (GPCR), an immunoglobulm, a growth factor, an enzyme, or a milk protein
2 A mammal according to claim 1 wherein said mammal is a farm animal
3 A mammal according to claim 2 wherein said farm animal is selected from the group consisting of cattle, sheep, pigs, horses and goats
4 A mammal according to claim 1 wherein said mammal is selected from the group consisting of mice, rats, rabbits , guinea pigs, hamsters and gerbils
5 A mammal according to claim 1 wherein said milk protein gene is a lactoglobulm gene
6 A mammal according to claim 5 wherein said lactoglobulm gene is the "-lactoglobulm gene or the $-lactoglobuhn gene
7 A mammal according to claim 6 wherein said modified "-lactoglobulm gene or $-lactoglobuhn gene does not encode any phenylalanme residues
8 A mammal according to claim 1 wherein said endogenous gene is disrupted by deletion of at least one nucleotide
9 A mammal according to claim 1 wherein said endogenous gene is disrupted by an insertion sequence
10 A mammal according to claim 9 wherein said insertion sequence is a pojyhnker sequence
11 A mammal according to claim 9 wherein said insertion sequence is a reporter gene
12 A mammal according to claim 11 wherein said reporter gene is selected from the group consisting of a luciferase gene, a β-galactosidase gene and green fluorescent protein (GFP), blue fluorescent protein (BFP), red fluorescent protein (RFP) and yellow fluorescent protein (YFP)
13 A mammal according to claim 9 wherein said insertion sequence is selected from the group consisting of a gene encoding human lysozyme, human growth hormone, human serum albumin, human globin, a human immunoglobulm, and a human enzyme
14 A mammal according to claim 12 wherein said human enzyme is α-1 antitrypsin
15 A mammal according to claim 12 wherein said human enzyme is anti-thrombin III
16 A mammal according to claim 12 wherein said human enzyme gene does not encode any phenylalanme residues
17 A mammal according to claim 9 wherein said insertion sequence is selected from the group consisting of a human gene under control of its endogenous promoter, a modified endogenous regulatory element for an endogenous gene, a transcriptional regulation cassette and a dimeπzing sequence
18 A mammal according to claim 17 wherein said endogenous regulatory element is disrupted by deletion of at least one nucleotide
19 A mammal according to claim 17 wherein said regulatory element is disrupted by an insertion sequence
20 A mammal according to claim 1 wherein said enzyme is a sugar transferase enzyme
21 A mammal according to claim 20 wherein said sugar transferase enzyme is "-galactosyl transferase
22 A mammal according to claim 21 wherein said "-galactosyl transferase gene is disrupted by deletion of at least one nucleotide
23 A mammal according to claim 21 wherein said "-galactosyl transferase gene is disrupted by an insertion sequence 24 A mammal according to claim 23 wherein said insertion sequence is a hormone receptor gene
25 A mammal according to claim 23 wherein said insertion sequence is a viral receptor gene
26 A mammal according to claim 23 wherein said insertion sequence is a G-protem coupled receptor gene
27 A pnmate comprising a modified endogenous gene
28 A pnmate according to claim 27 wherein said endogenous gene is disrupted by deletion of at least one nucleotide
29 A pnmate according to claim 27 wherein said endogenous gene is disrupted by an insertion sequence
30 A pnmate according to claim 29 wherein said insertion sequence is a human therapeutic gene
31 A pnmate according to claim 29 wherein said insertion sequence is a human antibody gene
PCT/US1999/010731 1998-05-15 1999-05-14 Transgenic animals produced by homologous sequence targeting WO1999060108A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002341350A CA2341350A1 (en) 1998-05-15 1999-05-14 Transgenic animals produced by homologous sequence targeting
AU40797/99A AU4079799A (en) 1998-05-15 1999-05-14 Transgenic animals produced by homologous sequence targeting
NZ508665A NZ508665A (en) 1998-05-15 1999-05-14 Method of producing transgenic animals by homologus sequence targeting
EP99924251A EP1078048A2 (en) 1998-05-15 1999-05-14 Transgenic animals produced by homologous sequence targeting

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7987798A 1998-05-15 1998-05-15
US09/079,877 1998-05-15

Publications (2)

Publication Number Publication Date
WO1999060108A2 true WO1999060108A2 (en) 1999-11-25
WO1999060108A3 WO1999060108A3 (en) 2000-02-03

Family

ID=22153380

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/010731 WO1999060108A2 (en) 1998-05-15 1999-05-14 Transgenic animals produced by homologous sequence targeting

Country Status (5)

Country Link
EP (1) EP1078048A2 (en)
AU (1) AU4079799A (en)
CA (1) CA2341350A1 (en)
NZ (2) NZ508665A (en)
WO (1) WO1999060108A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001050847A2 (en) * 1999-12-23 2001-07-19 Pangene Corporation Reca / rad51 recombinase-mediated production of recombinant organisms
WO2003022220A2 (en) * 2001-09-07 2003-03-20 The Regents Of The University Of California Method of generating a transgenic livestock animal
WO2002054862A3 (en) * 2000-12-13 2003-04-10 Deltagen, Inc. TRANSGENIC MICE CONTAINING Kv8.1 POTASSIUM CHANNEL ALPHA SUBUNIT GENE DISRUPTIONS
WO2008106984A2 (en) * 2007-03-07 2008-09-12 Aarhus Universitet Pig model for breast cancer, mitochondria related protein folding disorders and/or epidermolysis bullosa simplex
US7807863B2 (en) 2002-11-08 2010-10-05 Kyowa Hakko Kirin Co., Ltd. Transgenic bovine having reduced prion protein activity and uses thereof
US7928285B2 (en) 2004-04-22 2011-04-19 Kyowa Hakko Kirin Co., Ltd. Method of producing xenogenous antibodies using a bovine
WO2022232545A1 (en) * 2021-04-30 2022-11-03 Logicbio Therapeutics, Inc. Viral vector compositions and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004669A1 (en) * 1992-08-21 1994-03-03 Medical Research Council Mice having cftr defect as model for cystic fibrosis

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004669A1 (en) * 1992-08-21 1994-03-03 Medical Research Council Mice having cftr defect as model for cystic fibrosis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CLARKE, L.L. ET AL.: "Defective epithelial chloride transport in a gene-targeted mouse model of cystic fibrosis" SCIENCE., vol. 257, no. 5073, 21 August 1992 (1992-08-21), pages 1125-1128, XP002113624 AAAS. LANCASTER, PA., US *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001050847A2 (en) * 1999-12-23 2001-07-19 Pangene Corporation Reca / rad51 recombinase-mediated production of recombinant organisms
WO2001050847A3 (en) * 1999-12-23 2002-07-11 Pangene Corporation Reca / rad51 recombinase-mediated production of recombinant organisms
WO2002054862A3 (en) * 2000-12-13 2003-04-10 Deltagen, Inc. TRANSGENIC MICE CONTAINING Kv8.1 POTASSIUM CHANNEL ALPHA SUBUNIT GENE DISRUPTIONS
WO2003022220A2 (en) * 2001-09-07 2003-03-20 The Regents Of The University Of California Method of generating a transgenic livestock animal
WO2003022220A3 (en) * 2001-09-07 2003-07-03 Univ California Method of generating a transgenic livestock animal
US7199281B2 (en) 2001-09-07 2007-04-03 The Regents Of The University Of California Method of generating a transgenic livestock animal
US7807863B2 (en) 2002-11-08 2010-10-05 Kyowa Hakko Kirin Co., Ltd. Transgenic bovine having reduced prion protein activity and uses thereof
US7928285B2 (en) 2004-04-22 2011-04-19 Kyowa Hakko Kirin Co., Ltd. Method of producing xenogenous antibodies using a bovine
WO2008106984A2 (en) * 2007-03-07 2008-09-12 Aarhus Universitet Pig model for breast cancer, mitochondria related protein folding disorders and/or epidermolysis bullosa simplex
WO2008106984A3 (en) * 2007-03-07 2008-12-31 Univ Aarhus Pig model for breast cancer, mitochondria related protein folding disorders and/or epidermolysis bullosa simplex
WO2022232545A1 (en) * 2021-04-30 2022-11-03 Logicbio Therapeutics, Inc. Viral vector compositions and methods of use thereof

Also Published As

Publication number Publication date
NZ508665A (en) 2003-09-26
NZ527435A (en) 2005-01-28
CA2341350A1 (en) 1999-11-25
AU4079799A (en) 1999-12-06
EP1078048A2 (en) 2001-02-28
WO1999060108A3 (en) 2000-02-03

Similar Documents

Publication Publication Date Title
US5948653A (en) Sequence alterations using homologous recombination
WO1998042727A9 (en) Sequence alterations using homologous recombination
US6255113B1 (en) Homologous sequence targeting in eukaryotic cells
US5763240A (en) In vivo homologous sequence targeting in eukaryotic cells
US20020061530A1 (en) Enhanced targeting of DNA sequences by recombinase protein and single-stranded homologous DNA probes using DNA analog activation
AU762766B2 (en) The use of consensus sequences for targeted homologous gene isolation and recombination in gene families
US20020108136A1 (en) Transgenic animals produced by homologous sequence targeting
WO2000009755A2 (en) Domain specific gene evolution
WO1999060108A2 (en) Transgenic animals produced by homologous sequence targeting
US20050214944A1 (en) In vivo homologous sequence targeting in cells
US20020090361A1 (en) In vivo homologous sequence targeting in cells
US20040019916A1 (en) In vivo homologous sequence targeting in eukaryotic cells
EP1244806A2 (en) Reca / rad51 recombinase-mediated production of recombinant organisms
AU2003223612B2 (en) Method to enhance homologous recombination
US20040023213A1 (en) Domain specific gene evolution

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
NENP Non-entry into the national phase in:

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 40797/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 508665

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1999924251

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999924251

Country of ref document: EP

ENP Entry into the national phase in:

Ref country code: CA

Ref document number: 2341350

Kind code of ref document: A

Format of ref document f/p: F

Ref document number: 2341350

Country of ref document: CA

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1999924251

Country of ref document: EP