CA2918540A1 - Methods of altering target dna in cells, and corresponding cells - Google Patents

Methods of altering target dna in cells, and corresponding cells Download PDF

Info

Publication number
CA2918540A1
CA2918540A1 CA2918540A CA2918540A CA2918540A1 CA 2918540 A1 CA2918540 A1 CA 2918540A1 CA 2918540 A CA2918540 A CA 2918540A CA 2918540 A CA2918540 A CA 2918540A CA 2918540 A1 CA2918540 A1 CA 2918540A1
Authority
CA
Canada
Prior art keywords
cell
talen
longer
dna
repeat sequences
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CA2918540A
Other languages
French (fr)
Inventor
George M. Church
Luhan Yang
Marc Guell CARGOL
Joyce Lichi YANG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harvard College
Original Assignee
Harvard College
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 Harvard College filed Critical Harvard College
Publication of CA2918540A1 publication Critical patent/CA2918540A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal 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
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/90Vectors containing a transposable element
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • C12N2830/003Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor tet inducible

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Transplantation (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Saccharide Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Methods of genome engineering in cells using a TALEN lacking repeat sequences or Cas9 is provided.

Description

GENOME ENGINEERING
RELATED APPLICATION DATA
This application claims priority to U.S. Provisional Patent Application No.
61/858,866 filed on July 26, 2013 and is hereby incorporated herein by reference in its entirety for all purposes.
STATEMENT OF GOVERNMENT INTERESTS
This invention was made with government support under P50 HG003170 from the National Human Genome Research Center for Excellence in Genomics Science. The government has certain rights in the invention.
BACKGROUND
Genome editing via sequence-specific nucleases is known. See references 1, 2, and 3 hereby incorporated by reference in their entireties. A nuclease-mediated double-stranded DNA
(dsDNA) break in the genome can be repaired by two main mechanisms: Non-Homologous End Joining (NHEJ), which frequently results in the introduction of non-specific insertions and deletions (indels), or homology directed repair (HDR), which incorporates a homologous strand as a repair template. See reference 4 hereby incorporated by reference in its entirety. When a sequence-specific nuclease is delivered along with a homologous donor DNA
construct containing the desired mutations, gene targeting efficiencies are increased by 1000-fold compared to just the donor construct alone. See reference 5 hereby incorporated by reference in its entirety. Use of single stranded oligodeoxyribonucleotides ("ssODNs") as DNA donors has been reported. See references 21 and 22 hereby incorporated by reference in their entireties.
Despite large advances in gene editing tools, many challenges and questions remain regarding the use of custom-engineered nucleases in human induced pluripotent stem cell ("hiPSC") engineering. First, despite their design simplicity, Transcription Activator-Like Effectors Nucleases (TALENs) target particular DNA sequences with tandem copies of Repeat Variable Diresidue (RVD) domains. See reference 6 hereby incorporated by reference in its entirtety. While the modular nature of RVDs simplifies TALEN design, their repetitive sequences complicate methods for synthesizing their DNA constructs (see references 2, 9, and 15-19 hereby incorporated by reference in their entireties) and also impair their use with lentiviral gene delivery vehicles. See reference 13 hereby incorporated by reference in its entirety.

In current practice, NHEJ and HDR are frequently evaluated using separate assays.
Mismatch-sensitive endonuclease assays (see reference 14 hereby incorporated by reference in its entirety) are often used for assessing NHEJ, but the quantitative accuracy of this method is variable and the sensitivity is limited to NHEJ frequencies greater than-3%. See reference 15 hereby incorporated by reference in its entirety. HDR is frequently assessed by cloning and sequencing, a completely different and often cumbersome procedure. Sensitivity is still an issue because, while high editing frequencies on the order of 50% are frequently reported for some cell types, such as U205 and K562 (see references 12 and 14 hereby incorporated by reference in their entireties), frequencies are generally lower in hiPSCs. See reference 10 hereby incorporated by reference in its entirety. Recently, high editing frequencies have been reported in hiPSC
and hESC using TALENs (see reference 9 hereby incorporated by reference in its entirety), and even higher frequencies with the CRISPR Cas9-gRNA system (see references 16-19 hereby incorporated by reference in their entireties. However, editing rates at different sites appear to vary widely (see reference 17 hereby incorporated by reference in its entirety), and editing is sometimes not detectable at all at some sites (see reference 20 hereby incorporated by reference in its entirety).
Bacterial and archaeal CRISPR-Cas systems rely on short guide RNAs in complex with Cas proteins to direct degradation of complementary sequences present within invading foreign nucleic acid. See Deltcheva, E. et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471, 602-607 (2011); Gasiunas, G., Barrangou, R., Horvath, P. &
Siksnys, V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA
cleavage for adaptive immunity in bacteria. Proceedings of the National Academy of Sciences of the United States of America 109, E2579-2586 (2012); Jinek, M. et al. A programmable dual-RNA-guided DNA
endonuclease in adaptive bacterial immunity. Science 337, 816-821 (2012);
Sapranauskas, R. et al.
The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli.
Nucleic acids research 39, 9275-9282 (2011); and Bhaya, D., Davison, M. &
Barrangou, R.
CRISPR-Cas systems in bacteria and archaea: versatile small RNAs for adaptive defense and regulation. Annual review of genetics 45, 273-297 (2011). A recent in vitro reconstitution of the S.
pyogenes type II CRISPR system demonstrated that crRNA ("CRISPR RNA") fused to a normally trans-encoded tracrRNA ("trans-activating CRISPR RNA") is sufficient to direct Cas9 protein to sequence-specifically cleave target DNA sequences matching the crRNA.
Expressing a gRNA
homologous to a target site results in Cas9 recruitment and degradation of the target DNA. See H.
Deveau et al., Phage response to CRISPR-encoded resistance in Streptococcus thermophilus.
Journal of Bacteriology 190, 1390 (Feb, 2008).
2
3 PCT/US2014/048140 SUMMARY
Aspects of the present disclosure are directed to the use of modified Transcription Activator-Like Effector Nucleases (TALENs) for genetically modifying a cell, such as a somatic cell or a stem cell. TALENs are known to include repeat sequences. Aspects of the present disclosure are directed to a method of altering target DNA in a cell including introducing into a cell a TALEN lacking repeat sequences 100 bp or longer wherein the TALEN cleaves the target DNA
and the cell undergoes nonhomologous end joining to produce altered DNA in the cell. According to certain aspects, repeat sequences of desired length have been removed from a TALEN.
According to certain aspects, the TALEN is devoid of repeat sequences of certain desired length.
According to certain aspects, a TALEN is provided with repeat sequences of desired length removed. According to certain aspects, a TALEN is modified to remove repeat sequences of desired length. According to certain aspects, a TALEN is engineered to remove repeat sequences of desired length.
Aspects of the present disclosure include methods of altering target DNA in a cell including combining within a cell a TALEN lacking repeat sequences 100 bp or longer and a donor nucleic acid sequence wherein the TALEN cleaves the target DNA and the donor nucleic acid sequence is inserted into the DNA in the cell. Aspects of the present disclosure are directed to a virus including a nucleic acid sequence encoding a TALEN lacking repeat sequences 100 bp or longer. Aspects of the present disclosure are directed to a cell including a nucleic acid sequence encoding a TALEN lacking repeat sequences 100 bp or longer. According to certain aspects described herein, the TALEN lacks repeat sequences 100 bp or longer, 90 bp or longer, 80 bp or longer, 70 bp or longer, 60 bp or longer, 50 bp or longer, 40 bp or longer, 30 bp or longer, 20 bp or longer, 19 bp or longer, 18 bp or longer, 17 bp or longer, 16 bp or longer, 15 bp or longer, 14 bp or longer, 13 bp or longer, 12 bp or longer, 11 bp or longer, or 10 bp or longer.
Aspects of the present disclosure are directed to making a TALE including combining an endonuclease, a DNA polymerase, a DNA ligase, an exonuclease, a plurality of nucleic acid dimer blocks encoding repeat variable diresidue domains and a TALE-N/TF backbone vector including an endonuclease cutting site, activating the endonuclease to cut the TALE-N/TF
backbone vector at the endonuclease cutting site to produce a first end and a second end, activating the exonuclease to create a 3' and a 5' overhang on the TALE-N/TF backbone vector and the plurality of nucleic acid dimer blocks and to anneal the TALE-N/TF backbone vector and the plurality of nucleic acid dimer blocks in a desired order, activating the DNA polymerase and the DNA
ligase to connect the TALE-N/TF backbone vector and the plurality of nucleic acid dimer blocks. One of skill in the art will readily based on the present disclosure be able to identify suitable endonucleases, DNA
polymerases, DNA ligases, exonucleases, nucleic acid dimer blocks encoding repeat variable diresidue domains and TALE-N/TF backbone vectors.

Aspects of the present disclosure are directed to a method of altering target DNA in a stem cell expressing an enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner including (a) introducing into the stem cell a first foreign nucleic acid encoding an RNA complementary to the target DNA
and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA, introducing into the stem cell a second foreign nucleic acid encoding a donor nucleic acid sequence, wherein the RNA and the donor nucleic acid sequences are expressed, wherein the RNA and the enzyme co-localize to the target DNA, the enzyme cleaves the target DNA and the donor nucleic acid is inserted into the target DNA to produce altered DNA in the stem cell.
Aspects of the present disclosure are directed to a stem cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA
complementary to target DNA and that cleaves the target DNA in a site specific manner.
Aspects of the present disclosure are directed to a cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA
complementary to target DNA and that cleaves the target DNA in a site specific manner and including an inducible promoter for promoting expression of the enzyme. In this manner, expression can be regulated, for example, it can be started and it can be stopped.
Aspects of the present disclosure are directed to a cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA
complementary to target DNA and that cleaves the target DNA in a site specific manner, wherein the first foreign nucleic acid is removable from genomic DNA of the cell using a removal enzyme, such as a transposase.
Aspects of the present disclosure are directed to a method of altering target DNA in a cell expressing an enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner including (a) introducing into the cell a first foreign nucleic acid encoding a donor nucleic acid sequence, introducing into the cell from media surrounding the cell an RNA complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA, wherein the donor nucleic acid sequence is expressed, wherein the RNA and the enzyme co-localize to the target DNA, the enzyme cleaves the target DNA and the donor nucleic acid is inserted into the target DNA to produce altered DNA in the cell.
Aspects of the present disclosure are directed to the use of an RNA guided DNA
binding protein for genetically modifying a stem cell. In one aspect, the stem cell has been genetically modified to include a nucleic acid encoding for the RNA guided DNA binding protein and the stem cell expresses the RNA guided DNA binding protein. According to a certain aspect, donor nucleic
4 acids for introducing specific mutations are optimized for genome editing using either the modified TALENs or the RNA guided DNA binding protein.
Aspects of the present disclosure are directed to the modification of DNA, such as multiplex modification of DNA, in a stem cell using one or more guide RNAs (ribonucleic acids) to direct an enzyme having nuclease activity expressed by the stem cell, such as a DNA binding protein having nuclease activity, to a target location on the DNA
(deoxyribonucleic acid) wherein the enzyme cuts the DNA and an exogenous donor nucleic acid is inserted into the DNA, such as by homologous recombination. Aspects of the present disclosure include cycling or repeating steps of DNA modification on a stem cell to create a stem cell having multiple modifications of DNA
within the cell. Modifications may include insertion of exogenous donor nucleic acids.
Multiple exogenous nucleic acid insertions can be accomplished by a single step of introducing into a stem cell, which expresses the enzyme, nucleic acids encoding a plurality of RNAs and a plurality of exogenous donor nucleic acids, such as by co-transformation, wherein the RNAs are expressed and wherein each RNA in the plurality guides the enzyme to a particular site of the DNA, the enzyme cuts the DNA and one of the plurality of exogenous nucleic acids is inserted into the DNA at the cut site. According to this aspect, many alterations or modification of the DNA in the cell are created in a single cycle.
Multiple exogenous nucleic acid insertions can be accomplished in a cell by repeated steps or cycles of introducing into a stem cell, which expresses the enzyme, one or more nucleic acids encoding one or more RNAs or a plurality of RNAs and one or more exogenous nucleic acids or a plurality of exogenous nucleic acids wherein the RNA is expressed and guides the enzyme to a particular site of the DNA, the enzyme cuts the DNA and the exogenous nucleic acid is inserted into the DNA at the cut site, so as to result in a cell having multiple alterations or insertions of exogenous DNA into the DNA within the stem cell. According to one aspect, the stem cell expressing the enzyme has been genetically altered to express the enzyme such as by introducing into the cell a nucleic acid encoding the enzyme and which can be expressed by the stem cell. In this manner, aspects of the present disclosure include cycling the steps of introducing RNA into a stem cell which expresses the enzyme, introducing exogenous donor nucleic acid into the stem cell, expressing the RNA, forming a co-localization complex of the RNA, the enzyme and the DNA, enzymatic cutting of the DNA by the enzyme, and insertion of the donor nucleic acid into the DNA. Cycling or repeating of the above steps results in multiplexed genetic modification of a stem cell at multiple loci, i.e., a stem cell having multiple genetic modifications.
According to certain aspects, DNA binding proteins or enzymes within the scope of the present disclosure include a protein that forms a complex with the guide RNA
and with the guide RNA guiding the complex to a double stranded DNA sequence wherein the complex binds to the DNA sequence. According to one aspect, the enzyme can be an RNA guided DNA
binding
5 protein, such as an RNA guided DNA binding protein of a Type II CRISPR System that binds to the DNA and is guided by RNA. According to one aspect, the RNA guided DNA
binding protein is a Cas9 protein.
This aspect of the present disclosure may be referred to as co-localization of the RNA and DNA binding protein to or with the double stranded DNA. In this manner, a DNA
binding protein-guide RNA complex may be used to cut multiple sites of the double stranded DNA
so as to create a stem cell with multiple genetic modifications, such as multiple insertions of exogenous donor DNA.
According to certain aspects, a method of making multiple alterations to target DNA in a stem cell expressing an enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner is provided including (a) introducing into the stem cell a first foreign nucleic acid encoding one or more RNAs complementary to the target DNA and which guide the enzyme to the target DNA, wherein the one or more RNAs and the enzyme are members of a co-localization complex for the target DNA, introducing into the stem cell a second foreign nucleic acid encoding one or more donor nucleic acid sequences, wherein the one or more RNAs and the one or more donor nucleic acid sequences are expressed, wherein the one or more RNAs and the enzyme co-localize to the target DNA, the enzyme cleaves the target DNA and the donor nucleic acid is inserted into the target DNA to produce altered DNA in the stem cell, and repeating step (a) multiple times to produce multiple alterations to the DNA in the stem cell.
According to one aspect, the RNA is between about 10 to about 500 nucleotides.

According to one aspect, the RNA is between about 20 to about 100 nucleotides.
According to one aspect, the one or more RNAs is a guide RNA. According to one aspect, the one or more RNAs is a tracrRNA-crRNA fusion.
According to one aspect, the DNA is genomic DNA, mitochondrial DNA, viral DNA, or exogenous DNA.
According to one aspect, a cell may be genetically modified to reversibly include a nucleic acid encoding a DNA binding enzyme using a vector which can be easily removed using an enzyme. Useful vectors methods are known to those of skill in the art and include lentivirus, adeno associated virus, nuclease and integrase mediated tarteget insertion methods and transposon mediated insertion methods. According to one aspect, the nucleic acid encoding a DNA binding enzyme that has been added, such as by using a cassette or vector can be removed in its entirety along with the cassette and vector and without leaving a portion of such nucleic acid, cassette or vector in the genomic DNA, for example. Such removal is referred to in the art as "scarless"
removal, as the genome is the same as it was before addition of the nucleic acid, cassette or vector.
6 One exemplary embodiment for insertion and scarless removal is a PiggyBac vector commercially available from System Biosciences.
Further features and advantages of certain embodiments of the present invention will become more fully apparent in the following description of embodiments and drawings thereof, and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
The foregoing and other features and advantages of the present embodiments will be more fully understood from the following detailed description of illustrative embodiments taken in conjunction with the accompanying drawings in which:
Fig. 1 is directed to functional tests of re-TALENs in human somatic and stem cells.
(a) Schematic representation of experimental design for testing genome targeting efficiency. A
genomically integrated GFP coding sequence is disrupted by the insertion of a stop codon and a 68bp genomic fragment derived from the AAVS1 locus (bottom). Restoration of the GFP sequence by nuclease-mediated homologous recombination with tGFP donor (top) results in GFP+ cells that can be quantitated by FACS. Re-TALENs and TALENs target identical sequences within AAVS1 fragments.
(b) Bar graph depicting GFP+ cell percentage introduced by tGFP donor alone, TALENs with tGFP donor, and re-TALENs with tGFP donor at the target locus, as measured by FACS. (N=3, error bar =SD) Representative FACS plots are shown below.
(c) Schematic overview depicting the targeting strategy for the native AAVS1 locus. The donor plasmid, containing splicing acceptor (SA)- 2A (self-cleaving peptides), puromycin resistant gene (PURO) and GFP were described (see reference 10 hereby incorporated by reference in its entirety.
The locations of PCR primers used to detect successful editing events are depicted as blue arrows.
(d) Successfully targeted clones of PGP1 hiPSCs were selected with puromycin (0.5ug/mL) for 2 weeks. Microscopy images of three representative GFP+ clones are shown. Cells were also stained for the pluripotency markers TRA-1-60. Scale bar: 200 !um.
(e) PCR assays performed on these the monoclonal GFP+ hiPSC clones demonstrated successful insertions of the donor cassettes at the AAVS1 site (lane 1,2,3), whereas plain hiPSCs show no evidence of successful insertion (lane C).
Fig. 2 relates to a comparison of reTALENs and Cas9-gRNA5 genome targeting efficiency on CCR5 in iPSCs.
(a) Schematic representation of genome engineering experimental design. At the re-TALEN pair or Cas9-gRNA targeting site, a 90mer ssODN carrying a 2bp mismatch against the genomic DNA
was delivered along with the reTALEN or Cas9-gRNA constructs into PGP1 hiPSCs.
The cutting sites of the nucleases are depicted as red arrows in the figure.
7 (b) Deep sequencing analysis of HDR and NHEJ efficiencies for re-TALEN pairs (CCR5 #3) and ssODN, or the Cas9-gRNA and ssODN. Alterations in the genome of hiPSCs were analyzed from high-throughput sequence data by GEAS. Top: HDR was quantified from the fraction of reads that contained a 2bp point mutation built into the center of the ssODN (blue), and NHEJ activity was quantified from the fraction of deletions (grey)/Insertions (red) at each specific position in the genome. For the reTALEN and ssODN graphs, green dashed lines are plotted to mark the outer boundary of the re-TALEN pair's binding sites, which are at positions -26bp and +26bp relative to the center of the two re-TALEN binding sites. For Cas9-gRNA and ssODN graphs, the green dashed lines mark the outer boundary of the gRNA targeting site, which are at positions -20 and -1 bp relative to the PAM sequence. Bottom: Deletion/Insertion size distribution in hiPSCs analyzed from the entire NHEJ population with treatments indicated above.
(c) The genome editing efficiency of re-TALENs and Cas9-gRNA5 targeting CCR5 in PGP1 hiPSCs.
Top: schematic representation of the targeted genome editing sites in CCR5.
The 15 targeting sites are illustrated by blue arrows below. For each site, cells were co-transfected with a pair of re-TALENs and their corresponding ssODN donor carrying 2bp mismatches against the genomic DNA. Genome editing efficiencies were assayed 6 days after transfection.
Similarly, 15 Cas9-gRNAs were transfected with their corresponding ssODNs individually into PGP1-hiPSCs to target the same 15 sites and analyzed the efficiency 6 days after transfection.
Bottom: the genome editing efficiency of re-TALENs and Cas9-gRNA5 targeting CCR5 in PGP1 hiPSCs. Panel 1 and 2 indicate NHEJ and HDR efficiencies mediated by reTALENs. Panel 3 and 4 indicate NHEJ and HDR efficiencies mediated by Cas9-gRNA5. NHEJ rates were calculated by the frequency of genomic alleles carrying deletions or insertions at the targeting region; HDR
rates were calculated by the frequency of genomic alleles carrying 2bp mismatches. Panel 5, the DNaseI HS profile of a hiPSC cell line from ENCODE database (Duke DNase HS, iPS NIHi7 DS). Of note, the scales of different panels are different.
(f) Sanger sequencing of the PCR amplicon from the three targeted hiPSC
colonies confirmed that the expected DNA bases at the genome-insertion boundary is present. M: DNA
ladder. C: control with plain hiPSCs genomic DNA.
Fig. 3 is directed to a study of functional parameters governing ssODN-mediated HDR
with re-TALENs ir Cas9-gRNA5 in PGP1 hiPSCs.
(a) PGP1 hiPSCs were co-transfected with re-TALENs pair (#3) and ssODNs of different lengths (50, 70, 90,110, 130,150,170 nts). All ssODNs possessed an identical 2bp mismatch against the genomic DNA in the middle of their sequence. A 90mer ssODN achieved optimal HDR in the targeted genome. The assessment of HDR, NHEJ-incurred deletion and insertion efficiency is as described herein.
8 (b) 90mer ssODNs corresponding to re-TALEN pair #3 each containing a 2bp mismatch (A) in the center and an additional 2bp mismatch (B) at different positions offset from A
(where offsets varied from -30bp430bp) were used to test the effects of deviations from homology along the ssODN. Genome editing efficiency of each ssODN was assessed in PGP1 hiPSCs.
The bottom bar graph shows the incorporation frequency of A only, B only, and A + B in the targeted genome.
HDR rates decrease as the distance of homology deviations from the center increase.
(c) ssODNs targeted to sites with varying distances (-620bp¨ 480bp) away from the target site of re-TALEN pair #3 were tested to assess the maximum distance within which ssODNs can be placed to introduce mutations. All ssODNs carried a 2bp mismatch in the middle of their sequences.
Minimal HDR efficiency (<=0.06%) was observed when the ssODN mismatch was positioned 40bp away from the middle of re-TALEN pair's binding site.
(d) PGP1 hiPSCs were co-transfected with Cas9-gRNA (AAVS1) and ssODNs of different orientation (Oc: complement to gRNA; On: non-complement to gRNA) and different lengths (30, 50, 70, 90, 110 nt). All ssODNs possessed an identical 2bp mismatch against the genomic DNA in the middle of their sequence. A 70mer Oc achieved optimal HDR in the targeted genome.
Fig. 4 is directed to using re-TALENs and ssODNs to obtain monoclonal genome edited hiPSC without selection.
(a) Timeline of the experiment.
(b) Genome engineering efficiency of re-TALENs pair and ssODN (#3) assessed by the NGS
platform described in Figure 2b.
(c) Sanger sequencing results of monoclonal hiPSC colonies after genome editing. The 2bp heterogeneous genotype (CT/CT-TA/CT) was successfully introduced into the genome of PGP1-iPS-3-11, PGP1-iPS-3-13 colonies.
(d) Immunofluorescence staining of targeted PGP1-iPS-3-11. Cells were stained for the pluripotency markers Tra-1-60 and SSEA4.
(e) Hematoxylin and eosin staining of teratoma sections generated from monoclonal PGP1-iPS-3-11 cells.
Fig. 5. Design of reTALE. (a) Sequence alignment of the original TALE RVD
monomer with monomers in re-TALE-16.5 (re-TALE-MI 4re-TALE-M17). Nucleotide alterations from the original sequence are highlighted in gray. (b) Test of repetitiveness of re-TALE by PCR. Top panel illustrates the structure of re-TALE/TALE and positions of the primers in the PCR reaction.
Bottom panel illustrates PCR bands with condition indicated below. The PCR
laddering presents with the original TALE template (right lane).
Fig. 6. Design and practice of TALE Single-incubation Assembly (TASA) assembly.
(a) Schematic representation of the library of re-TALE dimer blocks for TASA
assembly. There is a library of 10 re-TALE dimer blocks encoding two RVDs. Within each block, all 16 dimers share
9 the same DNA sequence except the RVD encoding sequences; Dimers in different blocks have distinct sequences but are designed such that they share 32bp overlaps with the adjacent blocks.
DNA and amino acid sequence of one dimer (Block6_AC) are listed on the right.
(b) Schematic representation of TASA assembly. The left panel illustrates the TASA assembly method: a one-pot incubation reaction is conducted with an enzyme mixture/re-TALE blocks/re-TALE-N/TF backbone vectors. The reaction product can be used directly for bacterial transformation. The right panel illustrates the mechanism of TASA. The destination vector is linearized by an endonuclease at 37 C to cut off ccdB counter-selection cassette; the exonuclease, which processes the end of blocks and linearized vectors, exposes ssDNA
overhangs at the end of fragments to allow blocks and vector backbones to anneal in a designated order. When the temperature rises up to 50 C, polymerases and ligases work together to seal the gap, producing the final constructs ready for transformation.
(c) TASA assembly efficiency for re-TALEs possessing different monomer lengths. The blocks used for assembly are illustrated on the left and the assembly efficiency is presented on the right.
Fig. 7. The functionality and sequence integrity of Lenti-reTALEs.
Fig 8. The sensitivity and reproducibility of GEAS.
(A) Information-based analysis of HDR detection limit. Given the dataset of re-TALENs (#10)/ssODN, the reads containing the expected editing (HDR) were identified and these HDR
reads were systematically removed to generate different artificial datasets with a "diluted" editing signal. Datasets with 100, 99.8, 99.9, 98.9, 97.8, 89.2, 78.4, 64.9, 21.6,
10.8, 2.2, 1.1, 0.2, 0.1, 0.02, and 0% removal of HDR reads were generated to generate artificial datasets with HR efficiency ranging from 0-0.67%. For each individual dataset, mutual information (MI) of the background signal (in purple) and the signal obtained in the targeting site (in green) was estimated. MI at the targeting site is remarkably higher than the background when the HDR
efficiency is above 0.0014%. A limit of HDR detection between 0.0014% and 0.0071% was estimated.
MI calculation is described herein.
(B) The test of reproducibility of genome editing assessment system. The pairs of plots (Top and Bottom) show the HDR and NHEJ assessment results of two replicates with re-TALENs pair and cell type indicated above. For each experiment, nucleofection, targeted genome amplification, deep-sequencing and data analysis were conducted independently. The genome editing assessment variation of replicates was calculated as -\12 (1HDR1-HDR21)/((HDR+HDR2)/2) =AHDR/HDR and -\12 (INHEJ1-NHEJ21) ANHEJ1+NHEJ2)/2) =ANHEJ/NHEJ and the variation results are listed below the plots. The average variation of the system was (19%+11%+4%+9%+10%+35%)/6=15%.
Factors that may contribute to the variations include the status of cells under nucleofection, nucleofection efficiency, and sequencing coverage and quality.

Fig. 9. Statistical analysis of NHEJ and HDR efficiencies by reTALENs and Cas9-gRNAs on CCR5.
(a) The correlation of HR and NHEJ efficiencies mediated by reTALENs at identical sites in iPSCs (r=0.91, P< 1X 10-5).
(b) The correlation of HR and NHEJ efficiencies mediated by Cas9-gRNA at identical sites in iPSCs (r=0.74, P=0.002).
(c) The correlation of NHEJ efficiencies mediated by Cas9-gRNA and the Tm temperature of gRNA targeting site in iPSCs (r=0.52, P=0.04) Fig. 10. The correlation analysis of genome editing efficiency and epigenetic state.
Pearson correlation was used to study possible associations between DNase I
sensitivity and genome engineering efficiencies (HR, NHEJ). The observed correlation was compared to a randomized set (N=100000). Observed correlations higher than the 95th percentile, or lower than the 5th percentile of the simulated distribution were considered as potential associations. No remarkable correlation between DNasel sensitiivty and NHEJ/HR efficiencies was observed.
Fig. 11. The impact of homology pairing in the ssODN-mediated genome editing.
(a) In the experiment described in Figure 3b, overall HDR as measured by the rate at which the middle 2b mismatch (A) was incorporated decreased as the secondary mismatches B increased their distance from the A (relative position of B to A varies from -30430bp).
The higher rates of incorporation when B is only 10bp away from A (-10bp and +10b) may reflect a lesser need for pairing of the ssODN against genomic DNA proximal to the dsDNA break.
(b) Distribution of gene conversion lengths along the ssODN. At each distance of B from A, a fraction of HDR events incorporates only A while another fraction incorporates both A and B.
These two events may be interpretable in terms of gene conversion tracts (Elliott et al., 1998), whereby A+B events represent long conversion tracts that extend beyond B and A-only events represent shorter ones that do not reach to B. Under this interpretation, a distribution of gene conversion lengths in both directions along the oligo can be estimated (the middle of ssODN is defined as 0, conversion tracks towards the 5' end of ssODN as - direction, and 3' end as +
direction). Gene conversion tracts progressively decrease in incidence as their lengths increase, a result very similar to gene conversion tract distributions seen with dsDNA
donors, but on a highly compressed distance scale of tens of bp for the ssDNA oligo vs. hundreds of bases for dsDNA
donors.
(c) Assays for gene conversion tracts using a single ssODN that contains a series of mutations and measuring contiguous series of incorporations. A ssODN donor with three pairs of 2bp mismatches (orange) spaced at intervals of lOnt on either side of the central 2bp mismatch (Top) was used. Few genomic sequencing reads were detected (see reference 62 hereby incorporated by reference in its entirety) carrying >=1 mismatches defined by ssODN among >300,000 reads sequencing this
11 region. All these reads were plotted (bottom) and the sequence of the reads was color coded.
Orange: defined mismatches; green: wild type sequence. Genome editing with this ssODN gave rise to a pattern in which middle mutation alone was incorporated 85% (53/62) of the time, with multiple B mismatches incorporated at other times. Although numbers of B
incorporation events were too low to estimate a distribution of tract lengths > 10bp, it is clear that the short tract region from - 10- 10bp predominates.
Fig. 12. Cas9-gRNA nuclease and nickases genome editing efficiencies.
PGP1 iPSCs were co-transfected with combination of nuclease (C2) (Cas9-gRNA) or nickase (Cc) (Cas9D10A-gRNA) and ssODNs of different orientation (Oc and On). All ssODNs possessed an identical 2bp mismatch against the genomic DNA in the middle of their sequence. The assessment of HDR is described herein.
Fig. 13. The design and optimization of re-TALE sequence.
The re-TALE sequence was evolved in several design cycles to eliminate repeats. In each cycle, synonymous sequences from each repeat are evaluated. Those with the largest hamming distance to the evolving DNA are selected. The final sequence with cai = 0.59 AG= -9.8 kcal/mol. An R package was provided to carry out this general framework for synthetic protein design.
Fig. 14 is a gel image showing PCR validation of the genomic insertion of Cas 9 in PGP1 cells. Line 3, 6, 9, 12 are PCR product of plain PGP1 cell lines.
Fig. 15 is a graph of the mRNA expression level of Cas9 mRNA under the induction.
Fig. 16 is a graph showing genome targeting efficiency by different RNA
designs.
Fig. 17 is a graph showing genome targeting efficiency of 44% homologous recombination achieved by a guide RNA ¨ donor DNA fusion.
Fig. 18 is a diagram showing the genotype of isogenic PGP1 cell lines generated by system decribed herein. PGP1-iPS-BTHH has the single nucleotides deletion phenotype as the BTHH
patient. PGP1-NHEJ has 4bp deletions that generated frame-shift mutations in a different way.
Fig. 19 is a graph showing that cardiomyocyte derived from isogenic PGP1 iPS
recapitulated defective ATP production and FIFO ATPase specific activity as demonstrated in patient specific cells.
DETAILED DESCRIPTION
Aspects of the present invention are directed to the use of a TALEN that lacks certain repeat sequences, for nucleic acid engineering, for example by cutting double stranded nucleic acid. The use of the TALEN to cut double stranded nucleic acid can result in nonhomologous end joining (NHEJ) or homologous recombination (HR). Aspects of the present disclosure also
12 contemplate the use of a TALEN that lacks repeat sequences for nucleic acid engineering, for example by cutting double stranded nucleic acid, in the presence of a donor nucleic acid and insertion of the donor nucleic acid into the double stranded nucleic acid, such as by nonhomologous end joining (NHEJ) or homologous recombination (HR).
Transcription activator-like effector nucleases (TALENs) are known in the art and include artificial restriction enzymes generated by fusing a TAL effector DNA binding domain to a DNA
cleavage domain. Restriction enzymes are enzymes that cut DNA strands at a specific sequence.
Transcription activator-like effectors (TALEs) can be engineered to bind to a desired DNA
sequence. See Boch, Jens (February 2011). "TALEs of genome targeting". Nature Biotechnology 29 (2): 135-6 hereby incorporated by reference in its entirety. By combining such an engineered TALE with a DNA cleavage domain (which cuts DNA strands), a TALEN is produced which is a restriction enzyme that is specific for any desired DNA sequence. According to certain aspects, the TALEN is introduced into a cell for target nucleic acid editing in situ, such as genome editing in situ.
According to one aspect, the non-specific DNA cleavage domain from the end of the FokI
endonuclease can be used to construct hybrid nucleases that are active in yeast cells, plant cells and animal cells. The FokI domain functions as a dimer, requiring two constructs with unique DNA
binding domains for sites in the target genome with proper orientation and spacing. Both the number of amino acid residues between the TALE DNA binding domain and the FokI
cleavage domain and the number of bases between the two individual TALEN binding sites affect activity.
The relationship between amino acid sequence and DNA recognition of the TALE
binding domain allows for designable proteins. Software programs such as DNAWorks can be used to design TALE constructs. Other methods of designing TALE constructs are known to those of skill in the art. See Cermak, T.; Doyle, E. L.; Christian, M.; Wang, L.; Zhang, Y.;
Schmidt, C.; Baller, J. A.; Somia, N. V. et al. (2011). "Efficient design and assembly of custom TALEN and other TAL
effector-based constructs for DNA targeting". Nucleic Acids Research.
doi:10.1093/nar/gkr218;
Zhang, Feng; et.al. (February 2011). "Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription", Nature Biotechnology 29 (2): 149-53;
Morbitzer, R.;
Elsaesser, J.; Hausner, J.; Lahaye, T. (2011). "Assembly of custom TALE-type DNA binding domains by modular cloning". Nucleic Acids Research. doi:10'1093/nar/gkr151;
Li, T.; Huang, S.;
Zhao, X.; Wright, D. A.; Carpenter, S.; Spalding, M. H.; Weeks, D. P.; Yang, B. (2011).
"Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes". Nucleic Acids Research. doi"10.1093/nar/gkr188;
GeiPler, R.;
Scholze, H.; Hahn, S.; Streubel, J.; Bonas, U.; Behrens, S. E.; Boch, J.
(2011). "Transcriptional
13 Activators of Human Genes with Programmable DNA-Specificity". In Shiu, Shin-Han. PLoS ONE
6 (5): e19509; Weber, E.; Gruetzner, R.; Werner, S.; Engler, C.; Marillonnet, S. (2011).
"Assembly of Designer TAL Effectors by Golden Gate Cloning". In Bendahmane, Mohammed.
PLoS ONE 6 (5): e19722 hereby incorporated by reference in their entireties.
According to an exemplary aspect, once the TALEN genes have been assembled they may inserted into plasmids according to certain embodiments; the plasmids are then used to transfect the target cell where the gene products are expressed and enter the nucleus to access the genome.
According to exemplary aspects, TALENs as described herein can be used to edit target nucleic acids, such as genomes, by inducing double-strand breaks (DSB), which cells respond to with repair mechanisms. Exemplary repair mechanisms include non-homologous end joining (NHEJ) which reconnects DNA from either side of a double-strand break where there is very little or no sequence overlap for annealing. This repair mechanism induces errors in the genome via insertion or deletion (indels), or chromosomal rearrangement; any such errors may render the gene products coded at that location non-functional. See Miller, Jeffrey; et.al. (February 2011). "A TALE
nuclease architecture for efficient genome editing". Nature Biotechnology 29 (2): 143-8 hereby incorporated by reference in its entirety. Because this activity can vary depending on the species, cell type, target gene, and nuclease used, the activity can be monitored by using a heteroduplex cleavage assay which detects any difference between two alleles amplified by PCR. Cleavage products can be visualized on simple agarose gels or slab gel systems.
Alternatively, DNA can be introduced into a genome through NHEJ in the presence of exogenous double-stranded DNA fragments. Homology directed repair can also introduce foreign DNA at the DSB as the transfected double-stranded sequences are used as templates for the repair enzymes. According to certain aspects the TALENs described herein can be used to generate stably modified human embryonic stem cell and induced pluripotent stem cell (IPSCs) clones.
According to certain aspects the TALENs described herein can be used to generate knockout species such as C. elegans, knockout rats, knockout mice or knockout zebrafish.
According to one aspect of the present disclosure, embodiments are directed to the use of exogenous DNA, nuclease enzymes such as DNA binding proteins and guide RNAs to co-localize to DNA within a stem cell and digest or cut the DNA with insertion of the exogenous DNA. Such DNA binding proteins are readily known to those of skill in the art to bind to DNA for various purposes. Such DNA binding proteins may be naturally occurring. DNA binding proteins included within the scope of the present disclosure include those which may be guided by RNA, referred to herein as guide RNA. According to this aspect, the guide RNA and the RNA guided DNA binding protein form a co-localization complex at the DNA. Such DNA
binding proteins
14 having nuclease activity are known to those of skill in the art, and include naturally occurring DNA
binding proteins having nuclease activity, such as Cas9 proteins present, for example, in Type II
CRISPR systems. Such Cas9 proteins and Type II CRISPR systems are well documented in the art. See Makarova et al., Nature Reviews, Microbiology, Vol. 9, June 2011, pp.
467-477 including all supplementary information hereby incorporated by reference in its entirety.
Exemplary DNA binding proteins having nuclease activity function to nick or cut double stranded DNA. Such nuclease activity may result from the DNA binding protein having one or more polypeptide sequences exhibiting nuclease activity. Such exemplary DNA
binding proteins may have two separate nuclease domains with each domain responsible for cutting or nicking a particular strand of the double stranded DNA. Exemplary polypeptide sequences having nuclease activity known to those of skill in the art include the McrA-HNH nuclease related domain and the RuvC-like nuclease domain. Accordingly, exemplary DNA binding proteins are those that in nature contain one or more of the McrA-HNH nuclease related domain and the RuvC-like nuclease domain.
An exemplary DNA binding protein is an RNA guided DNA binding protein of a Type II
CRISPR System. An exemplary DNA binding protein is a Cas9 protein.
In S. pyogenes, Cas9 generates a blunt-ended double-stranded break 3bp upstream of the protospacer-adjacent motif (PAM) via a process mediated by two catalytic domains in the protein:
an HNH domain that cleaves the complementary strand of the DNA and a RuvC-like domain that cleaves the non-complementary strand. See Jinke et al., Science 337, 816-821 (2012) hereby incorporated by reference in its entirety. Cas9 proteins are known to exist in many Type II
CRISPR systems including the following as identified in the supplementary information to Makarova et al., Nature Reviews, Microbiology, Vol. 9, June 2011, pp. 467-477:
Methanococcus maripaludis C7; Corynebacterium diphtheriae; Corynebacterium efficiens YS-314;
Corynebacterium glutamicum ATCC 13032 Kitasato; Corynebacterium glutamicum Bielefeld; Corynebacterium glutamicum R; Corynebacterium kroppenstedtii DSM
44385;
Mycobacterium abscessus ATCC 19977; Nocardia farcinica IFM10152; Rhodococcus erythropolis PR4; Rhodococcus jostii RHAl; Rhodococcus opacus B4 uid36573; Acidothermus cellulolyticus 11B; Arthrobacter chlorophenolicus A6; Kribbella flavida DSM 17836 uid43465;
Thermomonospora curvata DSM 43183; Bifidobacterium dentium Bdl;
Bifidobacterium longum DJ010A; Slackia heliotrinireducens DSM 20476; PersephoneIla marina EX Hi;
Bacteroides fragilis NCTC 9434; Capnocytophaga ochracea DSM 7271; Flavobacterium psychrophilum JIP02 86; Akkermansia muciniphila ATCC BAA 835; Roseiflexus castenholzii DSM 13941;
Roseiflexus RS1; Synechocystis PCC6803; Elusimicrobium minutum Pei191; uncultured Termite group 1 bacterium phylotype Rs D17; Fibrobacter succinogenes S85; Bacillus cereus ATCC
10987;

Listeria innocua;Lactobacillus casei; Lactobacillus rhamnosus GG;
Lactobacillus salivarius UCC118; Streptococcus agalactiae A909; Streptococcus agalactiae NEM316;
Streptococcus agalactiae 2603; Streptococcus dysgalactiae equisimilis GGS 124; Streptococcus equi zooepidemicus MGC510565; Streptococcus gallolyticus UCN34 uid46061;
Streptococcus gordonii Challis subst CH1; Streptococcus mutans NN2025 uid46353; Streptococcus mutans;
Streptococcus pyogenes M1 GAS; Streptococcus pyogenes MGAS5005; Streptococcus pyogenes MGA52096;
Streptococcus pyogenes MGA59429; Streptococcus pyogenes MGAS10270;
Streptococcus pyogenes MGAS6180; Streptococcus pyogenes MGAS315; Streptococcus pyogenes 55I-1;
Streptococcus pyogenes MGAS 10750; Streptococcus pyogenes NZ131; Streptococcus thermophiles CNRZ1066; Streptococcus thermophiles LMD-9; Streptococcus thermophiles LMG
18311; Clostridium botulinum A3 Loch Maree; Clostridium botulinum B Eklund 17B; Clostridium botulinum Ba4 657; Clostridium botulinum F Langeland; Clostridium cellulolyticum H10;
Finegoldia magna ATCC 29328; Eubacterium rectale ATCC 33656; Mycoplasma gallisepticum;
Mycoplasma mobile 163K; Mycoplasma penetrans; Mycoplasma synoviae 53;
Streptobacillus moniliformis DSM 12112; Bradyrhizobium BTAil; Nitrobacter hamburgensis X14;
Rhodopseudomonas palustris BisB18; Rhodopseudomonas palustris BisB5;
Parvibaculum lavamentivorans DS-1; Dinoroseobacter shibae DFL 12; Gluconacetobacter diazotrophicus Pal 5 FAPERJ; Gluconacetobacter diazotrophicus Pal 5 JGI; Azospirillum B510 uid46085;
Rhodospirillum rubrum ATCC 11170; Diaphorobacter TPSY uid29975;
Verminephrobacter eiseniae EF01-2; Neisseria meningitides 053442; Neisseria meningitides alpha 14; Neisseria meningitides Z2491; Desulfovibrio salexigens DSM 2638; Campylobacter jejuni doylei 269 97;
Campylobacter jejuni 81116; Campylobacter jejuni; Campylobacter lari RM2100;
Helicobacter hepaticus; Wolinella succinogenes; Tolumonas auensis DSM 9187;
Pseudoalteromonas atlantica T6c; Shewanella pealeana ATCC 700345; Legionella pneumophila Paris;
Actinobacillus succinogenes 130Z; Pasteurella multocida; Francisella tularensis novicida U112; Francisella tularensis holarctica; Francisella tularensis FSC 198; Francisella tularensis tularensis; Francisella tularensis WY96-3418; and Treponema denticola ATCC 35405. Accordingly, aspects of the present disclosure are directed to a Cas9 protein present in a Type II CRISPR
system.
The Cas9 protein may be referred by one of skill in the art in the literature as Csnl. The S.
pyogenes Cas9 protein is shown below. See Deltcheva et al., Nature 471, 602-607 (2011) hereby incorporated by reference in its entirety.
MDKKY SI GLDIGTN SVGWAVITDEYKVP SKKFKVLGNTD RH S IKKNLIGALLFD S GETAE
ATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFG
NIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSD

VDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGN
LIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI
LL SD ILRVNTEITKAPL SAS MIKRYDEHHQ DLTLLKALVRQ QLPEKYKEIFFD Q SKNGYA
GYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELH
AILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEE
VVDKGASAQ S FIERMTNFDKNLPNEKVLPKH SLLYEYFTVYNELTKVKYVTEGMRKPAFL
SGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFD SVEIS GVEDRFNASLGTYHDLLKI
IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWG
RLSRKLINGIRDKQ S GKTILDFLK SD GFANRNFMQLIHDD SLTFKEDIQKAQVS GQGD SL
HEHIANLAG SPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKN S RER
MKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDH
IVPQ SFLKDD SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNL
TKAERGGLSELDKAGFIKRQLVETRQITKHVAQILD SRMNTKYDENDKLIREVKVITLKS
KLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVR
K
MIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDF
ATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFD SPTVA
YSVLVVAKVEKGKSKKLKSVKELLGITIMERS SFEKNPIDFLEAKGYKEVKKDLIIKLPK
YSLFELENGRKRMLASAGELQKGNELALP SKYVNFLYLASHYEKLKGSPEDNEQKQLFVE
QHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGA
PAAFKYFDTTIDRKRYTSTKEVLDATLIHQ SITGLYETRIDL S Q LGGD -According to one aspect, the RNA guided DNA binding protein includes homologs and orthologs of Cas9 which retain the ability of the protein to bind to the DNA, be guided by the RNA
and cut the DNA. According to one aspect, the Cas9 protein includes the sequence as set forth for naturally occurring Cas9 from S. pyogenes and protein sequences having at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% homology thereto and being a DNA binding protein, such as an RNA guided DNA binding protein.
According to one aspect, an engineered Cas9-gRNA system is provided which enables RNA-guided genome cutting in a site specific manner in a stem cell, if desired, and modification of the stem cell genome by insertion of exogenous donor nucleic acids. The guide RNAs are complementary to target sites or target loci on the DNA. The guide RNAs can be crRNA-tracrRNA chimeras. The guide RNAs can be introduced from media surrounding the cell. In this manner a method of continuously modifying a cell is provided to the extent that various guide RNAs are provided to surrounding media and with the uptake by the cell of the guide RNAs and with supplementation of the media with additional guide RNAs. Supplementation may be in a continuous manner. The Cas9 binds at or near target genomic DNA. The one or more guide RNAs bind at or near target genomic DNA. The Cas9 cuts the target genomic DNA
and exogenous donor DNA is inserted into the DNA at the cut site.
Accordingly, methods are directed to the use of a guide RNA with a Cas9 protein and an exogenous donor nucleic acid to multiplex insertions of exogenous donor nucleic acids into DNA
within a stem cell expressing Cas9 by cycling the insertion of nucleic acid encoding the RNA (or providing RNA from the surrounding media) and exogenous donor nucleic acid, expressing the RNA (or uptaking the RNA), colocalizing the RNA, Cas9 and DNA in a manner to cut the DNA, and insertion of the exogenous donor nucleic acid. The method steps can be cycled in any desired number to result in any desired number of DNA modifications. Methods of the present disclosure are accordingly directed to editing target genes using the Cas9 proteins and guide RNAs described herein to provide multiplex genetic and epigenetic engineering of stem cells.
Further aspects of the present disclosure are directed to the use of DNA
binding proteins or systems (such as the modified TALENS or Cas9 described herein) in general for the multiplex insertion of exogenous donor nucleic acids into the DNA, such as genomic DNA, of a stem cell, such as a human stem cell. One of skill in the art will readily identify exemplary DNA binding systems based on the present disclosure.
Cells according to the present disclosure unless otherwise specified include any cell into which foreign nucleic acids can be introduced and expressed as described herein. It is to be understood that the basic concepts of the present disclosure described herein are not limited by cell type. Cells according to the present disclosure include somatic cells, stem cells, eukaryotic cells, prokaryotic cells, animal cells, plant cells, fungal cells, archael cells, eubacterial cells and the like.
Cells include eukaryotic cells such as yeast cells, plant cells, and animal cells. Particular cells include mammalian cells, such as human cells. Further, cells include any in which it would be beneficial or desirable to modify DNA.
Target nucleic acids include any nucleic acid sequence to which a TALEN or RNA
guided DNA binding protein having nuclease activity as described herein can be useful to nick or cut.
Target nucleic acids include any nucleic acid sequence to which a co-localization complex as described herein can be useful to nick or cut. Target nucleic acids include genes. For purposes of the present disclosure, DNA, such as double stranded DNA, can include the target nucleic acid and a co-localization complex can bind to or otherwise co-localize with the DNA or a TALEN can otherwise bind with the DNA at or adjacent or near the target nucleic acid and in a manner in which the co-localization complex or the TALEN may have a desired effect on the target nucleic acid. Such target nucleic acids can include endogenous (or naturally occurring) nucleic acids and exogenous (or foreign) nucleic acids. One of skill based on the present disclosure will readily be able to identify or design guide RNAs and Cas9 proteins which co-localize to a DNA or a TALEN

which binds to a DNA, including a target nucleic acid. One of skill will further be able to identify transcriptional regulator proteins or domains, such as transcriptional activators or transcriptional repressors, which likewise co-localize to a DNA including a target nucleic acid. DNA includes genomic DNA, mitochondrial DNA, viral DNA or exogenous DNA. According to one aspect, materials and methods useful in the practice of the present disclosure include those described in Di Carlo, et al., Nucleic Acids Research, 2013, vol. 41, No. 7 4336-4343 hereby incorporated by reference in its entirety for all purposes including exemplary strains and media, plasmid construction, transformation of plasmids, electroporation of transcient gRNA
cassette and donor nucleic acids, transformation of gRNA plasmid with donor DNA into Cas9-expressing cells, galactose induction of Cas9, identification of CRISPR-Cas targets in yeast genome, etc.
Additional references including information, materials and methods useful to one of skill in carrying out the invention are provided in Mali,P., Yang,L., Esvelt,K.M., Aach,J., Guell,M., DiCarlo,J.E., Norville,J.E. and Church,G.M. (2013) RNA-Guided human genome engineering via C as 9. Science, 10. 1126 fscience .1232033 ; Storici,F., Durham,C . L., Gordenin,D.A. and Resnick,M.A. (2003) Chromosomal site-specific double-strand breaks are efficiently targeted for repair by oligonucleotides in yeast. PNAS, 100, 14994-14999 and Jinek,M., Chylinski,K., Fonfara,1., Hauer,M., Doudna,J.A. and Charpentier,E. (2012) A programmable dual-RNA-Guided DNA endonuclease in adaptive bacterial immunity. Science, 337, 816-821 each of which are hereby incorporated by reference in their entireties for all purposes.
Foreign nucleic acids (i.e. those which are not part of a cell's natural nucleic acid composition) may be introduced into a cell using any method known to those skilled in the art for such introduction.
Such methods include transfection, transduction, viral transduction, microinjection, lipofection, nucleofection, nanoparticle bombardment, transformation, conjugation and the like. One of skill in the art will readily understand and adapt such methods using readily identifiable literature sources.
Donor nucleic acids include any nucleic acid to be inserted into a nucleic acid sequence as described herein.
The following examples are set forth as being representative of the present disclosure.
These examples are not to be construed as limiting the scope of the present disclosure as these and other equivalent embodiments will be apparent in view of the present disclosure, figures and accompanying claims.

EXAMPLE I
Guide RNA Assembly 19bp of the selected target sequence (i.e. 5'-N19 of 5'-N19-NGG-3') were incorporated into two complementary 100mer oligonucleotides (TTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCGN19GTTTTAGAGCTAGAA
ATAGCAAGTTAAAATAAGGCTAGTCC). Each 100mer oligonucleotide was suspended at 100mM in water, mixed with equal volume and annealed in thermocycle machine (95 C, 5min;
Ramp to 4 C, 0.1 C/sec). To prepare the destination vector, the gRNA cloning vector (Addgene plasmid ID 41824) was linearized using AfIII and the vector was purified. The (10u1) gRNA
assembly reaction was carried out with 1 Ong annealed 100bp fragment, 10Ong destination backbone, 1X Gibson assembly reaction mix (New England Biolabs) at 50 C for 30min. The reaction can be processed directly for bacterial transformation to colonize individual assemblies.
EXAMPLE II
Re-Coded TALEs Design and Assembly re-TALEs were optimized at different levels to facilitate assembly, and improve expression.
re-TALE DNA sequences were first co-optimized for a human codon-usage, and low mRNA
folding energy at the 5' end (GeneGA, Bioconductor). The obtained sequence was evolved through several cycles to eliminate repeats (direct or inverted) longer than 11 bp (See Fig. 12). In each cycle, synonymous sequences for each repeat are evaluated. Those with the largest hamming distance to the evolving DNA are selected. The sequence of one of re-TALE
possessing 16.5 monomers as follows CTAACCCCTGAACAGGTAGTCGCTATAGCTTCAAATATCGGGGGCAAGCAAGCACTTG
AGACCGTTCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCGGAGCA
AGTCGTCGCGATCGCGAGCAACGGCGGGGGGAAGCAGGCGCTGGAAACTGTTCAGAG
ACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAGGTTGTCGCAATA
GCAAGTAATATAGGCGGTAAGCAAGCCCTAGAGACTGTGCAACGCCTGCTCCCCGTGC
TGTGTCAGGCTCACGGTCTGACACCTGAACAAGTTGTCGCGATAGCCAGTCACGACGG
GGGAAAACAAGCTCTAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAAGCACAT
GGGTTAACACCCGAACAAGTAGTAGCGATAGCGTCAAATAACGGGGGTAAACAGGCT
TTGGAGACGGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTACGCCAG
AACAGGTGGTTGCAATTGCCTCCAACATCGGCGGGAAACAAGCGTTGGAAACTGTGC
AGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAGGTTGTGGC
CATCGCTAGCCACGACGGAGGGAAGCAGGCTCTTGAAACCGTACAGCGACTTCTCCCA

GTTTTGTGCCAAGCTCACGGGCTAACCCCCGAGCAAGTAGTTGCCATAGCAAGCAACG
GAGGAGGAAAACAGGCATTAGAAACAGTTCAGCGCTTGCTCCCGGTACTCTGTCAGG
CACACGGTCTAACTCCGGAACAGGTCGTAGCCATTGCTTCCCATGATGGCGGCAAACA
GGCGCTAGAGACAGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGCCTCACC
CCGGAGCAGGTCGTTGCCATCGCCAGTAATATCGGCGGAAAGCAAGCTCTCGAAACA
GTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCAGGTAG
TGGCAATCGCATCT CAC GATGGAGGTAAACAAGCACTCGAGACTGT CCAAAGATTGTT
ACCCGTACTATGCCAAGCGCATGGTTTAACCCCAGAGCAAGTTGTGGCTATTGCATCT
AACGGCGGTGGCAAACAAGCCTTGGAGACAGTGCAACGATTACTGCCTGTCTTATGTC
AGGC CCATGGC CTTACTC CTGAGCAAGTC GTAGCTATC GC CAG CAACATAGGTGG GAA
ACAGGCCCTGGAAACCGTACAACGTCTCCTCCCAGTACTTTGTCAAGCACACGGGTTG
ACACCGGAACAAGTGGTGGCGATTGCGTCCAACGGCGGAGGCAAGCAGGCACTGGAG
ACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAGAGCAGG
TGGTAGCAATAGCGTCGAACATCGGTGGTAAGCAAGCGCTTGAAACGGTCCAGCGTCT
TCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACCAGAACAAGTGGTTGCTATTGCT
AGTAACAACGGTGGAAAGCAGGCCCTCGAGACGGTGCAGAGGTTACTTCCCGTCCTCT
GTCAAGCGCACGGCCTCACTCCAGAGCAAGTGGTTGCGATCGCTTCAAACAATGGTGG
AAGACCTGCCCTGGAA
According to certain aspects, TALEs may be used having at least 80% sequence identity, at least 85% sequence identity, at least 90% sequence identity, at least 95%
sequence identity, at least 98% sequence identity, or at least 99% sequence identity to the above sequence. One of skill will readily understand where the above sequence may vary while still maintaining the DNA
binding activity of the TALE.
re-TALE dimer blocks encoding two RVDs (see Fig. 6A) were generated by two rounds of PCR under standard Kapa HIFI (KPAP) PCR conditions, in which the first round of PCR
introduced the RVD coding sequence and the second round of PCR generated the entire dimer blocks with 36bp overlaps with the adjacent blocks. PCR products were purified using QIAquick 96 PCR Purification Kit (QIAGEN) and the concentrations were measured by Nano-drop. The primer and template sequences are listed in Table 1 and Table 2 below.
Table 1. re-TALE blocks sequences CGCAATGCGCTCACGGGAGCACCCCTCAACCTAACCCCTGAACAGGTA
GTCGCTATAGCTTCANThThThGGGGGCAAGCAAGCACTTGAGACCGT
block0 TCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCGGA
GCAAGTCGTCGCGATCGCGAGCNNNNNNGGGGGGAAGCAGGCGCTGG

AAACTGTTCAGAGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTC
AGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAG
GTTGTCGCAATAGCAAGTN
GGCGGTAAGCAAGCCCTAGAGAC
TGTGCAACGCCTGCTCCCCGTGCTGTGTCAGGCTCACGGTCTGACACC
blockl TGAACAAGTTGTCGCGATAGCCAGTNNGGGGGAAAACAAGCTC
TAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAAGCACATGGGT
TA
TGCGCTCACGGGAGCACCCCTCAACCTCACCCCCGAACAGGTTGTCGC
AATAGCAAGTNNNNNNGGCGGTAAGCAAGCCCTAGAGACTGTGCAAC
block 1! GCCTGCTCCCCGTGCTGTGTCAGGCTCACGGTCTGACACCTGAACAAG
TTGTCGCGATAGCCAGTNThThThGGGGGAAAACAAGCTCTAGAAACG
GTTCAAAGGTTGTTGCCCGTTCTGTGCCAAGCACATGGGTTA
AGGTTGTTGCCCGTTCTGTGCCAAGCACATGGGTTAACACCCGAACAA
GTAGTAGCGATAGCGTCANThGGGGGTAAACAGGCTTTGGAGAC
GGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTACGCC
block2 AGAACAGGTGGTTGCAATTGCCTCC
NGGCGGGAAACAAGCGT
TGGAAACTGTGCAGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTT
GACGCCT
AGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAG
GTTGTGGCCATCGCTAGCNNQGAGGGAAGCAGGCTCTTGAAAC
CGTACAGCGACTTCTCCCAGTTTTGTGCCAAGCTCACGGGCTAACCCC
block3 CGAGCAAGTAGTTGCCATAGCAAGCNThThThGGAGGAAAACAGGCAT
TAGAAACAGTTCAGCGCTTGCTCCCGGTACTCTGTCAGGCACACGGTC
TA
CGCTTGCTCCCGGTACTCTGTCAGGCACACGGTCTAACTCCGGAACAG
GTCGTAGCCATTGCTTCCNThGGCGGCAAACAGGCGCTAGAGAC
CGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGCCTCACCCC
block4 GGAGCAGGTCGTTGCCATCGCCAGTNNNNNNGGCGGAAAGCAAGCTC
TCGAAACAGTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGAC
TG
CGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCAG
block5 GTAGTGGCAATCGCATCTNNGGAGGTAAACAAGCACTCGAGAC
TGTCCAAAGATTGTTACCCGTACTATGCCAAGCGCATGGTTTAACCCC

AGAGCAAGTTGTGGCTATTGCATCTNNNNNNGGTGGCAAACAAGCCTT
GGAGACCGTGCAACGATTACTGCCTGTCTTATGTCAGGCCCATGGCCT
T
CGATTACTGCCTGTCTTATGTCAGGCCCATGGCCTTACTCCTGAGCAGG
TGGTCGCTATCGCCAGC
NGGGGGCAAGCAAGCACTGGAAACA
GTCCAGCGTTTGCTTCCAGTACTTTGTCAGGCGCATGGATTGACACCG
block6 GAACAAGTGGTGGCTATAGCCTCANNNNNNGGAGGAAAGCAGGCGCT
GGAAACCGTCCAACGTCTTTTACCGGTGCTTTGCCAGGCGCACGGGCT
C
CGATTACTGCCTGTCTTATGTCAGGCCCATGGCCTTACTCCTGAGCAAG
TCGTAGCTATCGCCAGC
NGGTGGGAAACAGGCCCTGGAAACC
GTACAACGTCTCCTCCCAGTACTTTGTCAAGCACACGGGTTGACACCG
block6' GAACAAGTGGTGGCGATTGCGTCC
NGGAGGCAAGCAGGCACT
GGAGACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCT
C
CGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAGAGCAG
GTGGTAGCAATAGCGTCGNThGGTGGTAAGCAAGCGCTTGAAAC
GGTCCAGCGTCTTCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACC
block7 AGAACAAGTGGTTGCTATTGCTAGTNThThThGGTGGAAAGCAGGCCC
TCGAGACGGTGCAGAGGTTACTTCCCGTCCTCTGTCAAGCGCACGGCC
TC
Table 2. re-TALE blocks primer sequences CGCAATGCGCTCACGGGAGCACCCCTCAACctAACCCCTGAACAGGT*
block0-F A*G
GAGACCATGCGCCTGACAAAGTACAGGCAGCAGTCTCTGAACAG*T*
block0-R T
block 1 '-F TGGCGCAATGCGCTCACGGGAGCACCCCTCA*A*C
AGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACA*
block 1 -F G*G
block 1 -R/block 1 '-R TAACCCATGTGCTTGGCACAGAACGGGCAACAACCTTTGAACCG*T*T
block2-F AGGTTGTTGCCCGTTCTGTGCCAAGCACATGGGTTAACACCCgaac*a*a AGGCGTCAAGCCGTGGGCTTGACACAAAACAGGAAGGAGTCTCTGCA
blcok2-R CAG*T*t block3-F AGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTG*A*G
block3-R TAGAC CGTGTGC CT GACAGAGTACC GGGAGCAAGC GCT* G*A
block4-F CGCTTGCTCCCGGTACTCTGTCAGGCACACGGTCTAA*C*T
block4-R CAGTCCATGAGCTTGACATAGGACTGGCAACAGCCGTT*G*T
block5-F CGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGA*C*G
block5-R AAGGCCATGGGCCTGACATAAGACAGGCAGTAATCGTT*G*C
block6-F CGATTACTGCCTGTCTTATGTCAGGCCCATGGCCTTA*C*T
block6-R GAGCCCGTGCGCCTGGCAAAGCACCGGTAAAAGACGTTGGA*C*G
CGATTACTGCCTGTCTTATGTCAGGCCCATGGCCTTACTCCTGAGCAA
block6'-F *G*T
block6'-R GAGCCCATGAGCCTGGCAAAGAACCGGAAGAAGCCGTT*G*G
CGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAGAGCAG
block7-F G*T*G
blcok7-R GAGGCCGTGCGCTTGACAGAGGACGGGAAGTAACCTCT*G*C
re-TALENs and re-TALE-TF destination vectors were constructed by modifying the TALE-TF and TALEN cloning backbones (see reference 24 hereby incorporated by reference in its entirety). The 0.5 RVD regions on the vectors were re-coded and SapI cutting site was incorporated at the designated re-TALE cloning site. The sequences of re-TALENs and re-TALE-TF backbones are provided in Fig. 20. Plasmids can be pre-treated with SapI (New England Biolabs) with manufacturer recommended conditions and purified with QIAquick PCR
purification kit (QIAGEN).
A (10u1) one-pot TASA assembly reaction was carried out with 200ng of each block, 500ng destination backbone, 1X TASA enzyme mixture (2U SapI, 100U Ampligase (Epicentre), 10mU T5 exonuclease (Epicentre), 2.5U Phusion DNA polymerase (New England Biolabs)) and lx isothermal assembly reaction buffer as described before (see reference 25 hereby incorporated by reference in its entirety) (5% PEG-8000, 100 mM Tris-HC1 pH 7.5, 10 mM
MgC12, 10 mM
DTT, 0.2 mM each of the four dNTPs and 1 mM NAD). Incubations were performed at 37 C for 5min and 50 C for 30 min. TASA assembly reaction can be processed directly for bacterial transformation to colonize individual assemblies. The efficiency of obtaining full length construct is ¨20% with this approach. Alternatively, >90% efficiency can be achieved by a three-step assembly. First, lOul re-TALE assembly reactions are performed with 200ng of each block, 1X re-TALE enzyme mixture (100U Ampligase, 12.5mU T5 exonuclease, 2.5U Phusion DNA
polymerase) and lx isothermal assembly buffer at 50 C for 30min, followed by standardized Kapa HIFI PCR reaction, agarose gel electrophoresis, and QIAquick Gel extraction (Qiagen) to enrich the full length re-TALEs. 200ng re-TALE amplicons can then be mixed with 500ng Sap 1-pre-treated destination backbone, 1X re-TALE assembly mixture and lx isothermal assembly reaction buffer and incubated at 50 C for 30 min. The re-TALE final assembly reaction can be processed directly for bacterial transformation to colonize individual assemblies. One of skill in the art will readily be able to select endonucleases, exonucleases, polymerases and ligases from among those known to practice the methods described herein. For example, type Hs endonucleases can be used, such as: Fok 1, Bts I, Ear I, Sap I. Exonucleases which are titralable can be used, such as lamda exonuclease, T5 exonuclease and Exonuclease III. Non-hotstart polymerases can be used, such as phusion DNA polymerase, Taxi DNA polymerase and VentR DNA polymerase.
Thermostable ligases can be used in this reaction, such as Ampligase, pfu DNA ligase, Taxi DNA ligase. In addition, different reaction conditions can be used to activate such endonucleases, exonucleases, polymerases and ligases depending on the particular species used.
EXAMPLE III
Cell Line and Cell Culture PGP1 iPS cells were maintained on Matrigel (BD Biosciences)-coated plates in mTeSR1 (Stemcell Technologies). Cultures were passaged every 5-7 days with TrypLE
Express (Invitrogen). 293T and 293FT cells were grown and maintained in Dulbecco's modified Eagle's medium (DMEM, Invitrogen) high glucose supplemented with 10% fetal bovine serum (FBS, Invitrogen), penicillin/streptomycin (pen/strep, Invitrogen), and non-essential amino acids (NEAA, Invitrogen). K562 cells were grown and maintained in RPMI (Invitrogen) supplemented with 10%
fetal bovine serum (FBS, Invitrogen 15%) and penicillin/streptomycin (pen/strep, Invitrogen). All cells were maintained at 37 C and 5% CO2 in a humidified incubator.
A stable 293T cell line for detecting HDR efficiency was established as described in reference 26 hereby incorporated by reference in its entirety. Specifically, the reporter cell lines bear genomically integrated GFP coding sequences disrupted by the insertion of a stop codon and a 68bp genomic fragment derived from the AAVS1 locus.
EXAMPLE IV
Test of re-TALENs Activity 293T reporter cells were seeded at densities of 2 x 105 cells per well in 24-well plate and transfected them with 1 jig of each re-TALENs plasmid and 2lig DNA donor plasmid using Lipofectamine 2000 following the manufacturer's protocols. Cells were harvested using TrypLE
Express (Invitrogen) ¨18 h after transfection and resuspended in 200 1 of media for flow cytometry analysis using an LSRFortessa cell analyzer (BD Biosciences). The flow cytometry data were analyzed using FlowJo (FlowJo). At least 25,000 events were analyzed for each transfection sample. For endogenous AAVS1 locus targeting experiment in 293T, the transfection procedures were identical as described above and puromycin selection was conducted with drug concentration at 3[ig/m1 1 week after transfection.
EXAMPLE V
Functional Lentivirus Generation Assessment The lentiviral vectors were created by standard PCR and cloning techniques.
The lentiviral plasmids were transfected by Lipofectamine 2000 with Lentiviral Packaging Mix (Invitrogen) into cultured 293FT cells (Invitrogen) to produce lentivirus. Supernatant was collected 48 and 72h post-transfection, sterile filtered, and 100u1 filtered supernatant was added to 5 x 105 fresh 293T cells with polybrene. Lentivirus titration was calculated based on the following formula: virus titration =
(percentage of GFP+ 293T cell * initial cell numbers under transduction) /
(the volume of original virus collecting supernatant used in the transduction experiment). To test the functionality of lentivirus, 3 days after transduction, lentivirus transduced 293T cells were transfected with 30 ng plasmids carrying mCherry reporter and 50Ong pUC19 plasmids using Lipofectamine 2000 (Invitrogen). Cell images were analyzed using Axio Observer Z.1 (Zeiss) 18 hours after transfection and harvested using TrypLE Express (Invitrogen) and resuspended in 200 p1 of media for flow cytometry analysis using a LSRFortessa cell analyzer (BD
Biosciences). The flow cytometry data were analyzed using BD FACSDiva (BD Biosciences).
EXAMPLE VI
Test of re-TALENs and Cas9-gRNA genome editing efficiency PGP1 iPSCs were cultured in Rho kinase (ROCK) inhibitor Y-27632 (Calbiochem) 2h before nucleofection. Transfections were done using P3 Primary Cell 4D-Nucleofector X Kit (Lonza). Specifically, cells were harvested using TrypLE Express (Invitrogen) and 2x106 cells were resuspended in 20 p1 nucleofection mixture containing 16.4 pl P3 Nucleofector solution, 3.6 pl supplement, lpg of each re-TALENs plasmid or lug Cas9 and lug gRNA
construct, 2111 of 100 [iM ssODN. Subsequently, the mixtures were transferred to 20 1 Nucleocuvette strips and nucleofection was conducted using CB150 program. Cells were plated on Matrigel-coated plates in mTeSR1 medium supplemented with ROCK inhibitor for the first 24 hrs. For endogenous AAVS1 locus targeting experiment with dsDNA donor, the same procedure was followed except 2 [ig dsDNA donor was used and the mTeSR1 media was supplemented with puromycin at the concentration of 0.5ug/mL 1 week after transfection.
The information of reTALENs, gRNA and ssODNs used in this example are listed in Table 3 and Table 4 below.

Table 3. Information of re-TALEN pairs/Cas9-gRNA targeting CCR5 re- re-TAL TAL
ENs ENs gRA
pair N
target target #
ing pair ing targe . re-TALEN-L re-TALEN-R gRNA targeting she segue ting targeting sequence targeting sequence sequence . (start) nce site target start ing positi /chr3:
site on (end) /chr3:

Table 4. ssODN design for studying ssODN-mediated genome editing Dist 90- CTACTGTCATTCAGGGCAATACCCAGACGAGAAAGCTGAGGGTAT
ance *1 AACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
bet wee *2 ACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
n Fi the 90- CTACTGTCATTCAGCCCAATACCCAGACGAGAAAAGTGAGGGTATA
see *3 ACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
gu ndar re 3b mut 0 ACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
atio n *4 ACAGGTTTGTAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
and B *5 ACAGGTTTCAAGCTTGGCTCTCTGACTACAGAGGCCACTGGCTT

*6 ACAGGTTTCAAGCTTGGCAGTCTGACTAGTGAGGCCACTGGCTT

CACTTTATATTTCCCTGCTTAAACAGTCCCCCGAGGGTGGGTGCGG
bp 9 ¨ AAAAGGCTCTACACTTGTTATCATTCCCTCTCCACCACAGGCAT
OM

TTTGTATTTGGGTTTTTTTAAAACCTCCACTCTACAGTTAAGAATTC
bp 9 ¨ TAAGGCACAGAGCTTCAATAATTTGGTCAGAGCCAAGTAGCAG
OM

GGAGGTTAAACCCAGCAGCATGACTGCAGTTCTTAATCAATGCCCC
bp 9 TTGAATTGCACATATGGGATGAACTAGAACATTTTCTCGATGAT
OM

CTCGATGATTCGCTGTCCTTGTTATGATTATGTTACTGAGCTCTACT
bp 9 ¨ GTAGCACAGACATATGTCCCTATATGGGGCGGGGGTGGGGGTG
dist OM
ance L290 GGTGTCTTGATCGCTGGGCTATTTCTATACTGTTCTGGCTTTTCGGA
bet bp 9 ¨ AGCAGTCATTTCTTTCTATTCTCCAAGCACCAGCAATTAGCTT
wee OM
Fi n L200 gu GCTTCTAGTTTGCTGAAACTAATCTGCTATAGACAGAGACTCCGAC
ss0 bp 9 re ¨ GAACCAATTTTATTAGGATTTGATCAAATAAACTCTCTCTGACA
DN OM
3c and L114 GAAAGAGTAACTAAGAGTTTGATGTTTACTGAGTGCATAGTATGCA
the bp 9 ¨ CTAGATGCTGGCCGTGGATGCCTCATAGAATCCTCCCAACAACT
DS OM
B L45b GCTAGATGCTGGCCGTGGATGCCTCATAGAATCCTCCCAACAACCG
p90 ¨ ATGAAATGACTACTGTCATTCAGCCCAATACCCAGACGAGAAAG
M
R40b ACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTTTA
p90 ¨ CCCCTGGGTTAGTCTGCCTCTGTAGGATTGGGGGCACGTAATTT
M

TTAGTCTGCCTCTGTAGGATTGGGGGCACGTAATTTTGCTGTTTAAG
bp 9 ¨ GTCTCATTTGCCTTCTTAGAGATCACAAGCCAAAGCTTTTTAT
OM

GGAAGCCCAGAGGGCATCTTGTGGCTCGGGAGTAGCTCTCTGCTAC
bp 9 CTTCTCAGCTCTGCTGACAATACTTGAGATTTTCAGATGTCACC
OM

TCAGCTCTGCTGACAATACTTGAGATTTTCAGATGTCACCAACCAG
bp 9 CAAGAGAGCTTGATATGACTGTATATAGTATAGTCATAAAGAAC
OM

CATAAAGAACCTGAACTTGACCATATACTTATGTCATGTGGAAATC
bp 9 TTCTCATAGCTTCAGATAGATTATATCTGGAGTGAAGAATCCTG
OM

GTGGAAAATTTCTCATAGCTTCAGATAGATTATATCTGGAGTGAGC

¨ AATCCTGCCACCTATGTATCTGGCATAGTGTGAGTCCTCATAAA
OM

GGTTTGAAGGGCAACAAAATAGTGAACAGAGTGAAAATCCCCACC
bp 9 ¨ TAGATCCTGGGTCCAGAAAAAGATGGGAAACCTGTTTAGCTCACC
OM
Corn plem ent- GGCCACTAGGGACAAAATTGGTGAcagaaa 30me r Corn ss0 plem DN ent- CCCACAGTGGGGCCACTAGGGACAAAATTGGTGAcagaaaagccccatcc leng 50me th r and Corn one plem TCCCCTCCACCCCACAGTGGGGCCACTAGGGACAAAATTGGTGAcag Fi ntati ent-aaaagccccatccttaggcctcc gu on 70me re for r 3d Cas Corn 9- plem cifTTATCTGTCCCCTCCACCCCACAGTGGGGCCACTAGGGACAAAAT
gR ent-TGGTGAc agaaaagccccatccttaggcctectecttcctag NA 90me targ r etin Corn gttctgggtacttTTATCTGTCCCCTCCACCCCACAGTGGGGCCACTAGGGA
g plem CAAAATTGGTGAcagaaaagccccatccttaggcctectecttcctagtctcctgata ent-110m er Non-comp leme TTTCTGTCACCAATGGTGTCCCTAGTGGCC
nt-30me r Non-comp leme GGATGGGGCTTTTCTGTCACCAATGGTGTCCCTAGTGGCCCCACTG
nt- TGGG
50me r Non-comp leme GGAGGCCTAAGGATGGGGCTTTTCTGTCACCAATGGTGTCCCTAGT
nt- GGCCCCACTGTGGGGTGGAGGGGA
70me r Non-comp leme CTAGGAAGGAGGAGGCCTAAGGATGGGGCTTTTCTGTCACCAATG
nt- GTGTCCCTAGTGGCCCCACTGTGGGGTGGAGGGGACAGATAAAAG
90me r Non-comp TATCAGGAGACTAGGAAGGAGGAGGCCTAAGGATGGGGCTTTTCT
leme GTCACCAATGGTGTCCCTAGTGGCCCCACTGTGGGGTGGAGGGGAC
nt-AGATAAAAGTACCCAGAAC
110m er Fi ss0 Cas9 TTCTAGTAACCACTCAGGACAGGGGGGTTCAGCCCAAAAATTCACA
gu DN -AGAAAGTGGGGACCCATGGGAAAT
re don gRN

2c or A-for CCR
Cas 5-1 9- Cas9 gR -NA gRN CAGCAAGTCAGCAGCACAGCGTGTGTGACTCCGAGGGTGCTCCGCT
targ A- AGCCCACATTGCCCTCTGGGGGTG
etin CCR
g 5-2 CC Cas9 gRN GTCAGACTGCCAAGCTTGAAACCTGTCTTACCCTCTACTTTCTCGTC
A- TGGGTATTGGGCTGAATGACAGT
CCR

Cas9 gRN CAGAGCTGAGAAGACAGCAGAGAGCTACTCCCGAAGCACAAGATG
A- CCCTCTGGGCTTCCGTGACCTTGGC
CCR

Cas9 gRN CTGACAATACTTGAGATTTTCAGATGTCACCAACGACCAAGAGAGC
A- TTGATATGACTGTATATAGTATAG
CCR

Cas9 gRN CAGATACATAGGTGGCAGGATTCTTCACTCCAGACTTAATCTATCT
A- GAAGCTATGAGAAATTTTCCACAT
CCR

Cas9 TATATGATTGATTTGCACAGCTCATCTGGCCAGATAAGCTGAGACA
- TCCGTTCCCCTACAAGAAACTCTC

gRN
A-CCR

Cas9 gRN ATCTGGCCAGAAGAGCTGAGACATCCGTTCCCCTTGAAGAAACTCT
A- CCCCGGTAAGTAACCTCTCAGCTG
CCR

Cas9 gRN AGGCATCTCACTGGAGAGGGTTTAGTTCTCCTTAAGAGAAGATAAG
A- ATTTCAAGAGGGAAGCTAAGACTC
CCR

Cas9 gRN ATAATATAATAAAAAATGTTTCGTCTGCCACCACTAATGAATGTCA
A- TGCATTCTGGGTAGTTTAGTCTTA
CCR

Cas9 gRN TTTATAAAGTCCTAGAATGTATTTAGTTGCCCTCGTTGAATGCAAAC
A- TGTTTTATACATCAATAGGTTTT
CCR

Cas9 gRN GCTCAACCTGGCCATCTCTGACCTGTTTTTCCTTCCCACTGTCCCCT
A- TCTGGGCTCACTATGCTGCCGCC
CCR

Cas9 TTTTAAAGCAAACACAGCATGGACGACAGCCAGGCTCCTATCGATT

- GTCAGGAGGATGATGAAGAAGATT
gRN
A-CCR

Cas9 gRN GCTTGTCATGGTCATCTGCTACTCGGGAATCCTAATTACTCTGCTTC
A- GGTGTCGAAATGAGAAGAAGAGG
CCR

Cas9 gRN ATACTGCCCCCGCGAGGCCACATTGGCAAACCAGCTTGGGTGTTCA
A- ACTTCCAGACTTGGCCATGGAGAA
CCR

reTA
LEN- CTGAAGAATTTCCCATGGGTCCCCACTTTCTTGTGAATCCTTGGAGT
CCR GAACCCCCCTGTCCTGAGTGGTTACTAGAACACACCTCTGGAC
ss0 s_i DN reTA
don LEN- TGGAAGTATCTTGCCGAGGTCACACAGCAAGTCAGCAGCACAGCC
or CCR AGTGTGACTCCGAGCCTGCTCCGCTAGCCCACATTGCCCTCTGGG
for 5_2 reT reTA
AL LEN- CTACTGTCATTCAGCCCAATACCCAGACGAGAAAGCTGAGGGTATA
ENs CCR ACAGGTTTCAAGCTTGGCAGTCTGACTACAGAGGCCACTGGCTT
targ 5_3 etin reTA
g LEN- GGAAGCCCAGAGGGCATCTTGTGGCTCGGGAGTAGCTCTCTGCTAC
CC CCR CTTCTCAGCTCTGCTGACAATACTTGAGATTTTCAGATGTCACC
R5 5_4 reTA TCAGCTCTGCTGACAATACTTGAGATTTTCAGATGTCACCAACGCC
LEN- CAAGAGAGCTTGATATGACTGTATATAGTATAGTCATAAAGAAC

CCR

reTA
LEN- GTGGAAAATTTCTCATAGCTTCAGATAGATTATATCTGGAGTGAGC
CCR AATCCTGCCACCTATGTATCTGGCATAGTGTGAGTCCTCATAAA

reTA
LEN- GAAACAGCATTTCCTACTTTTATACTGTCTATATGATTGATTTGGTC
CCR AGCTCATCTGGCCAGAAGAGCTGAGACATCCGTTCCCCTACAA

reTA
LEN- TTGATTTGCACAGCTCATCTGGCCAGAAGAGCTGAGACATCCGTAT
CCR CCCTACAAGAAACTCTCCCCGGTAAGTAACCTCTCAGCTGCTTG

reTA
LEN- GGAGAGGGTTTAGTTCTCCTTAGCAGAAGATAAGATTTCAAGATGA
CCR GAGCTAAGACTCATCTCTCTGCAAATCTTTCTTTTGAGAGGTAA

reTA
LEN- TAATATAATAAAAAATGTTTCGTCTGCCACCACAGATGAATGTCGA
CCR GCATTCTGGGTAGTTTAGTCTTATAACCAGCTGTCTTGCCTAGT

reTA
LEN- TTAAAAACCTATTGATGTATAAAACAGTTTGCATTCATGGAGGGTG
CCR ACTAAATACATTCTAGGACTTTATAAAAGATCACTTTTTATTTA

reTA
LEN- GACATCTACCTGCTCAACCTGGCCATCTCTGACCTGTTTTTCCTATT
CCR TACTGTCCCCTTCTGGGCTCACTATGCTGCCGCCCAGTGGGAC

reTA
LEN- TCATCCTCCTGACAATCGATAGGTACCTGGCTGTCGTCCATGCTAC
CCR GTTTGCTTTAAAAGCCAGGACGGTCACCTTTGGGGTGGTGACAA

reTA
LEN- GGCTGGTCCTGCCGCTGCTTGTCATGGTCATCTGCTACTCGGGAGA
CCR CCTAAAAACTCTGCTTCGGTGTCGAAATGAGAAGAAGAGGCACA

reTA
LEN- GGCAAGCCTTGGGTCATACTGCCCCCGCGAGGCCACATTGGCAAGT
CCR CAGCAAGGGTGTTCAACTTCCAGACTTGGCCATGGAGAAGACAT

EXAMPLE VII
Amplicon Library Preparation of the Targeting Regions Cells were harvested 6 days after nucleofection and 0.1 [L1 prepGEM tissue protease enzyme (ZyGEM) and 1 [L1 prepGEM gold buffer (ZyGEM) were added to 8.9 ill of the 2-5 X 105 cells in the medium. lul of the reactions were then added to 9 I.E1 of PCR mix containing Sul 2X
KAPA Hifi Hotstart Readymix (KAPA Biosystems) and 100nM corresponding amplification primer pairs. Reactions were incubated at 95 C for 5 min followed by 15 cycles of 98 C, 20 s;
65 C, 20 s and 72 C, 20 s. To add the Illumina sequence adaptor used, 5 I.E1 reaction products were then added to 20 I.E1 of PCR mix containing 12.5 I.E1 2X KAPA HIFI Hotstart Readymix (KAPA
Biosystems) and 200 nM primers carrying Illumina sequence adaptors. Reactions were incubated at 95 C for 5min followed by 25 cycles of 98 C, 20s; 65 C, 20s and 72 C, 20s. PCR
products were purified by QIAquick PCR purification kit, mixed at roughly the same concentration, and sequenced with MiSeq Personal Sequencer. The PCR primers are listed in Table 5 below.
Table 5. CCR5 targeting site PCR primer sequences #
targeting name primer sequence in CCR5 ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATTTT
site 1-Fl GCAGTGTGCGTTACTCC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGTTT
sitel -F2 GCAGTGTGCGTTACTCC

sitel -F3 GCAGTGTGCGTTACTCC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGGTCATTT
sitel -F4 GCAGTGTGCGTTACTCC
site 1 -R CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTCCAAGCAA

CTAAGTCACAGCA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATATG
Site2-F 1 AGGAAATGGAAGCTTG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGAT
Site2-F2 GAGGAAATGGAAGCTTG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGCCTAAAT

Site2-F 3 GAGGAAATGGAAGCTTG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGGTCAATG
Site2-F4 AGGAAATGGAAGCTTG
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTCATTAGGG
Site2-R TATTGGAGGA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATAAT
site3 -Fl CCTCCCAACAACTCAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGAA
site3 -F2 TCCTCCCAACAACTCAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGCCTAAAA

site3 -F3 TCCTCCCAACAACTCAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGGTCAAAT
site3 -F4 CCTCCCAACAACTCAT
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTCCCAATCCT
site3_R ACAGAGGCAG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATAA
site4-F 1 GCCAAAGCTTTTTATTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGAA
site4-F2 GCCAAAGCTTTTTATTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGCCTAAAA

site4-F3 GCCAAAGCTTTTTATTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGGTCAAA
site4-F4 GCCAAAGCTTTTTATTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTAAGCCAAA
site4_R GCTTTTTATTCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATATC
site5-F 1 TTGTGGCTCGGGAGTAG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGATC
site5 -F2 TTGTGGCTCGGGAGTAG

CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTTGGCAGGA
site5-R TTCTTCACTCCA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATCTA
site6-F 1 TTTTGTTGCCCTTCAAA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGCTA

site6-F2 TTTTGTTGCCCTTCAAA
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAACCTGAA
site6-R CTTGACCATATACT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATCA
site7-F 1 GCTGAGAGGTTACTTACC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGCA

site7-F2 GCTGAGAGGTTACTTACC
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAATGATTTA
site7-R ACTCCACCCTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATACT
site8-F 1 CCACCCTCCTTCAAAAGA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGACT

site8-F2 CCACCCTCCTTCAAAAGA
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTTGGTGTTTG
site 8-R CCAAATGTCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATGG
site9_F 1 GCACATATTCAGAAGGCA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGGG

site9_F2 GCACATATTCAGAAGGCA
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAGTGAAAG
site9_R ACTTTAAAGGGAGCA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATCAC
site 1 O-F 1 AATTAAGAGTTGTCATA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGCA
site 1 0-F2 CAATTAAGAGTTGTCATA
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTCTCAGCTA
site 1 O-R GAGCAGCTGAAC
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTGACACTTG
1 1 site 1 1-Fl ATAATCCATC
site 11 -F2 ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGTCA

ATGTAGACATCTATGTAG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATTCA
sitell-R ATGTAGACATCTATGTAG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATACT
site12-F1 GCAAAAGGCTGAAGAGC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGACT
site12-F2 GCAAAAGGCTGAAGAGC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGCCTAAACT

site12-F3 GCAAAAGGCTGAAGAGC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGGTCAACT
site12-F4 GCAAAAGGCTGAAGAGC
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTGCCTATAA
site12-R AATAGAGCCCTGTCAA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATCTC
sitel3 -F1 TATTTTATAGGCTTCTTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGCTC

site 13 -F2 TATTTTATAGGCTTCTTC
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAGCCACCA
site13-R CCCAAGTGATC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACATCGTTC
site14-F1 CAGACATTAAAGATAGTC
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATTTC

site14-F2 CAGACATTAAAGATAGTC
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAATCATGA
site14-R TGGTGAAGATAAG
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGTGATCCG
site15-F1 GCAGAGACAAACATTAAA
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCCGGCAGA
site15-F2 GACAAACATTAAA
CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTAGCTAGGA
site15-R AGCCATGGCAAG
AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACA
illumina PE-PCR-F cgac*g*c adaptor PE-PCR-R CAAGCAGAAGACGGCATACGAGATCGGTCTCGGCATTCCT

GCTGAACc*g*c \\. sYr:,kRigkµ=
ACACTCTTTCCCTACACGACGCTCTTCCGATCTAGTGCATA
site3-M-F GTATGTGCTAGATGCTG

GTGACTGGAGTTCAGACGTGTGCTCTTCCGATCTTGATCTC
site3-M-R TAAGAAGGCAAATGAGAC

illumina Index-PCR TGGAGTTCAGACGTGTGCTCTTCCGATCT
adaptor universal- AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACA
PCR CGACGCTCTTCCGATCT
*index-PCR primers are purchased from epicentre (ScriptSeqTM Index PCR
Primers) EXAMPLE VIII
Genome Editing Assessment System (GEAS) Next generation sequencing has been utilized to detect rare genomic alterations. See references 27-30 hereby incorporated by reference in their entireties. To enable wide use of this approach to quickly assess HDR and NHEJ efficiency in hiPSCS, software was created, referred to as a "pipeline", to analyze the genome engineering data. This pipeline is integrated in one single Unix module, which uses different tools such as R, BLAT, and FASTX Toolkit.
Barcode splitting: Groups of samples were pooled together and sequenced using MiSeq 150bp paired end (PE150) (IIlumina Next Gen Sequencing), and later separated based on DNA
barcodes using FASTX Toolkit.
Quality filtering: Nucleotides with lower sequence quality (phred score<20) were trimmed.
After trimming, reads shorter than 80 nucleotides were discarded.
Mapping: BLAT was used to map the paired reads independently to the reference genome and .ps1 files were generated as output.
Indel calling: Indels were defined as the full length reads containing 2 blocks of matches in the alignment. Only reads following this pattern in both paired end reads were considered. As a quality control, the indel reads were required to possess minimal 7Ont matching with the reference genome and both blocks to be at least 20 nt long. Size and position of indels were calculated by the positions of each block to the reference genome. Non-homologous end joining (NHEJ) has been estimated as the percentage of reads containing indels (see equation 1 below).
The majority of NHEJ events have been detected at the targeting site vicinity.
Homology directed recombination (HDR) efficiency: Pattern matching (grep) within a 12bp window centering over DSB was used to count specific signatures corresponding to reads containing the reference sequence, modifications of the reference sequence (2bp intended mismatches), and reads containing only lbp mutation within the 2bp intended mismatches (see equation 1 below).
Equation 1. Estimation of NHEJ and HDR
A= reads identical to the reference: XXXXXABXXXXX
B= reads containing 2bp mismatch programed by ssODN: XXXXXabXXXXX
C= reads containing only 1 bp mutation in the target site: such as XXXXXaBXXXXX or XXXXXAbXXXXX
D = reads containing indels as described above Nigj = OEM X ________ A + C + Et) eNdvrizy =
A + C
EXAMPLE IX
Genotype Screening of Colonized hiPSCs Human iPS cells on feeder-free cultures were pre-treated with mTesr-1 media supplemented with SMC4 (5 uM thiazovivin,1 uM CHIR99021, 0.4 uM PD0325901, 2 uM
SB431542) (see reference 23 hereby incorporated by reference in its entirety for at least 2 hrs prior to FACS sorting. Cultures were dissociated using Accutase (Millipore) and resuspended in mTesr-1 media supplemented with SMC4 and the viability dye ToPro-3 (Invitrogen) at concentration of 1-2 X107 /mL. Live hiPS cells were single-cell sorted using a BD FACSAria II
SORP UV (BD
Biosciences) with 100um nozzle under sterile conditions into 96-well plates coated with irradiated CF-1 mouse embryonic fibroblasts (Global Stem). Each well contained hES cell medium (see reference 31 hereby incorporated by reference in its entirety) with 100 ng /
ml recombinant human basic Fibroblast Growth Factor (bFGF) (Millipore) supplemented with SMC4 and 5 ug / ml fibronectin (Sigma). After sorting, plates were centrifuged at 70 x g for 3 min. Colony formation was seen 4 days post sorting, and the culture media was replaced with hES cell medium with SMC4. SMC4 can be removed from hES cell medium 8 days after sorting.
A few thousand cells were harvested 8 days after Fluorescence-activated cell sorting (FACS) and 0.1u1 prepGEM tissue protease enzyme (ZyGEM) and lul prepGEM gold buffer (ZyGEM) were added to 8.9 1 of cells in the medium. The reactions were then added to 40 I of PCR mix containing 35.5m1 platinum 1.1X Supermix (Invitrogen), 250nM of each dNTP and 400nM primers. Reactions were incubated at 95 C for 3min followed by 30 cycles of 95 C, 20s;
65 C, 30s and 72 C, 20s. Products were Sanger sequenced using either one of the PCR primers in Table 5 and sequences were analyzed using DNASTAR (DNASTAR).

EXAMPLE X
Immunostaining and Teratoma Assays of hiPSCs Cells were incubated in the KnockOut DMEM/F-12 medium at 37 C for 60 minutes using the following antibody: Anti-SSEA-4 PE (Millipore) (1: 500 diluted); Tra-1-60 (BD Pharmingen) (1:100 diluted). After the incubation, cells were washed three times with KnockOut DMEM/F-12 and imaged on the Axio Observer Z.1 (ZIESS).
To conduct teratoma formation analysis, human iPSCs were harvested using collagenase type IV (Invitrogen) and the cells were resuspended into 200 Ill of Matrigel and injected intramuscularly into the hind limbs of Rag2gamma knockout mice. Teratomas were isolated and fixed in formalin between 4 - 8 weeks after the injection. The teratomas were subsequently analyzed by hematoxylin and eosin staining.
EXAMPLE XI
Targeting Genomic Loci in Human Somatic Cells and Human Stem Cells Using reTALENS
According to certain aspects, TALEs known to those of skill in the art are modified or re-coded to eliminate repeat sequences. Such TALEs suitable for modification and use in the genome editing methods in viral delivery vehicles and in various cell lines and organisms described herein are disclosed in references 2, 7-12 hereby incorporated by reference herein in their entireties.
Several strategies have been developed to assemble the repetitive TALE RVD
array sequences (see references 14 and 32-34 hereby incorporated by reference herein in their entireties. However, once assembled, the TALE sequence repeats remain unstable, which limits the wide utility of this tool, especially for viral gene delivery vehicles (see references 13 and 35 hereby incorporated by reference herein in their entireties. Accordingly, one aspect of the present disclosure is directed to TALEs lacking repeats, such as completely lacking repeats. Such a re-coded TALE is advantageous because it enables faster and simpler synthesis of extended TALE
RVD arrays.
To eliminate repeats, the nucleotide sequences of TALE RVD arrays were computationally evolved to minimize the number of sequence repeats while maintaining the amino acid composition. Re-coded TALE (Re-TALEs) encoding 16 tandem RVD DNA recognition monomers, plus the final half RVD repeat, are devoid of any 12bp repeats (see Fig. 5a). Notably, this level of recoding is sufficient to allow PCR amplification of any specific monomer or sub-section from a full-length re-TALE construct (see Fig. 5b). The improved design of re-TALEs may be synthesized using standard DNA synthesis technology (see reference 36 hereby incorporated by reference in its entirety without incurring the additional costs or procedures associated with repeat-heavy sequences. Furthermore, the recoded sequence design allows efficient assembly of re-TALE
constructs using a modified isothermal assembly reaction as described in the methods herein and with reference to Fig. 6.

Genome editing NGS data was statistically analyzed as follows. For HDR
specificity analysis, an exact binomial test was used to compute the probabilities of observing various numbers of sequence reads containing the 2bp mismatch. Based on the sequencing results of 10bp windows before and after the targeting site, the maximum base change rates of the two windows (P1 and P2) were estimated. Using the null hypothesis that the changes of each of the two target bp were independent, the expected probability of observing 2bp mismatch at the targeting site by chance as the product of these two probabilities (P1 *P2) was computed. Given a dataset containing N numbers of total reads and n number of HDR reads, we calculated the p-value of the observed HDR efficiency was calculated. For HDR sensitivity analysis, the ssODN DNA
donors contained a 2bp mismatch against the targeting genome, which made likely the co-presence of the base changes in the two target bp if the ssODN was incorporated into the targeting genome. Other non-intended observed sequence changes would not likely change at the same time.
Accordingly, non-intended changes were much less interdependent. Based on these assumptions, mutual information (MI) was used to measure the mutual dependence of simultaneous two base pair changes in all other pairs of positions, and the HDR detection limit was estimated as the smallest HDR where MI
of the targeting 2bp site is higher than MI of all the other position pairs.
For a given experiment, HDR reads with intended 2bp mismatch from the original fastq file were identified and a set of fastq files with diluted HDR efficiencies were simulated by systematically removing different numbers of HDR reads from the original data set. Mutual information (MI) was computed between all pairs of positions within a 20bp window centered on the targeting site. In these calculations, the mutual information of the base composition between any two positions is computed. Unlike the HDR specificity measure described above, this measure does not assess the tendency of position pairs to change to any particular pairs of target bases, only their tendency to change at the same time. (see Fig. 8A). Table 6 shows HDR and NHEJ efficiency of re-TALEN/ssODN
targeting CCR5 and NHEL efficiency of Cas9-gRNA. We coded our analysis in R and MI was computed using the package infotheo.
Table 6 HDR detection HDR NHEJ
limit based on NHEJ HDR
targeting cell type (reTALEN) (reTALE) Information (Cas9-gRNA) (Cas9-gRNA) site (/o) (%) analysis 1 PGP1-iPS 0.06% 0.80% 0.04% 0.58%
0.38%
2 PGP1-iPS 0.48% 0.26% 0.01% 16.02%
3.71%

3 PGP1-iPS 1.71% 0.07% 0.03% 3.44%
3.20%
4 PGP1-iPS 5 ' 002`',g ' '10_0'!;h .ai)2'94.0 ' ' :i.. i U:14`C
PGP1-iPS 0.80% 0.04% 0.00% 3.70% 0.39%
6 PGP1-iPS 0.20% 0.73% 0.00% 1.12%
0.49%
7 PGP1-iPS !II''''''''''' 4J()10:.I!I''''''''''''''''UTNII!!
U0 1 NW '''''''''1! rZOgq" 11''''''''''''''''''''' IL VC
8 PGP1-iPS 0.03% 0.00% 0.00% 1.85%
0.03%
9 PGP1-iPS 1.60% 0.06% 0.00% 0.50%
0.13%
PGP1-iPS 0.68% 1.25% 0.01% 8.77% 1.32%
11 PGP1-iPS 0.06% 0.27% 0.00% 0.62%
0.44%
12 PGP1-iPS 1.60% 0.03% 0.04% 0.18%
0.99%
13 PGP1-iPS 0.00% 1.47% 0.00% 0.65%
0.02%
14 PGP1-iPS 0.47% 0.13% 0.02% 2.50%
0.31%
PGP1-iPS 0.8 0.14 0.08% 1.50 1.10%
* The group where HDR detection limit exceeds the real HDR detected Correlations between genome editing efficiency and epigenetic state were addressed as 5 follows. Pearson correlation coefficients were computed to study possible associations between epigenetic parameters (DNase I HS or nucleosome occupancy) and genome engineering efficiencies (HDR, NHEJ). Dataset of DNAasel Hypersensitivity was downloaded from UCSC
genome browser. hiPSCs DNase I HS:
/gbdb/hg19/bbi/wgEncodeOpenChromDnaseIpsnihi7Sig.bigWig 10 To compute P-values, the observed correlation was compared to a simulated distribution which was built by randomizing the position of the epigenetic parameter (N=100000). Observed correlations higher than the 95th percentile, or lower than the 5th percentile of the simulated distribution were considered as potential associations.
The function of reTALEN in comparison with the corresponding non-recoded TALEN
in
15 human cells was determined. A HEK 293 cell line containing a GFP
reporter cassette carrying a frame-shifting insertion was used as described in reference 37 hereby incorporated by reference in its entirety. See also Fig. la. Delivery of TALENs or reTALENs targeting the insertion sequence, together with a promoter-less GFP donor construct, leads to DSB-induced HDR
repair of the GFP
cassette, so that GFP repair efficiency can be used to evaluate the nuclease cutting efficiency. See reference 38 hereby incorporated by reference in its entirety. reTALENs induced GFP repair in 1.4% of the transfected cells, similar to that achieved by TALENs (1.2%) (see Fig. lb). The activity of reTALENs at the AAVS1 locus in PGP1 hiPSCs was tested (see Fig.
lc) and successfully recovered cell clones containing specific insertions (see Fig.
ld,e), confirming that reTALENs are active in both somatic and pluripotent human cells.
The elimination of repeats enabled generation of functional lentivirus with a re-TALE
cargo. Specifically, lentiviral particles were packaged encoding re-TALE-2A-GFP and were tested for activity of the re-TALE-TF encoded by viral particles by transfecting a mCherry reporter into a pool of lenti-reTALE-2A-GFP infected 293T cells. 293T cells transduced by lenti-re-TALE-TF
showed 36X reporter expression activation compared with the reporter only negative (see Fig.
7a,b,c). The sequence integrity of the re-TALE-TF in the lentiviral infected cells was checked and full-length reTALEs in all 10 of the clones tested were detected. (see Fig.
7d).
EXAMPLE XII
Comparison of ReTALEs and Cas9-gRNA efficiency in hiPSCs with Genome Editing Assessment System (GEAS) To compare the editing efficiencies of re-TALENs versus Cas9-gRNA in hiPSCs, a next-generation sequencing platform (Genome Editing Assessment System) was developed to identify and quantify both NHEJ and HDR gene editing events. A re-TALEN pair and a Cas9-gRNA were designed and constructed, both targeting the upstream region of CCR5 (re-TALEN, Cas9-gRNA
pair #3 in Table 3), along with a 9Ont ssODN donor identical to the target site except for a 2bp mismatch (see Fig. 2a). The nuclease constructs and donor ssODN were transfected into hiPSCs.
To quantitate the gene editing efficiency, paired-end deep sequencing on the target genomic region was conducted 3 days after transfection. HDR efficiency was measured by the percentage of reads containing the precise 2bp mismatch. NHEJ efficiency was measured by the percentage of reads carrying indels.
Delivery of the ssODN alone into hiPSCs resulted in minimal HDR and NHEJ
rates, while delivery of the re-TALENs and the ssODN led to efficiencies of 1.7% HDR and 1.2% NHEJ (see Fig. 2b). The introduction of the Cas9-gRNA with the ssODN led to 1.2% HDR and 3.4% NHEJ
efficiencies. Notably, the rate of genomic deletions and insertions peaked in the middle of the spacer region between the two reTALENs binding site, but peaked 3-4bp upstream of the Protospacer Associated Motif (PAM) sequence of Cas9-gRNA targeting site (see Fig. 2b), as would be expected since double stranded breaks take place in these regions. A
median genomic deletion size of 6bp and insertion size of 3bp generated by the re-TALENs was observed and a median deletion size of 7bp and insertion of lbp by the Cas9-gRNA was observed (see Fig. 2b), consistent with DNA lesion patterns usually generated by NHEJ (see reference 4 hereby incorporated by reference in its entirety.) Several analyses of the next-generation sequencing platform revealed that GEAS can detect HDR detection rates as low as 0.007%, which is both highly reproducible (coefficient of variation between replicates = 15% *
measured efficiency) and 400X more sensitive than most commonly used mismatch sensitive endonuclease assays (see Fig. 8).
re-TALEN pairs and Cas9-gRNA5 targeted to fifteen sites at the CCR5 genomic locus were built to determine editing efficiency (see Fig. 2c, see Table 3). These sites were selected to represent a wide range of DNaseI sensitivities (see reference 39 hereby incorporated by reference in its entirety. The nuclease constructs were transfected with the corresponding ssODNs donors (see Table 3) into PGP1 hiPSCs. Six days after transfection, the genome editing efficiencies at these sites were profiled (Table 6). For 13 out of 15 re-TALEN pairs with ssODN donors, NHEJ
and HDR was detected at levels above statistical detection thresholds, with an average NHEJ
efficiency of 0.4% and an average HDR efficiency of 0.6% (see Fig. 2c). In addition, a statistically significant positive correlation (r2 =0.81) was found between HR and NHEJ
efficiency at the same targeting loci (P<1 X 10-4) (see Fig. 9a), suggesting that DSB generation, the common upstream step of both HDR and NHEJ, is a rate-limiting step for reTALEN-mediated genome editing.
In contrast, all 15 Cas9-gRNA pairs showed significant levels of NHEJ and HR, with an average NHEJ efficiency of 3% and an average HDR efficiency of 1.0% (see Fig.
2c). In addition, a positive correlation was also detected between the NHEJ and HDR efficiency introduced by Cas9-gRNA (see Fig. 9b) (r2=0.52, p=0.003), consistent with observations for reTALENs. The NHEJ efficiency achieved by Cas9-gRNA was significantly higher than that achieved by reTALENs (t-test, paired-end, P=0.02). A moderate but statistically significant correlation between NHEJ efficiency and the melting temperature of the gRNA targeting sequence was observed (see Fig. 9c) (r2=0.28, p=0.04), suggesting that the strength of base-pairing between the gRNA and its genomic target could explain as much as 28% of the variation in the efficiency of Cas9-gRNA-mediated DSB generation. Even though Cas9-gRNA produced NHEJ levels at an average of 7 times higher than the corresponding reTALEN, Cas9-gRNA only achieved HDR
levels (average=1.0%) similar to that of the corresponding reTALENs (average = 0.6%).
Without wishing to be bound by scientific theory, these results may suggest either that the ssODN
concentration at the DSB is the limiting factor for HDR or that the genomic break structure created by the Cas9-gRNA is not favorable for effective HDR. No correlation between DNaseI HS and the genome targeting efficiencies was observed for either method. (see Fig. 10).
EXAMPLE XIII
Optimization of ssODN Donor Design for HDR
Highly-performing ssODNs in hiPSCs were designed as follows. A set of ssODNs donors of different lengths (50-170nt), all carrying the same 2bp mismatch in the middle of the spacer region of the CCR5 re-TALEN pair #3 target sites was designed. HDR efficiency was observed to vary with ssODN length, and an optimal HDR efficiency of ¨1.8% was observed with a 90 nt ssODN , whereas longer ssODNs decreased HDR efficiency (see Fig. 3a). Since longer homology regions improve HDR rates when dsDNA donors are used with nucleases (see reference 40 hereby incorporated by reference in its entirety), possible reasons for this result may be that ssODNs are used in an alternative genome repair process; longer ssODNs are less available to the genome repair apparatus; or that longer ssODNs incur negative effects that offset any improvements gained by longer homology, compared to dsDNA donors (see reference 41 hereby incorporated by reference in its entirety.) Yet, if either of the first two reasons were the case, then NHEJ rates should either be unaffected or would increase with longer ssODNs because NHEJ
repair does not involve the ssODN donor. However, NHEJ rates were observed to decline along with HDR (see Fig. 3a), suggesting that the longer ssODNs present offsetting effects.
Possible hypotheses would be that longer ssODNs are toxic to the cell (see reference 42 hereby incorporated by reference in its entirety), or that transfection of longer ssODNs saturates the DNA processing machinery, thereby causing decreased molar DNA uptake, and reducing the capacity of the cells to take up or express re-TALEN plasmids.
How rate of incorporation of a mismatch carried by the ssODN donor varies with its distance to the double stranded break ("DSB") was examined. A series of 9Ont ssODNs all possessing the same 2bp mismatch (A) in the center of the spacer region of re-TALEN pair #3 was designed. Each ssODN also contained a second 2bp mismatch (B) at varying distances from the center (see Fig. 3b). A ssODN possessing only the center 2bp mismatch was used as a control.
Each of these ssODNs was introduced individually with re-TALEN pair #3 and the outcomes were analyzed with GEAS. We found that overall HDR -- as measured by the rate at which the A
mismatch was incorporated (A only or A+B) -- decreased as the B mismatches became farther from the center (see Fig. 3b, see Fig. 11a). The higher overall HDR rate observed when B is only 10bp away from A may reflect a lesser need for annealing of the ssODN against genomic DNA
immediately proximal to the dsDNA break.
For each distance of B from A, a fraction of HDR events only incorporated the A
mismatch, while another fraction incorporated both A and B mismatches (see Fig. 3b (A only and A+B)), These two outcomes may be due to gene conversion tracts (see reference 43 hereby incorporated by reference in its entirety) along the length of the ssDNA
oligo, whereby incoporation of A+B mismatches resulted from long conversion tracts that extended beyond the B
mismatch, and incorporation of the A-only mismatch resulted from shorter tracts that did not reach B. Under this interpretation, a distribution of gene conversion lengths in both directions along the ssODN were estimated (see Fig. 11b). The estimated distribution implies that gene conversion tracts progressively become less frequent as their lengths increase, a result very similar to gene conversion tract distributions seen with dsDNA donors, but on a highly compressed distance scale of tens of bases for the ssDNA donor vs. hundreds of bases for dsDNA donors.
Consistent with this result, an experiment with a ssODN containing three pairs of 2bp mismatches spaced at intervals of lOnt on either side of the central 2bp mismatch "A" gave rise to a pattern in which A
alone was incorporated 86% of the time, with multiple B mismatches incorporated at other times (see Fig. 11c). Although the numbers of B only incorporation events were too low to estimate a distribution of tract lengths less than 10bp, it is clear that the short tract region within 10bp of the nuclease site predominates (see Fig. 11b). Finally, in all experiments with single B mismatches, a small fraction of B-only incorporation events is seen (0.04 A-0.12%) that is roughly constant across all B distances from A.
Furthermore, analysis was carried out of how far the ssODN donor can be placed from the re-TALEN-induced dsDNA break while still observing incorporation. A set of 9Ont ssODNs with central 2bp mismatches targeting a range of larger distances (-600bp to +400bp) away from the re-TALEN-induced dsDNA break site were tested. When the ssODNs matched .40bp away, we observed >30x lower HDR efficiencies compared to the control ssODN positioned centrally over the cut region (see Fig. 3c). The low level of incorporation that was observed may be due to processes unrelated to the dsDNA cut, as seen in experiments in which genomes are altered by a ssDNA donor alone see reference 42 hereby incorporated by reference in its entirety. Meanwhile, the low level of HDR present when the ssODN is ¨40bp away may be due to a combination of weakened homology on the mismatch-containing side of the dsDNA cut along with insufficient ssODN oligo length on the other side of the dsDNA break.
The ssODNs DNA donor design for Cas9-gRNA mediated targeting was tested. Cas9-gRNA (C2) targeting the AAVS1 locus was constructed and ssODN donors of variable orientations (Oc: complementary to the gRNA and On: non-complementary to the gRNA) and lengths (30, 50, 70, 90, 110 nt) were designed. Oc achieved better efficiency than On, with a 70mer Oc achieving an optimal HDR rate of 1.5%. (see Fig. 3d) The same ssODN strand bias was detected using a Cas9-derived nickase (Cc: Cas9_D1OA), despite the fact that the HDR
efficiencies mediated by Cc with ssODN were significantly less than C2 (t-test, paired-end, P=0.02). (see Fig. 12).
EXAMPLE XIV
hiPSC Clonal Isolation of Corrected Cells GEAS revealed that re-TALEN pair #3 achieved precise genome editing with an efficiency of ¨1% in hiPSCs, a level at which correctly edited cells can usually be isolated by screening clones. HiPSCs have poor viability as single cells. Optimized protocols described in reference 23 hereby incorporated by reference in its entirety along with a single-cell FACS
sorting procedure was used to establish a robust platform for single hiPSCs sorting and maintenance, where hiPSC
clones can be recovered with survival rates of >25%. This method was combined with a rapid and efficient genotyping system to conduct chromosomal DNA extraction and targeted genome amplification in 1-hour single tube reactions, enabling large scale genotyping of edited hiPSCs.
Together, these methods comprise a pipeline for robustly obtaining genome-edited hiPSCs without selection.
To demonstrate this system (see Fig. 4a), PGP1 hiPSCs were transfected with a pair of re-TALENs and an ssODN targeting CCR5 at site #3 (see Table 3). GEAS was performed with a portion of the transfected cells, finding an HDR frequency of 1.7% (see Fig.
4b). This information, along with the 25% recovery of sorted single-cell clones, allow estimation of obtaining at least one correctly-edited clone from five 96-well plates with Poisson probability 98%
(assuming =0,017 90 .4'5 2). Six days after transfection, hiPSCs were FACS-sorted and eight days after sorting, 100 hiPSC clones were screened. Sanger sequencing revealed that 2 out of 100 of these unselected hiPSC colonies contained a heterozygous genotype possessing the 2bp mutation introduced by the ssODN donor see (Fig. 4c). The targeting efficiency of 1%
(1%=2/2*100, 2 mono-allelic corrected clones out of 100 cell screened) was consistent with the next-generation sequencing analysis (1.7%) (see Fig. 4b). The pluripotency of the resulting hiPSCs was confirmed with immunostaining for SSEA4 and TRA-1-60 (see Fig. 4d). The successfully targeted hiPSCs clones were able to generate mature teratomas with features of all three germ layers (see Fig. 4e).
EXAMPLE XV
Method for Continuous Cell Genome Editing According to certain aspects, a method is provided for genome editing in cells, including a human cell, for example a human stem cell, wherein the cell is genetically modified to include a nucleic acid encoding an enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner.
Such an enzyme includes an RNA guided DNA binding protein, such as an RNA-guided DNA binding protein of a Type II CRISPR system. An exemplary enzyme is Cas9. According to this aspect, the cell expresses the enzyme and guide RNA is provided to the cell from the media surrounding the cell.
The guide RNA and the enzyme form a co-localization complex at target DNA
where the enzyme cuts the DNA. Optionally, a donor nucleic acid may be present for insertion into the DNA at the cut site, for example by nonhomologous end joining or homologous recombination. According to one aspect, the nucleic acid encoding an enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner, such as Cas9, is under the influence of a promoter, such as the nucleic acid can be activated and silenced. Such promoters are well known to those of skill in the art. One exemplary promoter is the dox inducible promoter. According to one aspect, the cell is genetically modified by having reversibly inserted into its genome the nucleic acid encoding an enzyme that forms a co-localization complex with RNA complementary to the target DNA and that cleaves the target DNA

in a site specific manner. Once inserted, the nucleic acid can be removed by use of a reagent, such as a transposase. In this manner, the nucleic acid can be easily removed after use.
According to one aspect, a continuous genome editing system in human induced pluripotent stem cells (hiPSCs) using a CRISPR system is provided. According to an exemplary aspect, the method includes use of a hiPSC line with Cas9 reversibly inserted in the genome (Cas9-hiPSCs); and gRNAs which have been modified from their native form to allow their passage from media surrounding the cells into the cells for use with the Cas9. Such gRNA
has been treated with a phosphatase in a manner to remove phosphate groups. Genome editing in the cell is carried out with Cas9 by supplementing phosphatase treated gRNA in the tissue culture media. This approach enables scarless genome editing in HiPSCs with up to 50% efficiencies with single days of treatment, 2-10X times more efficient than the best efficiencies reported so far. Further, the method is easy to use and with significantly lower cellular toxicity. Embodiments of the present disclosure include single editing of hiPSCs for biological research and therapeutic applications, multiplex editing of hiPSCs for biological research and therapeutic applications, directional hiPSCs evolution and phenotype screening of hiPSCs and its derivative cells.
According to certain aspects, other cell lines and organisms described herein can be used in addition to stem cells. For example, the method described herein can be used to animal cells such as mouse or rat cells so that stable Cas9 integrated mouse cells and rat cells can be generated and tissue specific genome editing can be conducted by locally introducing phosphatase treated gRNA
from media surrounding the cells. Moreover, other Cas9 derivatives can be inserted into many cell lines and organisms, and targeted genomic manipulations, such as sequence specific nicking, gene activation, suppression and epigenetic modification can be conducted.
Aspects of the present disclosure are directed to making stable hiPSCs with Cas9 inserted into the genome. Aspects of the present disclosure are directed to modifying RNA to enable entry into a cell through the cell wall and co-localization with Cas9 while avoiding the immune response of the cell. Such modified guide RNA can achieve optimal transfection efficiencies with minimal toxicity. Aspects of the present disclosure are directed to optimzied genome editing in Cas9-hiPSCs using phosphatase treated gRNA. Aspects of the present disclosure include eliminating Cas9 from hiPSCs to achieve scarless genome editing, where the nucleic acid encoding Cas9 has been reversibly placed into the cell genome. Aspects of the present disclosure include biomedical engineering using hiPSCs with Cas9 inserted into the genome to create desired genetic mutations.
Such engineered hiPSCs maintain pluripotency and can be successfully differentiated into various cell types, including cardiomyocyte, which fully recapitulate the phenotype of patient cell lines.
Aspects of the present disclosure include libraries of phosphatase treated gRNAs for multiplex genome editing. Aspects of the present disclosure include generating a library of PGP
cell lines with each one carrying 1 to a few designated mutations in the genome, which can serve as resource for drug screening. Aspects of the present disclosure include generating PGP1 cell lines with all the retrotranselements barcoded with different sequences to track the location and activity of this element.
EXAMPLE XVI
Generating Stable hiPSCs with Cas9 inserted into the Genome Cas9 was encoded under the dox inducible promoter and the construct was placed into a Piggybac vector which can be inserted into and removed out of the genome with the help of Piggybac transposase. PCR reaction validated the stable insertion of the vector (see Fig. 14). The inducible Cas9 expression was determined via RT-QPCR. The mRNA level of Cas9 increased 1000X after 8 hours of lug/mL DOX supplementation in the culture media and the level of Cas9 mRNA dropped to normal level ¨ 20 hours after withdrawal of the DOX. (See Fig.
15).
According to one aspect, the Cas9-hiPSC system based genome editing bypasses the transfection procedure of Cas9 plasmid/RNA, a large construct usually with <
1% transfection efficiency in hiPSCs. The present Cas9-hiPSC system can serve as a platform to perform high efficient genomic engineering in human stem cells. In addition, the Cas9 cassette introduced into the hiPSCs using Piggybac system can be removed out from the genome easily upon introducing of transposases.
EXAMPLE XVII
Phosphatase Treated Guide RNA
To enable continuous genome editing on Cas9-hiPSCs, a series of modified RNA
encoding gRNA were generated and supplemented into Cas9-iPS culture medium in complex with liposome.
Phosphatase treated native RNA without any capping achieved the optimal HDR
efficiency of 13%, 30X more than previously reported 5'Cap-Mod RNA (see Fig. 16).
According to one aspect, guide RNA is physically attached to the donor DNA. In this manner, a method is provided of coupling Cas9 mediated genomic cutting and ssODN-mediated HDR, thus stimulating sequence specific genomic editing. gRNA linked with DNA
ssODN donor with optimized concentration achieved 44% HDR and unspecific NHEJ 2% (see Fig.
17). Of note, this procedure does not incurred visible toxicity as observed with nucleofection or electroporation.
According to one aspect, the present disclosure provides an in vitro engineered RNA
structure encoding gRNA, which achieved high transfection efficiency, genome editing efficiency in collaboration with genomically inserted Cas9. In addition, the present disclosure provides a gRNA-DNA chimeric construct to couple a genomic cutting event with the homology directed recombination reaction.

EXAMPLE XVIII
Eliminating Reversibly Engineered Cas9 from hiPSCs to Achieve Scarless Genome Editing According to certain aspects, a Cas9 cassette is inserted into the genome of hiPSC cells using a reversible vector. Accordingly, a Cas9 cassette was reversibly inserted into the genome of hiPSC cells using a PiggyBac vector. The Cas9 cassette was removed from the genome edited hiPSCs by transfecting the cell with transposase-encoding plasmid.
Accordingly, aspects of the present disclosure include use of a reversible vector, which is known to those of skill in the art. A
reversible vector is one which can be inserted into a genome, for example, and then removed with a corresponding vector removal enzyme. Such vectors and corresponding vector removal enzymes are known to those of skill in the art. A screen was performed on colonized iPS cells and colonies devoid of Cas9-cassette were recovered as confirmed by PCR reaction.
Accordingly, the present disclosure provides method of genome editing without affecting the rest of the genome by having a permanent Cas9 cassette present in the cell.
EXAMPLE XIX
Genome Editing in iPGP1 Cells Research into the pathogenesis of cardiomyopathy has historically been hindered by the lack of suitable model systems. Cardiomyocyte differentiation of patient-derived induced pluripotent stem cells (iPSCs) offers one promising avenue to surmount this barrier, and reports of iPSC modeling of cardiomyopathy have begun to emerge. However, realization of this promise will require approaches to overcome genetic heterogeneity of patient-derived iPSC lines.
Cas9-iPGP1 cell lines and phosphatase treated guide RNA bound to DNA were used to generated three iPSC lines that are isogenic except for the sequence at TAZ
exon 6, which was identified to carry single nucleotide deletion in Barth syndrome patients.
Single round of RNA
transfection achieved ¨30% HDR efficiency. Modified Cas9-iPGP1 cells with desired mutations were colonized (see Fig. 18) and the cell lines were differentiated into cardiomyocyte.
Cardiomyocyte derived from the engineered Cas9-iPGP1 fully recapitulated the cardiolipin, mitochondrial, and ATP deficits observed in patient-derived iPSCs and in the neonatal rat TAZ
knockdown model (see Fig. 19). Accordingly, methods are provided for correcting mutations causing diseases in pluripotent cells followed by differentiation of the cells into desired cell types.
EXAMPLE XX
Materials and Methods 1. Establishment of PiggyBac Cas9 dox inducible stable human iPS/ES
lines 1. After cells reached 70% confluence pretreat the culture with ROCK inhibitor Y27632 at final concentration of 10uM for overnight.

2. The next day prepare the nucleofection solution by combine the 82 [L1 of human stem cell nucleofector solution and 18u1 supplement 1 in a sterile 1.5 ml eppendorf tube. Mix well.
Incubate solution at 37 C for 5 mins.
3. Aspirate mTeSR1; gently rinse the cells with DPBS at 2 mL/well of a six-well plate.
4. Aspirate the DPBS, add 2 mL/well of Versene, and put the culture back to incubator at 37 C until they become rounded up and loosely adherent, but not detached. This requires 3-7 min.
5. Gently aspirate the Versene and add mTeSR1. Add lml mTeSR1 and dislodge the cells by gently flowing mTeSR1 over them with a 1,000 uL micropipette.
6. Collect the dislodged cells, gently triturate them into a single-cell suspension, and quantitate by hemacytometer and adjust cell density to lmillion cells per ml.
7. Add lml cell suspension to1.5m1 eppendorf tube and centrifuge at 1100 RPM for 5 min in a bench top centrifuge.
8. Resuspend cells in 100 [L1 of human stem cell nucleofector solution from Step 2.
9. Transfer cells to a nucleofector cuvette using a 1 ml pipette tip. Add 1 [tg of plasmid Transposonase and 5ug PB Cas9 plasmids into the cell suspension in the cuvette. Mix cells and DNA by gentle swirling.
10. Put the cuvette into the nucleofector. Programs B-016 was selected and nucleofect cells by pressing button X.
ii. Add 500u1 mTeSR1 medium with ROCK inhibitor in the cuvette after nucleofection.
12. Asperate the nucleofected cells from the cuvette using the provided Pasteur plastic pipette.
And transfer cells drop-wise into matrigel coated well of 6 well plate mTeSR1 mediun with ROCK inhibitor. Incubate the cells at 37 C overnight.
13. Change the medium to mTesrl the next day and after 72 hours of transfection;add puromycin at final concentration at lug/ml.And the line will be set up within 7 days.
2. RNA preparation 1. Prepare DNA template with T7 promoter upstream of gRNA coding sequence.
2. Purify the DNA using Mega Clear Purification and normalize the concentration.
3. Prepare Custom NTPS mixtures for different gRNA production.
#1 Native RNA Mix [Final] (mM) GTP 7.5 ATP 7.5 CTP 7.5 UTP 7.5 1 Total volume ______________________________________________ 1 #2 Capped Native RNA
Mix [Final] (mM) 3'-0-Me-m7G Cap structure analog (NEB) 6 GTP 1.5 ATP 7.5 CTP 7.5 UTP 7.5 Total volume #3 Modified RNA Mix [Final] (mM) GTP 7.5 ATP 7.5 5-Me-CTP (Tr-Link) 7.5 Pseudo-UTP (Tr-Link) 7.5 Total volume #4 Capped/Modified RNA
Mix [Final] (mM) 3'-0-Me-m7G Cap structure analog (NEB) 6 GTP 1.5 ATP 7.5 5-Me-CTP (Tri-Link) 7.5 Pseudo-UTP (Tr-Link) 7.5 Total volume 4. Prepare the in vitro transcription mix at room temperature.
Amt (ul) Custom NTPS (*Add NA

vol/IVT rxn as indicated above) on ice PCR product (10Ong/u1) =
1600ng total 16 ([final]=4Ong/u1) Buffer X10 (MEGAscript kit from Ambion) @ RT
T7 Enzyme (MEGAscript kit from Ambion) 5. Incubate for 4 hours (3-6hrs ok) at 37 C (thermocycler).
6. Add 2 1 Turbo DNAse (MEGAscript kit from Ambion) to each sample. Mix gently and incubate at 37 C for 15.
7. Purify DNAse treated reaction using MegaClear from Ambion according to the manufacturer's instructions.
8. Purify RNA using MEGAclear. (Purified RNA can be stored at -80 for several months).
9. To remove phosphate groups to avoid Toll 2 immune reaction from the host cell.
RNA Phosphatase treatment 1X 12 For each RNA sample ¨100u1 NA
10X Antarctic Phosphatase buffer llul 132 Antarctic Phosphatase 2u1 24 Gently mix sample and incubate at 37 C for 30' (30'- 1 hr ok) 3. RNA transfection 1. Plate 10K-20K cells per 48 well without antibiotics. Cells should be 30-50% confluent for transfection.
2. Change the cell media to with B18R (200ng/m1), DOX (lug/m1), Puromycin (2ug/m1) at least two hours before the transfection.
3. Prepare the transfection reagent containing gRNA (0.5ug-2ug), donor DNA
(0.5ug-2ug)and RNAiMax, incubate the mixture in rm temperature for 15 minutes and transfer to the cell.
4. Single human iPS cells seed and single clone pickup 1. After 4 days of dox induction and 1 day dox withdraw,asperate the medium,rinse gently with the DPBS.add 2 mL/well of Versene, and put the culture back to incubator at 37 C until they become rounded up and loosely adherent, but not detached. This requires 3-7 min.
2. Gently aspirate the Versene and add mTeSR1 .Add lml mTeSR1 and dislodge the cells by gently flowing mTeSR1 over them with a 1,000 uL micropipette.
3. Collect the dislodged cells, gently triturate them into a single-cell suspension, and quantitate by hemacytometer and adjust cell density to 100K cells per ml.
4. Seeding the cells into matrigel coated 10 cm dishes with mTeSR1 plus ROCK
inhibitor at cell density of 50K,100K and 400K per 10 cm dish.
5. Single cell formed Clones screening 1. After 12 days culture in 10 cm dish and clones are big enough to be identified by naked eyes and labeled by colon marker. Do not allow clones become too big and adhere to each other.
2. Put the 10 cm dish to the culture hood and using a P20 pipette(set at 10u1) with filter tips. Aspirate lOul medium for one well of 24 well plates. Pick up clone by scratching the clone into small pieces and transfer to one well of 24 well plate. Each filter tip for each clone.
3. After 4-5 days the clones inside one well of 24 well plate become big enough to split.
4. Aspirate the medium and rinse with 2 mL/well DPBS.
5. Aspirate DPBS, replace with 250u1/well dispase (0.1 U/mL).
and incubate the cells in dispase at 37 C for 7 min.
6. Replace the dispase with 2m1 DPBS.
7. Add 250u1 mTeSR1. Using a cell scraper to lodge off the cells and collect the cells.
8. Tranfer 125u1 cell suspension into a well of matrigel coated 24 wells plates.
9. Transfer 125u1 cell suspension into 1.5m1 eppendorf tube for genomic DNA
extraction.
6. Clone screening 1. Centrifuge the tube from step7.7 2. Aspirate the medium and add 250u1 lysis buffer per well (10mM+TrispH7.5+(or+8.0),10mMEDTA,10mM.
3. NaC1,+10%SDS,4Oug/mL+proteinase K(added fresh before using the buffer).
4. Incubate at 55 overnight.
5. PrecipitateDNA by adding 250u1Isopropanol.
6. Spin for 30 minutes at highest speed. Wash with 70% ethanol.
7. Gently remove ethanol.Air dry for 5 min.
8. Resuspend gDNA with100-200u1 dH20.

9. PCR amplification of the targeted genomic region with specific primers.
10. Sanger sequencing the PCR product with respective primer.
11. Analysis of Sanger sequence data and expansion of targeted clones.
7. Piggybac vector remove 1. Repeat the step 2.1-2.9 2. Transfer cells to a nucleofector cuvette using a 1 ml pipette tip.Add 2 [ig plasmid of Transposonase into the cell suspension in the cuvette. Mix cells and DNA by gentle swirling.
3. Repeat the step 2.10-2.11 4. Asperate the nucleofected cells from the cuvette using the provided Pasteur plastic pipette. And transfer cells drop-wise into matrigel coated well of 10 cm dish with mTeSR1 medium plus ROCK inhibitor. Incubate the cells at 37 C overnight.
5. The next day change the medium to mTesrl and change the medium every day for 4 following days.
6. After the clones became big enough pick up 20-50 clones and seeding into 24 well.
7. Genotype the clones with PB Cas9 PiggyBac vector primers and expansion negative clones.
References References are designated throughout the specification by their number below and are incorporated into the specification as if fully set forth therein. Each of the following references is hereby incorporated by reference in its entirety.
1. Carroll,D. (2011) Genome engineering with zinc-finger nucleases. Genetics, 188, 773-82.
2. Wood,A.J., Lo,T.-W., Zeitler,B., Pickle,C.S., Ralston,E.J., Lee,A.H., Amora,R., Miller,J.C., Leung,E., Meng,X., et al. (2011) Targeted genome editing across species using ZFNs and TALENs. Science (New York, N.Y.), 333, 307.
3. Perez-Pinera,P., Ousterout,D.G. and Gersbach,C.A. (2012) Advances in targeted genome editing. Current opinion in chemical biology, 16, 268-77.
4. Symington,L.S. and Gautier,J. (2011) Double-strand break end resection and repair pathway choice. Annual review of genetics, 45, 247-71.

5. Urnov,F.D., Miller,J.C., Lee,Y.-L., Beausejour,C.M., Rock,J.M., Augustus,S., Jamieson,A.C., Porteus,M.H., Gregory,P.D. and Holmes,M.C. (2005) Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature, 435, 646-51.
6. Boch,J., Scholze,H., Schomack,S., Landgraf,A., Hahn,S., Kay,S., Lahaye,T., Nickstadt,A. and Bonas,U. (2009) Breaking the code of DNA binding specificity of TAL-type III
effectors.
Science (New York, N.Y.), 326, 1509-12.
7. Cell,P., Replacement,K.S., Talens,A., Type,A., Collection,C., Ccl-,A. and Quickextract,E.
Genetic engineering of human pluripotent cells using TALE nucleases.
8. Mussolino,C., Morbitzer,R., Liitge,F., Dannemann,N., Lahaye,T. and Cathomen,T. (2011) A
novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic acids research, 39, 9283-93.
9. Ding,Q., Lee,Y., Schaefer,E.A.K., Peters,D.T., Veres,A., Kim,K., Kuperwasser,N., Motola,D.L., Meissner,T.B., Hendriks,W.T., et al. (2013) Resource A TALEN Genome-Editing System for Generating Human Stem Cell-Based Disease Models.
10. Hockemeyer,D., Wang,H., Kiani,S., Lai,C.S., Gao,Q., Cassady,J.P., Cost,G.J., Zhang,L., Santiago,Y., Miller,J.C., et al. (2011) Genetic engineering of human pluripotent cells using TALE nucleases. Nature biotechnology, 29, 731-4.
11. Bedell,V.M., Wang,Y., Campbell,J.M., Poshusta,T.L., Starker,C.G., Krug Ii,R.G., Tan,W., Penheiter,S.G., Ma,A.C., Leung,A.Y.H., et al. (2012) In vivo genome editing using a high-efficiency TALEN system. Nature, 490, 114-118.
12. Miller,J.C., Tan,S., Qiao,G., Barlow,K. a, Wang,J., Xia,D.F., Meng,X., Paschon,D.E., Leung,E., Hinkley,S.J., et al. (2011) A TALE nuclease architecture for efficient genome editing. Nature biotechnology, 29, 143-8.
13. Holkers,M., Maggio,I., Liu,J., Janssen,J.M., Miselli,F., Mussolino,C., Recchia,A., Cathomen,T.
and Goncalves,M. a F. V (2012) Differential integrity of TALE nuclease genes following adenoviral and lentiviral vector gene transfer into human cells. Nucleic acids research, 10.1093/nar/gks1446.

14. Reyon,D., Tsai,S.Q., Khayter,C., Foden,J. a, Sander,J.D. and Joung,J.K.
(2012) FLASH
assembly of TALENs for high-throughput genome editing. Nature Biotechnology, 30, 460-465.
15. Qiu,P., Shandilya,H., D'Alessio,J.M., O'Connor,K., Durocher,J. and Gerard,G.F. (2004) Mutation detection using Surveyor nuclease. BioTechniques, 36, 702-7.
16. Mali,P., Yang,L., Esvelt,K.M., Aach,J., Guell,M., DiCarlo,J.E., Norville,J.E. and Church,G.M.
(2013) RNA-guided human genome engineering via Cas9. Science (New York, N.Y.), 339, 823-6.
17. Ding,Q., Regan,S.N., Xia,Y., Oostrom,L.A., Cowan,C.A. and Musunuru,K.
(2013) Enhanced Efficiency of Human Pluripotent Stem Cell Genome Editing through Replacing TALENs with CRISPRs. Cell Stem Cell, 12, 393-394.
18. Cong,L., Ran,F.A., Cox,D., Lin,S., Barretto,R., Habib,N., Hsu,P.D., Wu,X., Jiang,W., Marraffini,L. a, et al. (2013) Multiplex genome engineering using CRISPR/Cas systems.
Science (New York, N.Y.), 339, 819-23.
19. Cho,S.W., Kim,S., Kim,J.M. and Kim,J.-S. (2013) Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nature biotechnology, 31, 230-232.
20. Hwang,W.Y., Fu,Y., Reyon,D., Maeder,M.L., Tsai,S.Q., Sander,J.D., Peterson,R.T., Yeh,J.-R.J. and Joung,J.K. (2013) Efficient genome editing in zebrafish using a CRISPR-Cas system. Nature biotechnology, 31, 227-229.
21. Chen,F., Pruett-Miller,S.M., Huang,Y., Gjoka,M., Duda,K., Taunton,J., Collingwood,T.N., Frodin,M. and Davis,G.D. (2011) High-frequency genome editing using ssDNA
oligonucleotides with zinc-finger nucleases. Nature methods, 8, 753-5.
22. Soldner,F., Laganiere,J., Cheng,A.W., Hockemeyer,D., Gao,Q., Alagappan,R., Khurana,V., Golbe,L.I., Myers,R.H., Lindquist,S., et al. (2011) Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations.
Cell, 146, 318-31.
23. Valamehr,B., Abujarour,R., Robinson,M., Le,T., Robbins,D., Shoemaker,D.
and Flynn,P.
(2012) A novel platform to enable the high-throughput derivation and characterization of feeder-free human iPSCs. Scientific reports, 2, 213.
24. Sanjana,N.E., Cong,L., Zhou,Y., Cunniff,M.M., Feng,G. and Zhang,F. (2012) A transcription activator-like effector toolbox for genome engineering. Nature protocols, 7, 171-92.
25. Gibson,D.G., Young,L., Chuang,R., Venter,J.C., Iii,C.A.H., Smith,H.O. and America,N. (2009) Enzymatic assembly of DNA molecules up to several hundred kilobases. 6, 12-16.
26. Zou,J., Maeder,M.L., Mali,P., Pruett-Miller, S.M., Thibodeau-Beganny,S., Chou,B.-K., Chen,G., Ye,Z., Park,I.-H., Daley,G.Q., et al. (2009) Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell stem cell, 5, 97-110.
27. Perez,E.E., Wang,J., Miller,J.C., Jouvenot,Y., Kim,K. a, Liu,O., Wang,N., Lee,G., Bartsevich,V. V, Lee,Y.-L., et al. (2008) Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nature biotechnology, 26, 808-16.
28. Bhakta,M.S., Henry,I.M., Ousterout,D.G., Das,K.T., Lockwood,S.H., Meckler,J.F., Wallen,M.C., Zykovich,A., Yu,Y., Leo,H., et al. (2013) Highly active zinc-finger nucleases by extended modular assembly. Genome research, 10.1101/gr.143693.112.
29. Kim,E., Kim,S., Kim,D.H., Choi,B.-S., Choi,I.-Y. and Kim,J.-S. (2012) Precision genome engineering with programmable DNA-nicking enzymes. Genome research, 22, 1327-33.
30. Gupta,A., Meng,X., Zhu,L.J., Lawson,N.D. and Wolfe,S. a (2011) Zinc finger protein-dependent and -independent contributions to the in vivo off-target activity of zinc finger nucleases. Nucleic acids research, 39, 381-92.
31. Park,I.-H., Lerou,P.H., Zhao,R., Huo,H. and Daley,G.Q. (2008) Generation of human-induced pluripotent stem cells. Nature protocols, 3, 1180-6.
32. Cermak,T., Doyle,E.L., Christian,M., Wang,L., Zhang,Y., Schmidt,C., Baller,J.A., Somia,N. V, Bogdanove,A.J. and Voytas,D.F. (2011) Efficient design and assembly of custom TALEN
and other TAL effector-based constructs for DNA targeting. Nucleic acids research, 39, e82.
33. Briggs,A.W., Rios,X., Chari,R., Yang,L., Zhang,F., Mali,P. and Church,G.M.
(2012) Iterative capped assembly: rapid and scalable synthesis of repeat-module DNA such as TAL
effectors from individual monomers. Nucleic acids research, 10.1093/nar/gks624.
34. Zhang,F., Cong,L., Lodato,S., Kosuri,S., Church,G.M. and Arlotta,P. (2011) LETTErs Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. 29, 149-154.
35. Pathak,V.K. and Temin,H.M. (1990) Broad spectrum of in vivo forward mutations, hypermutations, and mutational hotspots in a retroviral shuttle vector after a single replication cycle: substitutions, frameshifts, and hypermutations. Proceedings of the National Academy of Sciences of the United States of America, 87, 6019-23.
36. Tian,J., Ma,K. and Saaem,I. (2009) Advancing high-throughput gene synthesis technology.
Molecular bioSystems, 5, 714-22.
37. Zou,J., Mali,P., Huang,X., Dowey,S.N. and Cheng,L. (2011) Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease.
Blood, 118, 4599-608.
38. Mali,P., Yang,L., Esvelt,K.M., Aach,J., Guell,M., Dicarlo,J.E., Norville,J.E. and Church,G.M.
(2013) RNA-Guided Human Genome.
39. Boyle,A.P., Davis,S., Shulha,H.P., Meltzer,P., Margulies,E.H., Weng,Z., Furey,T.S. and Crawford,G.E. (2008) High-resolution mapping and characterization of open chromatin across the genome. Cell, 132, 311-22.
40. Orlando,S.J., Santiago,Y., DeKelver,R.C., Freyvert,Y., Boydston,E. a, Moehle,E. a, Choi,V.M., Gopalan,S.M., Lou,J.F., Li,J., et al. (2010) Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic acids research, 38, e152.
41. Wang,Z., Zhou,Z.-J., Liu,D.-P. and Huang,J.-D. (2008) Double-stranded break can be repaired by single-stranded oligonucleotides via the ATM/ATR pathway in mammalian cells.
Oligonucleotides, 18, 21-32.
42. Rios,X., Briggs,A.W., Christodoulou,D., Gorham,J.M., Seidman,J.G. and Church,G.M. (2012) Stable gene targeting in human cells using single-strand oligonucleotides with modified bases. PloS one, 7, e36697.
43. Elliott,B., Richardson,C., Winderbaum,J., Jac,A., Jasin,M. and Nickoloff,J.A.C.A. (1998) Gene Conversion Tracts from Double-Strand Break Repair in Mammalian Cells Gene Conversion Tracts from Double-Strand Break Repair in Mammalian Cells. 18.
44. Lombardo,A., Genovese,P., Beausejour,C.M., Colleoni,S., Lee,Y.-L., Kim,K.
a, Ando,D., Urnov,F.D., Galli,C., Gregory,P.D., et al. (2007) Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nature biotechnology, 25, 1298-306.
45. Jinek,M., Chylinski,K., Fonfara,I., Hauer,M., Doudna,J. a and Charpentier,E. (2012) A
programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.
Science (New York, N.Y.), 337, 816-21.
46. Shrivastav,M., De Haro,L.P. and Nickoloff,J. a (2008) Regulation of DNA
double-strand break repair pathway choice. Cell research, 18, 134-47.
47. Kim,Y.G., Cha,J. and Chandrasegaran,S. (1996) Hybrid restriction enzymes:
zinc finger fusions to Fok I cleavage domain. Proceedings of the National Academy of Sciences of the United States of America, 93, 1156-60.
48. Mimitou,E.P. and Symington,L.S. (2008) Sae2, Exol and Sgsl collaborate in DNA double-strand break processing. Nature, 455, 770-4.
49. Doyon,Y., Choi,V.M., Xia,D.F., Vo,T.D., Gregory,P.D. and Holmes,M.C.
(2010) Transient cold shock enhances zinc-finger nuclease-mediated gene disruption. Nature methods, 7, 459-60.

Claims

Claims:
1. A method of altering target DNA in a cell comprising introducing into a cell a TALEN lacking repeat sequences 100 bp or longer wherein the TALEN cleaves the target DNA and the cell undergoes nonhomologous end joining to produce altered DNA in the cell.
2. The method of claim 1 wherein the TALEN lacks repeat sequences 90 bp or longer.
3. The method of claim 1 wherein the TALEN lacks repeat sequences 80 bp or longer.
4. The method of claim 1 wherein the TALEN lacks repeat sequences 70 bp or longer.
5. The method of claim 1 wherein the TALEN lacks repeat sequences 60 bp or longer.
6. The method of claim 1 wherein the TALEN lacks repeat sequences 50 bp or longer.
7. The method of claim 1 wherein the TALEN lacks repeat sequences 40 bp or longer.
8. The method of claim 1 wherein the TALEN lacks repeat sequences 30 bp or longer.
9. The method of claim 1 wherein the TALEN lacks repeat sequences 20 bp or longer.
10. The method of claim 1 wherein the TALEN lacks repeat sequences 19 bp or longer.
11. The method of claim 1 wherein the TALEN lacks repeat sequences 18 bp or longer.
12. The method of claim 1 wherein the TALEN lacks repeat sequences 17 bp or longer.
13. The method of claim 1 wherein the TALEN lacks repeat sequences 16 bp or longer.
14. The method of claim 1 wherein the TALEN lacks repeat sequences 15 bp or longer.
15. The method of claim 1 wherein the TALEN lacks repeat sequences 14 bp or longer.
16. The method of claim 1 wherein the TALEN lacks repeat sequences 13 bp or longer.

17. The method of claim 1 wherein the TALEN lacks repeat sequences 12 bp or longer.
18. The method of claim 1 wherein the TALEN lacks repeat sequences 11 bp or longer.
19. The method of claim 1 wherein the TALEN lacks repeat sequences 10 bp or longer.
20. The method of claim 1 wherein the cell is a eukaryotic cell.
21. The method of claim 1 wherein the cell is a yeast cell, a plant cell or an animal cell.
22. The method of claim 1 wherein the cell is a somatic cell.
23. The method of claim 1 wherein the cell is a stem cell.
24. The method of claim 1 wherein the cell is a human stem cell.
25. The method of claim 1 comprising introducing into the cell a first foreign nucleic acid encoding the TALEN, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
26. The method of claim 1 comprising introducing into the cell a virus including a first foreign nucleic acid encoding the TALEN, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
27. The method of claim 1 comprising introducing into the cell a first foreign nucleic acid encoding the TALEN
having a TALE
sequence CTAACCCCTGAACAGGTAGTCGCTATAGCTTCAAATATCGGGGGCAAGCAAGC
ACTTGAGACCGTTCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCG
GAGCAAGTCGTCGCGATCGCGAGCAACGGCGGGGGGAAGCAGGCGCTGGAAACTGTT
CAGAGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAGGTTGTCG
CAATAGCAAGTAATATAGGCGGTAAGCAAGCCCTAGAGACTGTGCAACGCCTGCTCC
CCGTGCTGTGTCAGGCTCACGGTCTGACACCTGAACAAGTTGTCGCGATAGCCAGTCA
CGACGGGGGAAAACAAGCTCTAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAA

GCACATGGGTTAACACCCGAACAAGTAGTAGCGATAGCGTCAAATAACGGGGGTAAA
CAGGCTTTGGAGACGGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTA
CGCCAGAACAGGTGGTTGCAATTGCCTCCAACATCGGCGGGAAACAAGCGTTGGAAA
CTGTGCAGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAGGTT
GTGGCCATCGCTAGCCACGACGGAGGGAAGCAGGCTCTTGAAACCGTACAGCGACTT
CTCCCAGTTTTGTGCCAAGCTCACGGGCTAACCCCCGAGCAAGTAGTTGCCATAGCAA
GCAACGGAGGAGGAAAACAGGCATTAGAAACAGTTCAGCGCTTGCTCCCGGTACTCT
GTCAGGCACACGGTCTAACTCCGGAACAGGTCGTAGCCATTGCTTCCCATGATGGCGG
CAAACAGGCGCTAGAGACAGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGC
CTCACCCCGGAGCAGGTCGTTGCCATCGCCAGTAATATCGGCGGAAAGCAAGCTCTCG
AAACAGTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCA
GGTAGTGGCAATCGCATCTCACGATGGAGGTAAACAAGCACTCGAGACTGTCCAAAG
ATTGTTACCCGTACTATGCCAAGCGCATGGTTTAACCCCAGAGCAAGTTGTGGCTATT
GCATCTAACGGCGGTGGCAAACAAGCCTTGGAGACAGTGCAACGATTACTGCCTGTCT
TATGTCAGGCCCATGGCCTTACTCCTGAGCAAGTCGTAGCTATCGCCAGCAACATAGG
TGGGAAACAGGCCCTGGAAACCGTACAACGTCTCCTCCCAGTACTTTGTCAAGCACAC
GGGTTGACACCGGAACAAGTGGTGGCGATTGCGTCCAACGGCGGAGGCAAGCAGGCA
CTGGAGACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAG
AGCAGGTGGTAGCAATAGCGTCGAACATCGGTGGTAAGCAAGCGCTTGAAACGGTCC
AGCGTCTTCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACCAGAACAAGTGGTTGC
TATTGCTAGTAACAACGGTGGAAAGCAGGCCCTCGAGACGGTGCAGAGGTTACTTCCC
GTCCTCTGTCAAGCGCACGGCCTCACTCCAGAGCAAGTGGTTGCGATCGCTTCAAACA
ATGGTGGAAGACCTGCCCTGGAA, or a sequence having at least 90% sequence identity to the TALE sequence, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
28. The method of claim 1 comprising introducing into the cell a virus including a first foreign nucleic acid encoding the TALEN
having a TALE sequence CTAACCCCTGAACAGGTAGTCGCTATAGCTTCAAATATCGGGGGCAAGCAAGC
ACTTGAGACCGTTCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCG
GAGCAAGTCGTCGCGATCGCGAGCAACGGCGGGGGGAAGCAGGCGCTGGAAACTGTT
CAGAGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAGGTTGTCG
CAATAGCAAGTAATATAGGCGGTAAGCAAGCCCTAGAGACTGTGCAACGCCTGCTCC
CCGTGCTGTGTCAGGCTCACGGTCTGACACCTGAACAAGTTGTCGCGATAGCCAGTCA

CGACGGGGGAAAACAAGCTCTAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAA
GCACATGGGTTAACACCCGAACAAGTAGTAGCGATAGCGTCAAATAACGGGGGTAAA
CAGGCTTTGGAGACGGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTA
CGCCAGAACAGGTGGTTGCAATTGCCTCCAACATCGGCGGGAAACAAGCGTTGGAAA
CTGTGCAGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAGGTT
GTGGCCATCGCTAGCCACGACGGAGGGAAGCAGGCTCTTGAAACCGTACAGCGACTT
CTCCCAGTTTTGTGCCAAGCTCACGGGCTAACCCCCGAGCAAGTAGTTGCCATAGCAA
GCAACGGAGGAGGAAAACAGGCATTAGAAACAGTTCAGCGCTTGCTCCCGGTACTCT
GTCAGGCACACGGTCTAACTCCGGAACAGGTCGTAGCCATTGCTTCCCATGATGGCGG
CAAACAGGCGCTAGAGACAGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGC
CTCACCCCGGAGCAGGTCGTTGCCATCGCCAGTAATATCGGCGGAAAGCAAGCTCTCG
AAACAGTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCA
GGTAGTGGCAATCGCATCTCACGATGGAGGTAAACAAGCACTCGAGACTGTCCAAAG
ATTGTTACCCGTACTATGCCAAGCGCATGGTTTAACCCCAGAGCAAGTTGTGGCTATT
GCATCTAACGGCGGTGGCAAACAAGCCTTGGAGACAGTGCAACGATTACTGCCTGTCT
TATGTCAGGCCCATGGCCTTACTCCTGAGCAAGTCGTAGCTATCGCCAGCAACATAGG
TGGGAAACAGGCCCTGGAAACCGTACAACGTCTCCTCCCAGTACTTTGTCAAGCACAC
GGGTTGACACCGGAACAAGTGGTGGCGATTGCGTCCAACGGCGGAGGCAAGCAGGCA
CTGGAGACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAG
AGCAGGTGGTAGCAATAGCGTCGAACATCGGTGGTAAGCAAGCGCTTGAAACGGTCC
AGCGTCTTCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACCAGAACAAGTGGTTGC
TATTGCTAGTAACAACGGTGGAAAGCAGGCCCTCGAGACGGTGCAGAGGTTACTTCCC
GTCCTCTGTCAAGCGCACGGCCTCACTCCAGAGCAAGTGGTTGCGATCGCTTCAAACA
ATGGTGGAAGACCTGCCCTGGAA, or a sequence having at least 90% sequence identity to the TALE sequence, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
29. A method of altering target DNA in a cell comprising combining within a cell a TALEN lacking repeat sequences 100 bp or longer and a donor nucleic acid sequence wherein the TALEN cleaves the target DNA and the donor nucleic acid sequence is inserted into the DNA in the cell.
30. The method of claim 29 wherein the cell undergoes nonhomologous end joining to produce altered DNA in the cell.

31. The method of claim 29 wherein the cell undergoes homologous recombination to produced altered DNA in the cell.
32. The method of claim 29 wherein the TALEN lacks repeat sequences 90 bp or longer.
33. The method of claim 29 wherein the TALEN lacks repeat sequences 80 bp or longer.
34. The method of claim 29 wherein the TALEN lacks repeat sequences 70 bp or longer.
35. The method of claim 29 wherein the TALEN lacks repeat sequences 60 bp or longer.
36. The method of claim 29 wherein the TALEN lacks repeat sequences 50 bp or longer.
37. The method of claim 29 wherein the TALEN lacks repeat sequences 40 bp or longer.
38. The method of claim 29 wherein the TALEN lacks repeat sequences 30 bp or longer.
39. The method of claim 29 wherein the TALEN lacks repeat sequences 20 bp or longer.
40. The method of claim 29 wherein the TALEN lacks repeat sequences 19 bp or longer.
41. The method of claim 29 wherein the TALEN lacks repeat sequences 18 bp or longer.
42. The method of claim 29 wherein the TALEN lacks repeat sequences 17 bp or longer.
43. The method of claim 29 wherein the TALEN lacks repeat sequences 16 bp or longer.
44. The method of claim 29 wherein the TALEN lacks repeat sequences 15 bp or longer.
45. The method of claim 29 wherein the TALEN lacks repeat sequences 14 bp or longer.
46. The method of claim 29 wherein the TALEN lacks repeat sequences 13 bp or longer.
47. The method of claim 29 wherein the TALEN lacks repeat sequences 12 bp or longer.
48. The method of claim 29 wherein the TALEN lacks repeat sequences 11 bp or longer.

48. The method of claim 29 wherein the TALEN lacks repeat sequences 10 bp or longer.
50. The method of claim 29 wherein the cell is a eukaryotic cell.
51. The method of claim 29 wherein the cell is a yeast cell, a plant cell or an animal cell.
52. The method of claim 29 wherein the cell is a somatic cell.
53. The method of claim 29 wherein the cell is a stem cell.
54. The method of claim 29 wherein the cell is a human stem cell.
55. The method of claim 29 comprising introducing into the cell a first foreign nucleic acid encoding the TALEN, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
56. The method of claim 29 comprising introducing into the cell a virus including a first foreign nucleic acid encoding the TALEN, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
57. The method of claim 29 comprising introducing into the cell a first foreign nucleic acid encoding the TALEN
having a TALE
sequence CTAACCCCTGAACAGGTAGTCGCTATAGCTTCAAATATCGGGGGCAAGCAAGC
ACTTGAGACCGTTCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCG
GAGCAAGTCGTCGCGATCGCGAGCAACGGCGGGGGGAAGCAGGCGCTGGAAACTGTT
CAGAGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAGGTTGTCG
CAATAGCAAGTAATATAGGCGGTAAGCAAGCCCTAGAGACTGTGCAACGCCTGCTCC
CCGTGCTGTGTCAGGCTCACGGTCTGACACCTGAACAAGTTGTCGCGATAGCCAGTCA
CGACGGGGGAAAACAAGCTCTAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAA
GCACATGGGTTAACACCCGAACAAGTAGTAGCGATAGCGTCAAATAACGGGGGTAAA
CAGGCTTTGGAGACGGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTA
CGCCAGAACAGGTGGTTGCAATTGCCTCCAACATCGGCGGGAAACAAGCGTTGGAAA

CTGTGCAGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAGGTT
GTGGCCATCGCTAGCCACGACGGAGGGAAGCAGGCTCTTGAAACCGTACAGCGACTT
CTCCCAGTTTTGTGCCAAGCTCACGGGCTAACCCCCGAGCAAGTAGTTGCCATAGCAA
GCAACGGAGGAGGAAAACAGGCATTAGAAACAGTTCAGCGCTTGCTCCCGGTACTCT
GTCAGGCACACGGTCTAACTCCGGAACAGGTCGTAGCCATTGCTTCCCATGATGGCGG
CAAACAGGCGCTAGAGACAGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGC
CTCACCCCGGAGCAGGTCGTTGCCATCGCCAGTAATATCGGCGGAAAGCAAGCTCTCG
AAACAGTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCA
GGTAGTGGCAATCGCATCTCACGATGGAGGTAAACAAGCACTCGAGACTGTCCAAAG
ATTGTTACCCGTACTATGCCAAGCGCATGGTTTAACCCCAGAGCAAGTTGTGGCTATT
GCATCTAACGGCGGTGGCAAACAAGCCTTGGAGACAGTGCAACGATTACTGCCTGTCT
TATGTCAGGCCCATGGCCTTACTCCTGAGCAAGTCGTAGCTATCGCCAGCAACATAGG
TGGGAAACAGGCCCTGGAAACCGTACAACGTCTCCTCCCAGTACTTTGTCAAGCACAC
GGGTTGACACCGGAACAAGTGGTGGCGATTGCGTCCAACGGCGGAGGCAAGCAGGCA
CTGGAGACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAG
AGCAGGTGGTAGCAATAGCGTCGAACATCGGTGGTAAGCAAGCGCTTGAAACGGTCC
AGCGTCTTCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACCAGAACAAGTGGTTGC
TATTGCTAGTAACAACGGTGGAAAGCAGGCCCTCGAGACGGTGCAGAGGTTACTTCCC
GTCCTCTGTCAAGCGCACGGCCTCACTCCAGAGCAAGTGGTTGCGATCGCTTCAAACA
ATGGTGGAAGACCTGCCCTGGAA, or a sequence having at least 90% sequence identity to the TALE sequence, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
58. The method of claim 29 comprising introducing into the cell a virus including a first foreign nucleic acid encoding the TALEN
having a TALE sequence CTAACCCCTGAACAGGTAGTCGCTATAGCTTCAAATATCGGGGGCAAGCAAGC
ACTTGAGACCGTTCAACGACTCCTGCCAGTGCTCTGCCAAGCCCATGGATTGACTCCG
GAGCAAGTCGTCGCGATCGCGAGCAACGGCGGGGGGAAGCAGGCGCTGGAAACTGTT
CAGAGACTGCTGCCTGTACTTTGTCAGGCGCATGGTCTCACCCCCGAACAGGTTGTCG
CAATAGCAAGTAATATAGGCGGTAAGCAAGCCCTAGAGACTGTGCAACGCCTGCTCC
CCGTGCTGTGTCAGGCTCACGGTCTGACACCTGAACAAGTTGTCGCGATAGCCAGTCA
CGACGGGGGAAAACAAGCTCTAGAAACGGTTCAAAGGTTGTTGCCCGTTCTGTGCCAA
GCACATGGGTTAACACCCGAACAAGTAGTAGCGATAGCGTCAAATAACGGGGGTAAA
CAGGCTTTGGAGACGGTACAGCGGTTATTGCCGGTCCTCTGCCAGGCCCACGGACTTA

CGCCAGAACAGGTGGTTGCAATTGCCTCCAACATCGGCGGGAAACAAGCGTTGGAAA
CTGTGCAGAGACTCCTTCCTGTTTTGTGTCAAGCCCACGGCTTGACGCCTGAGCAGGTT
GTGGCCATCGCTAGCCACGACGGAGGGAAGCAGGCTCTTGAAACCGTACAGCGACTT
CTCCCAGTTTTGTGCCAAGCTCACGGGCTAACCCCCGAGCAAGTAGTTGCCATAGCAA
GCAACGGAGGAGGAAAACAGGCATTAGAAACAGTTCAGCGCTTGCTCCCGGTACTCT
GTCAGGCACACGGTCTAACTCCGGAACAGGTCGTAGCCATTGCTTCCCATGATGGCGG
CAAACAGGCGCTAGAGACAGTCCAGAGGCTCTTGCCTGTGTTATGCCAGGCACATGGC
CTCACCCCGGAGCAGGTCGTTGCCATCGCCAGTAATATCGGCGGAAAGCAAGCTCTCG
AAACAGTACAACGGCTGTTGCCAGTCCTATGTCAAGCTCATGGACTGACGCCCGAGCA
GGTAGTGGCAATCGCATCTCACGATGGAGGTAAACAAGCACTCGAGACTGTCCAAAG
ATTGTTACCCGTACTATGCCAAGCGCATGGTTTAACCCCAGAGCAAGTTGTGGCTATT
GCATCTAACGGCGGTGGCAAACAAGCCTTGGAGACAGTGCAACGATTACTGCCTGTCT
TATGTCAGGCCCATGGCCTTACTCCTGAGCAAGTCGTAGCTATCGCCAGCAACATAGG
TGGGAAACAGGCCCTGGAAACCGTACAACGTCTCCTCCCAGTACTTTGTCAAGCACAC
GGGTTGACACCGGAACAAGTGGTGGCGATTGCGTCCAACGGCGGAGGCAAGCAGGCA
CTGGAGACCGTCCAACGGCTTCTTCCGGTTCTTTGCCAGGCTCATGGGCTCACGCCAG
AGCAGGTGGTAGCAATAGCGTCGAACATCGGTGGTAAGCAAGCGCTTGAAACGGTCC
AGCGTCTTCTGCCGGTGTTGTGCCAGGCGCACGGACTCACACCAGAACAAGTGGTTGC
TATTGCTAGTAACAACGGTGGAAAGCAGGCCCTCGAGACGGTGCAGAGGTTACTTCCC
GTCCTCTGTCAAGCGCACGGCCTCACTCCAGAGCAAGTGGTTGCGATCGCTTCAAACA
ATGGTGGAAGACCTGCCCTGGAA, or a sequence having at least 90% sequence identity to the TALE sequence, wherein the TALEN is expressed, and wherein the TALEN cleaves the target DNA to produce altered DNA in the cell.
59. A virus including a nucleic acid sequence encoding a TALEN lacking repeat sequences 100 bp or longer.
60. The virus of claim 59 wherein the TALEN lacks repeat sequences 90 bp or longer.
61. The virus of claim 59 wherein the TALEN lacks repeat sequences 80 bp or longer.
62. The virus of claim 59 wherein the TALEN lacks repeat sequences 70 bp or longer.
63. The virus of claim 59 wherein the TALEN lacks repeat sequences 60 bp or longer.

64. The virus of claim 59 wherein the TALEN lacks repeat sequences 50 bp or longer.
65. The virus of claim 59 wherein the TALEN lacks repeat sequences 40 bp or longer.
66. The virus of claim 59 wherein the TALEN lacks repeat sequences 30 bp or longer.
67. The virus of claim 59 wherein the TALEN lacks repeat sequences 20 bp or longer.
68. The virus of claim 59 wherein the TALEN lacks repeat sequences 19 bp or longer.
69. The virus of claim 59 wherein the TALEN lacks repeat sequences 18 bp or longer.
70. The virus of claim 59 wherein the TALEN lacks repeat sequences 17 bp or longer.
71. The virus of claim 59 wherein the TALEN lacks repeat sequences 16 bp or longer.
72. The virus of claim 59 wherein the TALEN lacks repeat sequences 15 bp or longer.
73. The virus of claim 59 wherein the TALEN lacks repeat sequences 14 bp or longer.
74. The virus of claim 59 wherein the TALEN lacks repeat sequences 13 bp or longer.
75. The virus of claim 59 wherein the TALEN lacks repeat sequences 12 bp or longer.
76. The virus of claim 59 wherein the TALEN lacks repeat sequences 11 bp or longer.
77. The virus of claim 59 wherein the TALEN lacks repeat sequences 10 bp or longer.
78. The virus of claim 59 being a lentivirus.
79. A cell including a nucleic acid sequence encoding a TALEN lacking repeat sequences 100 bp or longer.
80. The cell of claim 70 wherein the TALEN lacks repeat sequences 90 bp or longer.
81. The cell of claim 70 wherein the TALEN lacks repeat sequences 80 bp or longer.

82. The cell of claim 70 wherein the TALEN lacks repeat sequences 70 bp or longer.
83. The cell of claim 70 wherein the TALEN lacks repeat sequences 60 bp or longer.
84. The cell of claim 70 wherein the TALEN lacks repeat sequences 50 bp or longer.
85. The cell of claim 70 wherein the TALEN lacks repeat sequences 40 bp or longer.
86. The cell of claim 70 wherein the TALEN lacks repeat sequences 30 bp or longer.
87. The cell of claim 70 wherein the TALEN lacks repeat sequences 20 bp or longer.
88. The cell of claim 70 wherein the TALEN lacks repeat sequences 19 bp or longer.
89. The cell of claim 70 wherein the TALEN lacks repeat sequences 18 bp or longer.
90. The cell of claim 70 wherein the TALEN lacks repeat sequences 17 bp or longer.
91. The cell of claim 70 wherein the TALEN lacks repeat sequences 16 bp or longer.
92. The cell of claim 70 wherein the TALEN lacks repeat sequences 15 bp or longer.
93. The cell of claim 70 wherein the TALEN lacks repeat sequences 14 bp or longer.
94. The cell of claim 70 wherein the TALEN lacks repeat sequences 13 bp or longer.
95. The cell of claim 70 wherein the TALEN lacks repeat sequences 12 bp or longer.
96. The cell of claim 70 wherein the TALEN lacks repeat sequences 11 bp or longer.
97. The cell of claim 70 wherein the TALEN lacks repeat sequences 10 bp or longer 98. The cell of claim 70 being a eukaryotic cell.
99. The cell of claim 70 being a yeast cell, a plant cell or an animal cell.

100. The cell of claim 70 being a somatic cell.
101. The cell of claim 70 being a stem cell.
102. The cell of claim 70 being a human stem cell.
103. A method of making a TALE comprising combining an endonuclease, a DNA polymerase, a DNA ligase, an exonuclease, a plurality of nucleic acid dimer blocks encoding repeat variable diresidue domains and a TALE-N/TF
backbone vector including an endonuclease cutting site, activating the endonuclease to cut the TALE-N/TF backbone vector at the endonuclease cutting site to produce a first end and a second end, activating the exonuclease to create a 3' and a 5' overhang on the TALE-N/TF
backbone vector and the plurality of nucleic acid dimer blocks and to anneal the TALE-N/TF backbone vector and the plurality of nucleic acid dimer blocks in a desired order, activating the DNA polymerase and the DNA ligase to connect the TALE-N/TF
backbone vector and the plurality of nucleic acid dimer blocks.
104. A method of altering target DNA in a stem cell expressing an enzyme that forms a co-localization complex with RNA complementary to the target DNA and that cleaves the target DNA
in a site specific manner comprising (a) introducing into the stem cell a first foreign nucleic acid encoding an RNA
complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA, introducing into the stem cell a second foreign nucleic acid encoding a donor nucleic acid sequence, wherein the RNA and the donor nucleic acid sequences are expressed, wherein the RNA and the enzyme co-localize to the target DNA, the enzyme cleaves the target DNA and the donor nucleic acid is inserted into the target DNA to produce altered DNA in the stem cell.
105. The method of claim 104 wherein the enzyme is an RNA-guided DNA binding protein.
106. The method of claim 104 wherein the enzyme is an RNA-guided DNA binding protein of a Type II CRISPR system.

107. The method of claim 104 wherein the enzyme is Cas9.
108. The method of claim 104 wherein the RNA is between about 10 to about 500 nucleotides.
109. The method of claim 104 wherein the RNA is between about 20 to about 100 nucleotides.
110. The method of claim 104 wherein the RNA is a guide RNA.
111. The method of claim 104 wherein the RNA is a tracrRNA-crRNA fusion.
112. The method of claim 104 wherein the DNA is genomic DNA, mitochondrial DNA, viral DNA, or exogenous DNA.
113. The method of claim 104 wherein the donor nucleic acid sequence is inserted by recombination.
114. The method of claim 104 wherein the donor nucleic acid sequence is inserted by homologous recombination.
115. The method of claim 104 wherein the donor nucleic acid sequence is inserted by nonhomologous end joining.
116. The method of claim 104 wherein the RNA and the donor nucleic acid sequences are present on one or more plasmids.
117. The method of claim 104 further comprising repeating step (a) multiple times to produce multiple alterations to the DNA in the cell.
118. The method of claim 104 wherein after producing altered DNA in a stem cell, a nucleic acid encoding the enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner is removed from the stem cell genome.
119. The method of claim 104 wherein the RNA and the donor nucleic acid sequences are expressed as a bound nucleic acid sequence.

120. A stem cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA complementary to target DNA and that cleaves the target DNA in a site specific manner.
121. The stem cell of claim 120 further including a second foreign nucleic acid encoding for an RNA complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA.
122. The stem cell of claim 121 further including a third foreign nucleic acid encoding a donor nucleic acid sequence.
123. The stem cell of claim 120 further including an inducible promoter for promoting expression of the enzyme.
124. The stem cell of claim 120 wherein the first foreign nucleic acid is removable from genomic DNA of the cell using a transposase.
125. The stem cell of claim 120 wherein the enzyme is an RNA-guided DNA
binding protein.
126. The stem cell of claim 120 wherein the enzyme is an RNA-guided DNA
binding protein of a Type II CRISPR system.
127. The stem cell of claim 120 wherein the enzyme is Cas9.
128. The stem cell of claim 120 wherein the RNA is between about 10 to about 500 nucleotides.
129. The stem cell of claim 120 wherein the RNA is between about 20 to about 100 nucleotides.
130. The stem cell of claim 120 wherein the RNA is a guide RNA.
131. The stem cell of claim 120 wherein the RNA is a tracrRNA-crRNA fusion.
132. The stem cell of claim 120 wherein the target DNA is genomic DNA, mitochondrial DNA, viral DNA, or exogenous DNA.

133. A cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA complementary to target DNA and that cleaves the target DNA in a site specific manner and including an inducible promoter for promoting expression of the enzyme.
134. The cell of claim 133 further including a second foreign nucleic acid encoding for an RNA
complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA.
135. The stem cell of claim 134 further including a third foreign nucleic acid encoding a donor nucleic acid sequence.
136. A cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA complementary to target DNA and that cleaves the target DNA in a site specific manner, wherein the first foreign nucleic acid is removable from genomic DNA of the cell using a transposase.
137. The cell of claim 136 further including a second foreign nucleic acid encoding for an RNA
complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA.
138. The stem cell of claim 137 further including a third foreign nucleic acid encoding a donor nucleic acid sequence.
139. A cell including a first foreign nucleic acid encoding for an enzyme that forms a co-localization complex with RNA complementary to target DNA and that cleaves the target DNA in a site specific manner, wherein the first foreign nucleic acid is reversibly inserted into genomic DNA of the cell.
140. The cell of claim 139 further including a second foreign nucleic acid encoding for an RNA
complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA.
141. The stem cell of claim 140 further including a third foreign nucleic acid encoding a donor nucleic acid sequence.

142. A method of altering target DNA in a cell expressing an enzyme that forms a co-localization complex with RNA complementary to the target DNA and that cleaves the target DNA
in a site specific manner comprising (a) introducing into the cell a first foreign nucleic acid encoding a donor nucleic acid sequence, introducing into the cell from media surrounding the cell an RNA complementary to the target DNA and which guides the enzyme to the target DNA, wherein the RNA and the enzyme are members of a co-localization complex for the target DNA, wherein the donor nucleic acid sequence is expressed, wherein the RNA and the enzyme co-localize to the target DNA, the enzyme cleaves the target DNA and the donor nucleic acid is inserted into the target DNA to produce altered DNA in the cell.
143. The method of claim 142 wherein the RNA includes a 5' Cap structure.
144. The method of claim 142 wherein the RNA lacks phosphate groups.
145. The method of claim 142 wherein the enzyme is an RNA-guided DNA binding protein.
146. The method of claim 142 wherein the enzyme is an RNA-guided DNA binding protein of a Type II CRISPR system.
147. The method of claim 142 wherein the enzyme is Cas9.
148. The method of claim 142 wherein the RNA is between about 10 to about 500 nucleotides.
149. The method of claim 142 wherein the RNA is between about 20 to about 100 nucleotides.
150. The method of claim 142 wherein the RNA is a guide RNA.
151. The method of claim 142 wherein the RNA is a tracrRNA-crRNA fusion.
152. The method of claim 142 wherein the DNA is genomic DNA, mitochondrial DNA, viral DNA, or exogenous DNA.

153. The method of claim 142 wherein the donor nucleic acid sequence is inserted by recombination.
154. The method of claim 142 wherein the donor nucleic acid sequence is inserted by homologous recombination.
155. The method of claim 142 wherein the donor nucleic acid sequence is inserted by nonhomologous end joining.
156. The method of claim 142 further comprising repeating step (a) multiple times to produce multiple alterations to the DNA in the cell.
157. The method of claim 142 wherein after producing altered DNA in a cell, a nucleic acid encoding the enzyme that forms a co-localization complex with RNA
complementary to the target DNA and that cleaves the target DNA in a site specific manner is removed from the cell genome.
158. The method of claim 142 wherein the RNA and the donor nucleic acid sequences are expressed as a bound nucleic acid sequence.
CA2918540A 2013-07-26 2014-07-25 Methods of altering target dna in cells, and corresponding cells Pending CA2918540A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361858866P 2013-07-26 2013-07-26
US61/858,866 2013-07-26
PCT/US2014/048140 WO2015013583A2 (en) 2013-07-26 2014-07-25 Genome engineering

Publications (1)

Publication Number Publication Date
CA2918540A1 true CA2918540A1 (en) 2015-01-29

Family

ID=52390824

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2918540A Pending CA2918540A1 (en) 2013-07-26 2014-07-25 Methods of altering target dna in cells, and corresponding cells

Country Status (14)

Country Link
US (4) US10563225B2 (en)
EP (4) EP3483277B1 (en)
JP (4) JP6739335B2 (en)
KR (2) KR20160035017A (en)
CN (3) CN105473773B (en)
AU (4) AU2014293015B2 (en)
BR (1) BR112016001721B1 (en)
CA (1) CA2918540A1 (en)
ES (1) ES2715666T3 (en)
HK (1) HK1218940A1 (en)
IL (2) IL243560B (en)
RU (2) RU2688462C2 (en)
SG (3) SG11201600217QA (en)
WO (1) WO2015013583A2 (en)

Families Citing this family (111)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3613852A3 (en) 2011-07-22 2020-04-22 President and Fellows of Harvard College Evaluation and improvement of nuclease cleavage specificity
WO2013163394A1 (en) 2012-04-25 2013-10-31 Regeneron Pharmaceuticals, Inc. Nuclease-mediated targeting with large targeting vectors
EP3553174A1 (en) 2012-12-17 2019-10-16 President and Fellows of Harvard College Rna-guided human genome engineering
CN105518146B (en) 2013-04-04 2022-07-15 哈佛学院校长同事会 Therapeutic uses of genome editing with CRISPR/Cas systems
CN111500630A (en) 2013-04-16 2020-08-07 瑞泽恩制药公司 Targeted modification of rat genome
JP2016528890A (en) 2013-07-09 2016-09-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Therapeutic use of genome editing using the CRISPR / Cas system
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
ES2681622T3 (en) 2013-09-18 2018-09-14 Kymab Limited Methods, cells and organisms
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9546384B2 (en) 2013-12-11 2017-01-17 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse genome
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
MX2016010285A (en) 2014-02-11 2017-01-11 Univ Colorado Regents Crispr enabled multiplexed genome engineering.
WO2015134812A1 (en) 2014-03-05 2015-09-11 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating usher syndrome and retinitis pigmentosa
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
DK3116997T3 (en) 2014-03-10 2019-08-19 Editas Medicine Inc CRISPR / CAS-RELATED PROCEDURES AND COMPOSITIONS TO TREAT LEVER'S CONGENITAL AMAUROSIS 10 (LCA10)
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
WO2015191693A2 (en) 2014-06-10 2015-12-17 Massachusetts Institute Of Technology Method for gene editing
AU2015298571B2 (en) 2014-07-30 2020-09-03 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
WO2016061374A1 (en) 2014-10-15 2016-04-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating or maintaining pluripotent cells
WO2016065364A1 (en) * 2014-10-24 2016-04-28 Life Technologies Corporation Compositions and methods for enhancing homologous recombination
CA2963820A1 (en) 2014-11-07 2016-05-12 Editas Medicine, Inc. Methods for improving crispr/cas-mediated genome-editing
PT3221457T (en) 2014-11-21 2019-06-27 Regeneron Pharma Methods and compositions for targeted genetic modification using paired guide rnas
JP7068821B2 (en) 2014-12-03 2022-05-17 アジレント・テクノロジーズ・インク Guide RNA with chemical modification
KR102656470B1 (en) 2014-12-10 2024-04-09 리전츠 오브 더 유니버스티 오브 미네소타 Genetically modified cells, tissues, and organs for treating disease
WO2016150855A1 (en) * 2015-03-20 2016-09-29 Danmarks Tekniske Universitet Crispr/cas9 based engineering of actinomycetal genomes
US20180112213A1 (en) * 2015-03-25 2018-04-26 Editas Medicine, Inc. Crispr/cas-related methods, compositions and components
AU2016246450B2 (en) 2015-04-06 2022-03-17 Agilent Technologies, Inc. Chemically modified guide RNAs for CRISPR/Cas-mediated gene regulation
AU2016253150B2 (en) 2015-04-24 2022-04-21 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
EP3294896A1 (en) * 2015-05-11 2018-03-21 Editas Medicine, Inc. Optimized crispr/cas9 systems and methods for gene editing in stem cells
EP3303585A4 (en) 2015-06-03 2018-10-31 Board of Regents of the University of Nebraska Dna editing using single-stranded dna
AU2016276702B2 (en) 2015-06-09 2022-07-28 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for improving transplantation
US9790490B2 (en) 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems
CN108290933A (en) 2015-06-18 2018-07-17 布罗德研究所有限公司 Reduce the CRISPR enzyme mutants of undershooting-effect
AU2016279062A1 (en) 2015-06-18 2019-03-28 Omar O. Abudayyeh Novel CRISPR enzymes and systems
EP4339287A3 (en) 2015-07-31 2024-05-22 Regents Of The University Of Minnesota Modified cells and methods of therapy
US10526590B2 (en) 2015-08-31 2020-01-07 Agilent Technologies, Inc. Compounds and methods for CRISPR/Cas-based genome editing by homologous recombination
AU2016326711B2 (en) 2015-09-24 2022-11-03 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/Cas-mediated genome editing
CN108513575A (en) 2015-10-23 2018-09-07 哈佛大学的校长及成员们 Nucleobase editing machine and application thereof
CN115806940A (en) * 2015-11-04 2023-03-17 菲特治疗公司 Genome engineering of pluripotent cells
KR101885901B1 (en) * 2015-11-13 2018-08-07 기초과학연구원 RGEN RNP delivery method using 5'-phosphate removed RNA
EP3377086B1 (en) 2015-11-19 2024-05-01 The Brigham and Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
EP3383409A4 (en) * 2015-12-02 2019-10-02 The Regents of The University of California Compositions and methods for modifying a target nucleic acid
US11441146B2 (en) * 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
CN106957856B (en) * 2016-01-12 2020-07-28 中国科学院广州生物医药与健康研究院 Reconstructed egg of hairless model pig, construction method of reconstructed egg and construction method of model pig
EP3433363A1 (en) 2016-03-25 2019-01-30 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
KR102670601B1 (en) 2016-04-19 2024-05-29 더 브로드 인스티튜트, 인코퍼레이티드 The novel CRISPR enzyme and system
US11286478B2 (en) 2016-04-19 2022-03-29 The Broad Institute, Inc. Cpf1 complexes with reduced indel activity
EP4166661A1 (en) * 2016-04-29 2023-04-19 BASF Plant Science Company GmbH Fused donor - guide nucleic acid and methods for modification of target nucleic acids
BR112018074494A2 (en) * 2016-06-01 2019-03-19 Kws Saat Se & Co Kgaa hybrid nucleic acid sequences for genomic engineering
US10767175B2 (en) 2016-06-08 2020-09-08 Agilent Technologies, Inc. High specificity genome editing using chemically modified guide RNAs
AU2017280353B2 (en) 2016-06-24 2021-11-11 Inscripta, Inc. Methods for generating barcoded combinatorial libraries
MX2019000088A (en) 2016-06-27 2019-08-29 Broad Inst Inc Compositions and methods for detecting and treating diabetes.
US20180004537A1 (en) * 2016-07-01 2018-01-04 Microsoft Technology Licensing, Llc Molecular State Machines
US11359234B2 (en) 2016-07-01 2022-06-14 Microsoft Technology Licensing, Llc Barcoding sequences for identification of gene expression
BR112019001887A2 (en) 2016-08-02 2019-07-09 Editas Medicine Inc compositions and methods for treating cep290-associated disease
WO2018027078A1 (en) 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
CA3033327A1 (en) 2016-08-09 2018-02-15 President And Fellows Of Harvard College Programmable cas9-recombinase fusion proteins and uses thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
WO2018049168A1 (en) 2016-09-09 2018-03-15 The Board Of Trustees Of The Leland Stanford Junior University High-throughput precision genome editing
KR20240007715A (en) 2016-10-14 2024-01-16 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Aav delivery of nucleobase editors
EP3529359B1 (en) 2016-10-18 2023-12-13 Regents of the University of Minnesota Tumor infiltrating lymphocytes for use in therapy
WO2018094356A2 (en) * 2016-11-18 2018-05-24 Genedit Inc. Compositions and methods for target nucleic acid modification
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
WO2018152120A1 (en) * 2017-02-14 2018-08-23 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Methods of engineering human induced pluripotent stem cells to produce liver tissue
EP3592853A1 (en) 2017-03-09 2020-01-15 President and Fellows of Harvard College Suppression of pain by gene editing
JP2020510439A (en) 2017-03-10 2020-04-09 プレジデント アンド フェローズ オブ ハーバード カレッジ Base-editing factor from cytosine to guanine
WO2018170184A1 (en) 2017-03-14 2018-09-20 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
SG11201908658TA (en) 2017-03-23 2019-10-30 Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
US20210115407A1 (en) 2017-04-12 2021-04-22 The Broad Institute, Inc. Respiratory and sweat gland ionocytes
WO2018195486A1 (en) 2017-04-21 2018-10-25 The Broad Institute, Inc. Targeted delivery to beta cells
WO2018201086A1 (en) 2017-04-28 2018-11-01 Editas Medicine, Inc. Methods and systems for analyzing guide rna molecules
US11591601B2 (en) 2017-05-05 2023-02-28 The Broad Institute, Inc. Methods for identification and modification of lncRNA associated with target genotypes and phenotypes
EP3622070A2 (en) 2017-05-10 2020-03-18 Editas Medicine, Inc. Crispr/rna-guided nuclease systems and methods
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2018227114A1 (en) 2017-06-09 2018-12-13 Editas Medicine, Inc. Engineered cas9 nucleases
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
JP2020530307A (en) 2017-06-30 2020-10-22 インティマ・バイオサイエンス,インコーポレーテッド Adeno-associated virus vector for gene therapy
EP3652312A1 (en) 2017-07-14 2020-05-20 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
SG11201912235PA (en) 2017-07-31 2020-01-30 Regeneron Pharma Cas-transgenic mouse embryonic stem cells and mice and uses thereof
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US20200270632A1 (en) * 2017-09-15 2020-08-27 The Board Of Trustees Of The Leland Stanford Junior University Multiplex production and barcoding of genetically engineered cells
CN111757937A (en) 2017-10-16 2020-10-09 布罗德研究所股份有限公司 Use of adenosine base editor
US11268092B2 (en) 2018-01-12 2022-03-08 GenEdit, Inc. Structure-engineered guide RNA
CA3096274A1 (en) 2018-04-06 2019-10-10 Children's Medical Center Corporation Compositions and methods for somatic cell reprogramming and modulating imprinting
EP3794130A4 (en) 2018-05-16 2022-07-27 Synthego Corporation Methods and systems for guide rna design and use
US20210246505A1 (en) * 2018-06-21 2021-08-12 Memorial Sloan Kettering Cancer Center Compositions & methods for monitoring dna repair
SG11202102068TA (en) 2018-07-31 2021-03-30 Broad Inst Inc Novel crispr enzymes and systems
MX2021006783A (en) 2018-12-10 2021-07-15 Amgen Inc Mutated piggybac transposase.
DE112020001342T5 (en) 2019-03-19 2022-01-13 President and Fellows of Harvard College Methods and compositions for editing nucleotide sequences
US20230025039A1 (en) 2019-09-20 2023-01-26 The Broad Institute, Inc. Novel type vi crispr enzymes and systems
WO2021224633A1 (en) 2020-05-06 2021-11-11 Orchard Therapeutics (Europe) Limited Treatment for neurodegenerative diseases
EP4146804A1 (en) 2020-05-08 2023-03-15 The Broad Institute Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
AU2021364781A1 (en) 2020-10-21 2023-06-01 Massachusetts Institute Of Technology Systems, methods, and compositions for site-specific genetic engineering using programmable addition via site-specific targeting elements (paste)
TW202233660A (en) 2020-10-30 2022-09-01 美商安進公司 Overexpression of insulin-like growth factor receptor mutants to modulate igf supplementation
WO2023039586A1 (en) 2021-09-10 2023-03-16 Agilent Technologies, Inc. Guide rnas with chemical modification for prime editing
IL312452A (en) 2021-11-01 2024-06-01 Tome Biosciences Inc Single construct platform for simultaneous delivery of gene editing machinery and nucleic acid cargo
WO2023122764A1 (en) 2021-12-22 2023-06-29 Tome Biosciences, Inc. Co-delivery of a gene editor construct and a donor template
WO2023205744A1 (en) 2022-04-20 2023-10-26 Tome Biosciences, Inc. Programmable gene insertion compositions
WO2023225670A2 (en) 2022-05-20 2023-11-23 Tome Biosciences, Inc. Ex vivo programmable gene insertion
WO2024020587A2 (en) 2022-07-22 2024-01-25 Tome Biosciences, Inc. Pleiopluripotent stem cell programmable gene insertion
WO2024105633A1 (en) 2022-11-18 2024-05-23 Kyoto Prefectural Public University Corporation Compositions for mitophagy induction and uses thereof
WO2024138194A1 (en) 2022-12-22 2024-06-27 Tome Biosciences, Inc. Platforms, compositions, and methods for in vivo programmable gene insertion

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5641670A (en) * 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6689558B2 (en) 2000-02-08 2004-02-10 Sangamo Biosciences, Inc. Cells for drug discovery
AU2001261358B2 (en) * 2000-05-10 2006-07-13 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Resistance to acetohydroxyacid synthase-inhibiting herbicides
BRPI0307383B1 (en) 2002-01-23 2019-12-31 The Univ Of Utah Research Foundation directed genetic recombination method in host plant cell
JP2006508675A (en) 2002-12-09 2006-03-16 トゥールゲン・インコーポレイテッド Regulatory zinc finger protein
JP2007521823A (en) 2004-02-13 2007-08-09 ノボザイムス アクティーゼルスカブ Protease variant
DE102005062662A1 (en) 2005-12-23 2007-06-28 Basf Ag Preparation of optically active alkanol compound, useful for the treatment of depression or incontinence, comprises incubating an enzyme with a polypeptide sequence in a medium containing alkanone compound and isolating
DK2126130T3 (en) 2007-03-02 2015-06-29 Dupont Nutrition Biosci Aps CULTURES WITH IMPROVED phage resistance
WO2010011961A2 (en) 2008-07-25 2010-01-28 University Of Georgia Research Foundation, Inc. Prokaryotic rnai-like system and methods of use
US20100076057A1 (en) 2008-09-23 2010-03-25 Northwestern University TARGET DNA INTERFERENCE WITH crRNA
WO2010054108A2 (en) 2008-11-06 2010-05-14 University Of Georgia Research Foundation, Inc. Cas6 polypeptides and methods of use
PH12012500097A1 (en) * 2009-07-21 2011-01-27 Shanghai Inst Organic Chem Potent small molecule inhibitors of autophagy, and methods of use thereof
US8586526B2 (en) 2010-05-17 2013-11-19 Sangamo Biosciences, Inc. DNA-binding proteins and uses thereof
CN102770539B (en) 2009-12-10 2016-08-03 明尼苏达大学董事会 The DNA modification of TAL effector mediation
NO332016B1 (en) 2009-12-29 2012-05-21 Stiftelsen Sintef Sample processing cartridge and method for processing and / or analyzing a sample under centrifugal force
GB201000591D0 (en) 2010-01-14 2010-03-03 Ucb Pharma Sa Bacterial hoist strain
BR112012018631A8 (en) * 2010-01-28 2017-12-19 President And Fellows Of Harvard Colege compositions and methods for enhancing proteasome activity
US10087431B2 (en) 2010-03-10 2018-10-02 The Regents Of The University Of California Methods of generating nucleic acid fragments
EP2569425B1 (en) 2010-05-10 2016-07-06 The Regents of The University of California Endoribonuclease compositions and methods of use thereof
US20140113376A1 (en) 2011-06-01 2014-04-24 Rotem Sorek Compositions and methods for downregulating prokaryotic genes
JP6072788B2 (en) 2011-07-25 2017-02-01 サンガモ バイオサイエンシーズ, インコーポレイテッド Methods and compositions for altering the cystic fibrosis membrane conductance regulator (CFTR) gene
US8450107B1 (en) 2011-11-30 2013-05-28 The Broad Institute Inc. Nucleotide-specific recognition sequences for designer TAL effectors
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
DK2836226T3 (en) 2012-02-24 2017-09-18 Hutchinson Fred Cancer Res COMPOSITIONS AND PROCEDURES FOR TREATING HEMOGLOBINOPATHY
AU2013225950B2 (en) 2012-02-29 2018-02-15 Sangamo Therapeutics, Inc. Methods and compositions for treating huntington's disease
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
WO2013141680A1 (en) 2012-03-20 2013-09-26 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
DK3401400T3 (en) 2012-05-25 2019-06-03 Univ California METHODS AND COMPOSITIONS FOR RNA CONTROLLED TARGET DNA MODIFICATION AND FOR RNA-CONTROLLED TRANCE CRITICAL MODULATION
EP2880171B1 (en) 2012-08-03 2018-10-03 The Regents of The University of California Methods and compositions for controlling gene expression by rna processing
CN110643600A (en) 2012-10-23 2020-01-03 基因工具股份有限公司 System for cutting target DNA and use thereof
JP6620018B2 (en) 2012-12-06 2019-12-11 シグマ−アルドリッチ・カンパニー・リミテッド・ライアビリティ・カンパニーSigma−Aldrich Co., LLC Genomic modification and control based on CRISPR
MX2015007550A (en) 2012-12-12 2017-02-02 Broad Inst Inc Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications.
PT2898075E (en) 2012-12-12 2016-06-16 Harvard College Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
EP2971167B1 (en) 2013-03-14 2019-07-31 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9234213B2 (en) 2013-03-15 2016-01-12 System Biosciences, Llc Compositions and methods directed to CRISPR/Cas genomic engineering systems
US20140349400A1 (en) 2013-03-15 2014-11-27 Massachusetts Institute Of Technology Programmable Modification of DNA
CN105518146B (en) 2013-04-04 2022-07-15 哈佛学院校长同事会 Therapeutic uses of genome editing with CRISPR/Cas systems
WO2014190181A1 (en) 2013-05-22 2014-11-27 Northwestern University Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
US11685935B2 (en) 2013-05-29 2023-06-27 Cellectis Compact scaffold of Cas9 in the type II CRISPR system
WO2014198911A1 (en) 2013-06-14 2014-12-18 Cellectis Improved polynucleotide sequences encoding tale repeats
EP4245853A3 (en) 2013-06-17 2023-10-18 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
CA2933134A1 (en) 2013-12-13 2015-06-18 Cellectis Cas9 nuclease platform for microalgae genome engineering

Also Published As

Publication number Publication date
EP3916099A1 (en) 2021-12-01
JP7419328B2 (en) 2024-01-22
HK1218940A1 (en) 2017-03-17
EP3483277A1 (en) 2019-05-15
EP3024964A4 (en) 2017-01-18
RU2764757C2 (en) 2022-01-21
US20220235382A1 (en) 2022-07-28
KR20230096134A (en) 2023-06-29
RU2019114217A (en) 2019-08-23
JP6739335B2 (en) 2020-08-12
EP4234703A2 (en) 2023-08-30
AU2021202581A1 (en) 2021-05-27
IL243560B (en) 2020-02-27
US20150031132A1 (en) 2015-01-29
AU2014293015B2 (en) 2019-01-24
JP2020110152A (en) 2020-07-27
AU2014293015A1 (en) 2016-02-11
NZ716606A (en) 2022-03-25
BR112016001721B1 (en) 2023-10-03
RU2016106649A3 (en) 2018-08-10
US20160369301A1 (en) 2016-12-22
CN118028379A (en) 2024-05-14
WO2015013583A3 (en) 2015-04-23
JP2022033922A (en) 2022-03-02
IL243560A0 (en) 2016-02-29
RU2688462C2 (en) 2019-05-21
CN105473773B (en) 2024-03-08
IL272193B (en) 2022-05-01
JP2016528894A (en) 2016-09-23
US20150031133A1 (en) 2015-01-29
AU2018278911A1 (en) 2019-01-03
EP3916099B1 (en) 2023-04-12
ES2715666T3 (en) 2019-06-05
EP3024964A2 (en) 2016-06-01
RU2019114217A3 (en) 2020-07-16
SG11201600217QA (en) 2016-02-26
US10563225B2 (en) 2020-02-18
RU2016106649A (en) 2017-08-31
AU2018278911B2 (en) 2021-01-28
KR20160035017A (en) 2016-03-30
AU2023248167A1 (en) 2023-12-14
EP3024964B1 (en) 2018-12-19
BR112016001721A2 (en) 2017-09-05
AU2018278911C1 (en) 2021-09-23
WO2015013583A8 (en) 2015-03-05
CN105473773A (en) 2016-04-06
AU2021202581B2 (en) 2023-07-13
WO2015013583A2 (en) 2015-01-29
SG10201913000PA (en) 2020-03-30
EP4234703A3 (en) 2023-09-06
US9914939B2 (en) 2018-03-13
US11306328B2 (en) 2022-04-19
IL272193A (en) 2020-03-31
JP6993063B2 (en) 2022-01-13
EP3483277B1 (en) 2021-07-07
CN111304230A (en) 2020-06-19
JP2024029218A (en) 2024-03-05
SG10201800259WA (en) 2018-02-27

Similar Documents

Publication Publication Date Title
AU2021202581B2 (en) Genome engineering
Yang et al. Optimization of scarless human stem cell genome editing
RU2812848C2 (en) Genome engineering
NZ754903A (en) Genome engineering
NZ754903B2 (en) Genome engineering
NZ754904B2 (en) Genome engineering
NZ716606B2 (en) Genome engineering
NZ754902B2 (en) Genome engineering
BR122022013043B1 (en) COMBINATION COMPRISING A STEM CELL GENETICALLY MODIFIED TO INCLUDE A NUCLEIC ACID ENCODING A Cas9 ENZYME, A GUIDE RNA COMPLEMENTARY TO THE TARGET DNA, AND A DONOR NUCLEIC ACID SEQUENCE

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308

EEER Examination request

Effective date: 20190308