US20130209426A1 - Method for genome modification - Google Patents

Method for genome modification Download PDF

Info

Publication number
US20130209426A1
US20130209426A1 US13/594,053 US201213594053A US2013209426A1 US 20130209426 A1 US20130209426 A1 US 20130209426A1 US 201213594053 A US201213594053 A US 201213594053A US 2013209426 A1 US2013209426 A1 US 2013209426A1
Authority
US
United States
Prior art keywords
cell
genome
cells
nucleic acid
mutation
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.)
Abandoned
Application number
US13/594,053
Inventor
Allan Bradley
Kosuke Yusa
Sheikh Tamir Rashid
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.)
Cambridge Enterprise Ltd
Genome Research Ltd
Original Assignee
Cambridge Enterprise Ltd
Genome Research Ltd
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 Cambridge Enterprise Ltd, Genome Research Ltd filed Critical Cambridge Enterprise Ltd
Assigned to GENOME RESEARCH LIMITED reassignment GENOME RESEARCH LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRADLEY, ALLAN, YUSA, YOSUKE
Assigned to CAMBRIDGE ENTERPRISE LTD reassignment CAMBRIDGE ENTERPRISE LTD ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RASHID, SHEIK TAMIR
Publication of US20130209426A1 publication Critical patent/US20130209426A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • 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
    • 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
    • C12N5/067Hepatocytes
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/16Activin; Inhibin; Mullerian inhibiting substance

Definitions

  • a method of modifying the genome of a cell comprising recombining into the genome of the cell an exogenous nucleic acid, the nucleic acid comprising
  • the method further comprising the steps of:
  • transposase to excise the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence
  • a method of correcting or introducing a mutation in a genome of a cell comprising
  • nucleic acid comprising (i) a modifying sequence which corrects or introduces the mutation and (ii) a selection element flanked by transposon inverted repeats,
  • a cell comprising an exogenous nucleic acid, the exogenous comprising (i) a modifying sequence and
  • a cell comprising a genome which has been modified by insertion of a modifying sequence using a method according to claim 1 .
  • a modified cell comprising a modifying nucleic acid sequence, the cell having been selected for the insertion of the modifying nucleic acid sequence using a selectable element, and wherein the selectable element is then removed from the genome, such that genome of the modified cell is identical to the genome of the original starting cell except for the modifying nucleic acid.
  • a collection of cells such as an organ or tissue, wherein a mutation in the genomes of all, or a proportion, of the cells has been corrected by insertion of a modifying sequence using a method as described above.
  • a method for treating a disease in a patient comprising modifying the genome of a cell using a method as described above to correct a mutation associated with the disease in the cell, and using the cell, or cells derived or differentiated therefrom, in disease treatment.
  • a cell as described above for use in the treatment of disease.
  • FIG. 1 Correction of the G290T mutation in the Tyr gene in mIPSCs.
  • Top line structure of the Tyr gene; red line, 5′ external probe for Southern blot analysis; open arrow, piggyBac transposon carrying a PGK-puro ⁇ tk cassette; P1, P2 and P3, PCR primers; B, BamHI; E, EcoNI. b, c, Southern blot (b) and PCR analyses (c) showing insertion (c/PB) and excision (c/Rev) of the piggyBac transposon.
  • ES mouse ESCs as a control.
  • d, e Sequence analyses revealed correction of the G290T mutation (d) and seamless excision of the piggyBac transposon (e).
  • a The strategy for precise genome modification using ZFNs and the piggyBac transposon.
  • Top line structure of the A1AT gene; blue lines, Southern blot probes; thin and thick boxes, non-coding and coding exons, respectively; open arrow, piggyBac transposon; B, BamHI; A, AflIII.
  • b Sequences of wild-type (Reference), Z, PB, and Rev alleles. Amino acid position 342 (blue), recognition sites for ZFNs (green), piggyBac excision site (red) are shown. Sequence changes in Rev allele from Z allele were indicated by asterisks.
  • FIG. 3 Functional analysis of restored A1AT in c-hIPSCs-derived hepatocyte-like cells.
  • d Endoglycosidase H (E) and peptide:N-glycosidase (P) digestion of A1AT immunoprecipitated from uncorrected (ZZ), corrected (RR) and control (++) hIPSC-derived hepatocyte-like cells (upper panels) and corresponding culture medium (lower panels).
  • e Chymotrypsin ELISA showing that corrected cells (RR) have A1AT enzymatic inhibitory activity that is superior to uncorrected cells (ZZ) and close to adult hepatocytes.
  • f g, Immunofluorescence of transplanted liver sections detecting human albumin (f) and A1AT (g). DNA was counterstained with DAPI.
  • FIG. 4 Southern blot analyses of bi-allelic targeting of the A1AT gene in 3 independent A1ATD-iPSC lines.
  • a The structures of the Z allele and the targeted PB allele. Thin and thick open boxes, non-coding and coding exon, respectively; B, BamHI.
  • b The result obtained with A1ATD-iPSCs from patient B. Both alleles in line B-16 were correctly targeted while line B-17 carried one correctly targeted allele and one abnormal allele.
  • c-e Similar results were obtained with A1ATD-iPSCs generated from patient A (c) and from patient C (d, e). Genomic DNA was digested by BamHI and hybridized with the 5′ external probe. The first lane on each panel is DNA from the parental A1ATD-iPSC line. Correct homozygous clones are highlighted in red.
  • FIG. 6 Pluripotency of corrected iPSCs.
  • a Immunofluorescence showing the expression of pluripotency markers OCT4, SOX2, NANOG and TRA1-60 in corrected iPSCs grown for 20 passages after correction. DNAs were stained with DAPI (lower panels).
  • b Immunofluorescence of in vitro differentiated corrected iPSCs showing maintenance of differentiation capacity into three germ layers. Scale bars, 200 ⁇ m
  • aCGH result of the homozygous clone, C-B6 Top, an ideogram of chromosome 20; middle, genes in the region pointed by red in the chromosome ideogram; bottom, the log2 ratio plot of C-B6.
  • the amplified region is highlighted in red.
  • the log2 ratios in the adjacent region highlighted in gray are used as a normal copy number control for a comparison shown in c.
  • b SNP data showing B allele frequency (x axis on top) and smoothed log R ratio (x axis on bottom) of chromosome 20 of indicated cell lines. The 20q11.21 is highlighted in gray.
  • c Box plots showing log2 ratios of the affected (red) and the adjacent normal (gray) regions.
  • FIG. 8 SNP analyses. a, b, SNP array data showing entire chromosome 14 (a) and 830-kb region around the A1AT (SERPINA1) gene (b). Data for fibroblasts and correctly targeted homozygous clones from each patient are shown. Blue dots represent B allele frequency (left y axis) of each SNP and red lines represent smoothed Log R ratio (right y axis). Note that heterozygosity is maintained in all 6 targeted clones.
  • FIG. 9 Differentiation of corrected iPSCs into hepatocyte-like cells.
  • Corrected iPSC-derived hepatocyte-like cells display functional activity characteristic of primary human hepatocytes including presence of intracellular glycogen storage as shown by periodic acid Schiff staining (d), uptake of DiL-LDL (e), albumin secretion (f) and CytP450 metabolism (g). Scale bar, 100 ⁇ m
  • FIG. 10 Characterization of integration-free A1ATD-iPSC.
  • FIG. 11 Correction of the Z mutation in the integration-free A1ATD-iPSC line B.
  • a Validation of genetic modification by Southern blot analysis. See FIG. 2 for the detail.
  • b Sequencing analysis of gene correction. Note that the Z mutation (A) is corrected to wild-type sequence (G), while the piggyBac excisions produced TTAA as designed.
  • c CGH analysis showing that the 3-G5-A7 line kept the intact genome compared to the paretal fibroblasts.
  • d Immunostaining of 3-G5-A7-derived hepatocyte-like cells, showing that the corrected cells express normal A1AT porteins. Left, all forms of A1AT; middle, polymeric A1AT; right, DNA staining by DAPI. Scale bar, 50 ⁇ m.
  • the exogenous nucleic acid is suitably in a form that can be incorporated into the genome of the cell by homologous recombination and the nucleic acid is provided to the cell under conditions that allow homologous recombination to occur. These conditions are well known in the art.
  • the exogenous nucleic acid is DNA, which may be provided as a circular plasmid or linear nucleic acid.
  • the exogenous nucleic acid to be inserted suitably comprises two regions the same or a very similar sequence to the genome of the cell to be modified, such that homologous recombination between the homologous regions results in insertion of the exogenous nucleic acid into the chromosome, replacing the equivalent genomic sequence.
  • the exogenous nucleic acid to be inserted comprises a selectable marker and the insertion event can therefore be selected for by selection of the selection element under appropriate selection conditions.
  • the efficiency of targeting can be increased using Zinc Finger Nucleases (ZFNs) or Transcription activator like effector nucleases (TALENS) to affect double stranded breaks which can be repaired by, for example, homology directed DNA repair pathways using an exogenous donor plasmid or other exogenous nucleic acid sequence as a template.
  • ZFNs Zinc Finger Nucleases
  • TALENS Transcription activator like effector nucleases
  • the method of the invention also comprises providing to the cell a zinc finger nuclease or Transcription activator like effector nucleases (TALES), or nucleic acid encoding the same, to allow expression of the ZFN or TALES in the cell whose genome is to be modified.
  • TALES Transcription activator like effector nucleases
  • hIPSCs zinc finger nucleases 7 and piggyBac 8,9 technology in hIPSCs can achieve bi-allelic correction of a point mutation (Glu342Lys) in the ⁇ 1 antitrypsin (A1AT, also called SERPINA1) gene that is responsible for ⁇ 1 -antitrypsin deficiency (A1ATD).
  • A1AT also called SERPINA1
  • Genetic correction of hIPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene targeting technology that is currently available and prevents contamination of the host genome with residual non-human sequences.
  • the cell to be modified is a human cell or a non-human animal cell.
  • the cell is a pluripotent cell, such as a totipotent non-human cell, or a pluripotent human or non-human cell.
  • the cell may also be an iPS cell, such as a human iPS cell or non-human iPS cell.
  • An iPS cell may originate from a fibroblast cell, adipose tissue, bone marrow or other suitable somatic cell.
  • iPS cells may be generated using methods well known in the art, such as expression of Yamanaka factors as disclosed in EP1970446 or as disclosed in Wang et at www.pnas.org/cgi/doi/10.1073/pnas.1100893108
  • iPS cells may be derived from individuals with diseases caused by genetic mutation, for example diseases affecting the epithelial, endothelial, or interstitial compartments of the lung, such as cystic fibrosis, a-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease.
  • diseases affecting the epithelial, endothelial, or interstitial compartments of the lung such as cystic fibrosis, a-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease.
  • the cell to be modified is an iPS cell is derived from an individual in whom it is desired to re-transplant the iPS cell, or cells derived or differentiated therefrom.
  • an iPS cell is reprogrammed using an integration-free method, for example as disclosed herein using Sendai virus vectors (ref 25).
  • an integration-free method for example as disclosed herein using Sendai virus vectors (ref 25).
  • the reprogramming of an iPS cell does not leave any residual DNA in the genome of the iPS cell.
  • Another example of suitable methodology includes that disclosed in Somers et al, Stem cells 2010;28:1728-1740.
  • Another example of suitable integration free technology includes providing mRNA or DNA minicircle as disclosed in https://www.stemgent.com/applications/reprogramming.
  • the iPS cell modified by the method of the invention maintains expression of pluripotency markers for at least 10, at least 15, or at least 20 passages and or the ability to differentiate into cells expressing markers of three germ layers.
  • the modifying sequence is the sequence of exogenous nucleic acid which is not the selection element flanked by the transposase inverted repeat sequences. This sequence of the exogenous nucleic acid generally remains in the genome after transposase excision.
  • the modifying sequence may comprise a nucleic acid sequence which is substantially identical to a part of the genome of the cell to be modified but which has an addition, substitution or deletion of nucleotides with respect to that genome.
  • the modifying nucleic acid may be isogenic with the genome with the exception of the addition, substitution or deletion.
  • the modifying nucleic acid may be isogenic with the genome with the exception of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, for example, or even with the exception of up to 50 nucleotides, e.g. up to 100 nucleotides, up to 200 nucleotides, up to 500 nucleotides or up to 1 kb, or 2 kb.
  • the modifying sequence may be a sequence which is only partly isogenic with the genomic DNA, for example at the ends of the nucleic acid sequence to allow for homologous recombination, but may comprise a region of non-isogenic DNA of some length, such as more than 20, 30 ,40 or 50 nucleotides, such as a gene from a species different to that of the cell to be modified.
  • the modification causes a change in function of the cell, or the tissue or organism derivable from development of the cell.
  • the modification need not be associated with a disease phenotype.
  • the modification is correction of a mutation associated with disease, such as correction of a point mutation.
  • a point mutation may be changed to a nucleotide that is not associated with a disease phenotype, for example, changing of the Lys 342 to Glu in the ⁇ 1-antitrypsin (A1AT, also called SERPINA1) gene that is responsible for ⁇ 1-antitrypsin deficiency (A1ATD).
  • A1AT also called SERPINA1
  • A1ATD ⁇ 1-antitrypsin deficiency
  • the modification restores an enzymatic function of the cell.
  • the modification is a correction of one or both alleles of a gene.
  • the modification may be a correction of one or both alleles of a point mutation associated with a disease phenotype.
  • the method can be, for example:
  • a method of correcting a mutation in a genome of a cell comprising
  • the genome of the corrected cell is identical to the genome of the original cell with the only exception being mono or biallelic correction of the mutation.
  • the modifying sequence comprises the wild type nucleic acid sequence that would be found in individuals without disease which is inserted into the genome to replace the genome region comprising the mutation.
  • the modifying sequence can also be used to introduce a mutation into a cell, and thus the invention also relates to a method of correcting or introducing a mutation into a genome of a cell, as disclosed herein.
  • the genome of the corrected cell is identical to the genome of the original cell with the only exception being mono or biallelic correction of the mutation or mono or biallelic introduction of a mutation—such as a point mutation—but wherein, in one aspect, there may be additional mutations in the genome that occur due to the natural mutation processes that occur in during cell division.
  • the invention relates to a method of correcting or introducing a modification into the genome of a cell as disclosed herein, wherein after the transposon mediated excision process the genome of the corrected cell comprises the desired specific modification and may comprise a random mutation(s) but comprises no other nucleic acid sequence alteration, for example, no nucleic acid sequence alteration that might arise from insertion of, or recombination with, of a selectable element.
  • the exogenous nucleic acid may comprise a selection element located within transposon inverted repeats close to the site of the mutation to be corrected, for example the inverted repeat sequence may be between 3-100 nucleotides away from the site of the mutation to be corrected, such as 5-20 nucleotides, such as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 nucleotides.
  • the exogenous nucleic acid may be engineered to contain a transposon insertion site for insertion of a transposon carrying a selection element, to allow the selection element to be located close to the site of the mutation to be corrected.
  • the nucleic acid may be synthesized to contain a suitable site for insertion of a transposon carrying a selection element.
  • the exogenous nucleic acid has been engineered to contain a site for transposon integration then in one aspect, which can be preferred, insertion of the transposon site does not change the protein encoded by the nucleic acid into which the site is engineered.
  • the site is the TTAA piggyBac insertion sequence.
  • the method disclosed herein comprises use of an exogenous nucleic acid capable of recombination into the host genome, the nucleic acid comprising a transposon insertion site which is not found in the same relative location in the genome of the cell to be modified and which site does not affect any amino acid coding potential of the nucleic acid, such that—after transposase mediated excision—the genome of the cell comprises a mutation that has been corrected or introduced and additionally comprises a transposon insertion site.
  • the insertion of the modifying sequence into the genome and subsequent excision of the selection element may result in a genome which has the native genomic sequence at the point of excision of the selection element, or may result in a genome which has a non-native genomic sequence at the point of excision of the selection element, suitably a nucleic acid sequence which does not change the sequence of any protein encoded by the nucleic acid.
  • the inverted repeat sequences flanking the selectable marker are piggyBac inverted repeat sequences that can be excised by the action of the piggyBac transposase.
  • the inverted repeat sequences are, in one aspect, about 200-350 base pairs, such as 250 or 300 bp, and may be unequal lengths, such as 300 bp for 5′ side and 250 bp for 3′ side, which are typical repeat length for piggyBac vector.
  • the minimla inverted repeat size is 311 and 236 base pairs for the 5′ and 3′ respectively. Other transposase sequences may be used.
  • transposase enzyme may provide for complete excision of the sequence comprised between those repeats to leave only a single transposon target site and leave no nucleotide footprint remaining in the genome, or may leave a single transposon target site which alters the nucleotide sequence of the genome of the cell but not the protein(s) encoded by that genome, for example.
  • an exogenous nucleic acid may be provided comprising a selection element located within piggyBac transposon inverted repeats, inserted into a piggyBac target site (TTAA). Insertion of the nucleic acid into the genome is followed by removal of the selectable element from the genome by action of a transposase such as the piggyBac transposase on the transposon inverted repeats, which will leave a TTAA remaining in the genome which TTAA could be either the wild type sequence or a variant of the genome if there is no naturally occurring TTAA sequence at that point in the genome.
  • Transposase action may be provided by, for example, expression of the transposase gene from a plasmid within the cell.
  • the transposase for use in the method is the piggyBac transposase, which optionally may be engineered to have increased activity with respect to the wild type sequence, and may be a hyperactive piggyBac transposase as disclosed herein.
  • the method can achieve at least, or greater than, 30%, 40%, 45% or 50% efficiency in targeting on one allele.
  • any suitable selection element can be employed within the cell to select for insertion of the exogenous nucleic acid into the genome of the cell, and suitable selection systems are well known in the art.
  • the selectable element is a puro delta tk cassette, which can be selected by FIAU (1-(-2-deoxy-2-fluoro-1-b-D-arabinofuranosyl)-5-iodouracil (FIAU)).
  • the method is a method of modification of the genome of a cell comprising:
  • transposase within the cell to remove the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence.
  • the method is a method of modification of the genome of an iPS cell, the genome of which has a mutation associated with a disease phenotype, the method comprising delivering to the cell:
  • delivery is followed by selection for the insertion of the nucleic acid into the genome using the selection marker;
  • the only exogenous nucleic acid remaining in the genome is the modifying sequence.
  • the genome of the ipS cell can then be analysed to confirm that no changes have been introduced into the genome of the cell other than those intended.
  • the cell may be differentiated after removal of the selection element using methods known in the art.
  • transposase integrated into the genome of the cell.
  • the transposase may be provided to the cell on an expression plasmid, for example. Suitable plasmid expression systems are well known in the art.
  • a cell comprising an exogenous nucleic acid, the nucleic acid comprising (i) a modifying sequence and (ii) a selection element flanked by transposon inverted repeats.
  • This cell represents the intermediate of the process described in more detail above.
  • a cell comprising a genome which has been modified by insertion of a modifying sequence using a method according to claim 1 .
  • Such a cell represents the product of the method of the invention.
  • pairs of cells are isogenic.
  • a modified cell comprising a modifying nucleic acid sequence, the cell having been selected for the insertion of the modifying nucleic acid sequence using a selectable element, and wherein the selectable element is then removed from the genome, such that the genome of the modified cell is identical to the genome of the original starting cell except for the modifying nucleic acid.
  • a cell includes, for example, a cell selected for correction of a mutation and in which no other genetic changes have been made to the genome other than the correction event.
  • Also disclosed herein is a collection of cells, such as an organ or tissue, wherein the genomes of all, or a proportion, of the cells has been modified by insertion of a modifying sequence using a method as described above.
  • Also disclosed herein is a method for treating a disease in a patient, the method comprising modifying the genome of a cell using a method as described above to correct a mutation associated with the disease in the cell, and using the cell, or cells derived or differentiated therefrom, in disease treatment.
  • the method for treatment of disease may include isolation of a cell from a individual, conversion of that cell to an iPS cell, modification of the genome of the cell as disclosed herein to effect a desired change to the genome of the cell, and then either delivery of that cell to the individual or differentiation of the cell to form a different cell type followed by delivery of the differentiated cell to the individual.
  • the treatment is an autologous cell based therapy.
  • Cells of the invention may be used in combination with pharmaceutically acceptable excipients and/or carriers, such as buffers, examples of which are well known in the art.
  • iPS and stem cell therapies have been suggested or tried in the following disease areas, all of which may be applicable for the present methods; brain degeneration, such as in Parkinson's and Alzheimer's disease, cancer, Cystic fibrosis, cystic fibrosis, a-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease.
  • the method of the invention can also be used ex-vivo. i.e. directly on cells isolated from a patient and then returned in their original differentiation state.
  • the method of the invention can also be used to directly modify the genome of cells in a tissue.
  • the method of the invention therefore can comprise in vivo delivery of DNA and zinc-finger nucleases or TALENS to a tissue where the DNA is taken up, either as naked DNA or by an appropriate formulation, such as being packaged in a virus.
  • the present invention provides cells or tissues, such as iPS cell or tissues differentiated therefrom, into which specific modifications have been introduced, such as modifications that correct mutations.
  • the cells or tissues of the invention may be used as models for understanding disease, and the invention relates to uses of the modified cells of the invention or cells or tissues derived therefrom, optionally used in combination with the original unmodified cell, in the assessment of any of the following:
  • a therapeutic agent e.g. drug screening
  • modified cells of the invention may also be used to predict which patient sub-groups will respond to currently-available and future drug treatments.
  • the present invention provides pairs of cells or cell lines or tissues that differ only by a defined modification or modifications, and comparison of such cells or cell lines or tissues allows the precise effect of the modification(s) to be assessed.
  • the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
  • such open ended terms also comprise within their scope a restricted or closed definition, for example such as “consisting essentially of”, or “consisting of”.
  • A, B, C, or combinations thereof refers to all permutations and combinations of the listed items preceding the term.
  • A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB.
  • expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth.
  • BB BB
  • AAA AAA
  • AB BBC
  • AAABCCCCCC CBBAAA
  • CABABB CABABB
  • compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • mice induced pluripotent stem cells isolated from fibroblasts of the C57B16-Tyr c-Brd strain 15 .
  • the targeting vector was constructed, carrying a wild-type 290G sequence and a PGK-puro ⁇ tk cassette flanked by piggyBac repeats into the TTAA site ( FIG. 1 a ).
  • the selection cassette was excised from the mIPSCs genome by transient expression of the piggyBac transposase and subsequent FIAU selection.
  • Genomic modification was verified by Southern blot and PCR analyses ( FIGS. 1 b, c ).
  • the correction of the G290T mutation and seamless piggyBac excision were confirmed by sequence analyses ( FIGS. 1 d, e ).
  • Two introduced silent mutations were observed, confirming that the T290G substitution was mediated by gene correction, not by spontaneous reversion ( FIG. 1 e ).
  • the function of the reverted allele was tested by injecting the corrected mIPSCs into albino mouse blastocysts.
  • the resulting chimeric mice displayed a black coat color, indicating phenotypic correction of the albino mutation ( FIG. 1 f ).
  • A1ATD is an autosomal recessive disorder found in 1 out of 2000 individuals of North European descent and represents the most common inherited metabolic disease of the liver 17,18 . It results from a single point mutation in the MAT gene (the Z allele; Glu342Lys) that causes the protein to form ordered polymers within the endoplasmic reticulum of hepatocytes 17,18 . The resulting inclusions cause cirrhosis for which the only current therapy is liver transplantation.
  • Genomic instability is known to be associated with prolonged culture of hESCs 20,21 and those arising during genome modification would be another concern for clinical application of hIPSCs. Therefore, we analyzed the genomic integrity of the hIPSC lines using comparative genomic hybridization (CGH) (Table 4a-c).
  • CGH comparative genomic hybridization
  • Two out of three A1ATD-hIPSC primary lines differed from their parental fibroblasts, showing amplifications or deletions ranging from 20 kb to 1.3 Mb, including a gain of 20q11.21, a frequently amplified region in hESCs 22,23 (see FIG. 7 ).
  • Line A retained a normal genome content compared to its parental fibroblast.
  • ZFN off-target cleavage and imprecise excision after multiple piggyBac transposition might introduce mutations into the genome.
  • hIPSCs were differentiated in vitro into hepatocyte-like cells, the main cell type affected by the disease A1ATD. Differentiation of the corrected lines occurred as expected, resulting in a near homogenous population of hepatocyte-like cells ( FIGS. 9 a - c ).
  • CGH analysis of differentiated cells showed that hepatic differentiation neither increases the number of genetic abnormalities nor selects for cells with abnormal karyotype (Table 4d).
  • the resulting cells shared key functional attributes of their in vivo counterparts including glycogen storage, LDL-cholesterol uptake, albumin secretion and Cytochrome P450 activity ( FIGS.
  • A1ATD-hIPSCs were described previously 16 .
  • 2 ⁇ 10 6 hIPSCs were co-transfected with ZFN expression vectors and the donor template, and subjected to puromycin selection (1 ⁇ g ml ⁇ 1 ) initiated 4 days after transfection.
  • targeted cells were transfected with pCMV-hyPBase 8 , cultured for 4 days, replated and selected in 250 nM 1-(2-Deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-indouracil (FIAU).
  • ROCK inhibitor 26 Y-27632 (10 ⁇ M) 4 hours prior to dissociation and 24 hours post plating. Resulting colonies were picked 2 weeks later, analyzed by PCR and further verified by Southern blot analysis. Primer sequences are listed in Supplementary Table 7.
  • a Het. clones heterozygous for PB allele.
  • b Homo./Hemi. clones homozygous or hemizygous for PB allele.
  • Cells with one targeted allele and deletion of the other allele are undistinguishable from correctly targeted homozygous clones by PCR. Such cells are designated as hemizygotes.
  • c Vector backbone integration was analyzed by PCR.
  • d Clones showing incorrect PCR bands are included. e A sum of 2 independent experiments.
  • a destination vector pPB-R1R2-NP was constructed as follows. The attR1 and attR2 sites were PCR-generated and digested by NheI/HindIII and XhoI/SpeI, respectively. EM7-neo was PCR-generated and digested by HindIII/XhoI. These 3 fragments were then cloned into the NheI-SpeI site of pPB-LR527, resulting in pPB-R1R2-Neo.
  • An EcoRI-XbaI fragment containing PheS was excised from pR6K-R1R2-ZP28, blunt-ended and cloned into the blunted XhoI site of pPB-R1R2-Neo, resulting in pPB-R1R2-NP.
  • An entry vector pENTR-PGKpuro ⁇ tk was constructed by cloning a KpnI-NotI PGK-puroitk fragment into the KpnI-NotI site of pENTR-2B.
  • a targeting vector for Tyr The targeting vector was constructed using BAC recombineering. A BAC clone RP24-221M7 was introduced into Escherichia coli
  • strain EL350 29 A mini targeting vector was first constructed to modify the Tyr gene on the BAC. Left and right homology arms were PCR-generated and digested by AscI/BsiWI and NsiI/PacI, respectively. The transposon fragment was excised from pPB-R1R2-NP by NsiI/BsiWI digestion. These 3 fragments were then cloned into AscI/Pact site of pMCS, resulting in pMCS-Tyr-NP. An AscI-PacI fragment was excised from pMSC-Tyr-NP and used for BAC targeting.
  • a retrieving vector was constructed by cloning PCR-generated left and right homology arm into the XhoI/AscI site of pMSC-DTA, following AscI/HindIII and XhoI/HindIII digestion of the left and right arm, respectively.
  • the retrieving vector was linearized by HindIII digestion and used to retrieve 3.0-kb 5′ arm, the transposon and 6.5-kb 3′ arm.
  • the Neo-PheS cassette was replaced with the PGK-puro ⁇ tk cassette by Gateway cloning, resulting in pDTA-TyrPB.
  • the targeting vector was linearized by AscI prior to electroporation into the albino mIPSCs.
  • a donor template vector for A1AT A 2-kb fragment, which contained 1 kb at both side of Z mutation, was first PCR-amplified using genomic DNA from A1ATD-hIPSC line B as a template and cloned into pCR4-blunt-TOPO (Invitrogen), resulting in pCR4-AAT_Z.
  • To construct a donor template with corrected sequence and a piggyBac transposon the 5′ arm and 3′ arm were PCR-amplified and digested with AscI/NsiI and BsiWI/PacI, respectively.
  • the NsiI-BsiWI fragment containing a piggyBac transposon with the Neo-PheS cassette was excised from pPB-R1R2-NP.
  • the digested fragments were cloned into the AscI-Pact site of pMCS, resulting in pMCS-AAT-PB:NP.
  • the Neo-PheS cassette was subsequently replaced with a PGK-puro ⁇ tk cassette by Gateway cloning, resulting in the final donor vector, pMCS-AAT-PB:PGKpuro ⁇ tk.
  • the plasmids (pPB-R1R2-NP, pENTR-PGKpuro ⁇ tk, pMCS-AAT-PB:PGKpuro ⁇ tk) have been deposited in the Wellcome Trust Sanger Institute Archives and available upon request (http://www.sanger.ac.uk/technology/clonerequests/).
  • A1ATD-hIPSCs (ref. 16; A, patient 2 line 1; B, patient 1 line1; C, patient 3 line1) were cultured on MEF-feeder layers in hESC medium: DMEM/F12 supplemented with 20% knockout serum replacement, 1 mM GlutaMax, 0.1 mM 2-mercaptoethanol, 1 ⁇ non-essential amino acid and 4 ng ml-1 FGF2 (Invitrogen).
  • hIPSCs were cultured in DMEM containing 10% FCS, 2 mM Glutamine, 0.1 mM 2-melcaptoethanol and 1 ⁇ non-essential amino acid.
  • mIPSCs iPS25A1; ref.
  • KO-DMEM KO-DMEM supplemented with 15% FBS, 1 mM GlutaMax, 0.1 mM 2-mercaptoethanol, 1 ⁇ non-essential amino acid and 1000 unit ml-1 LIF (Millipore).
  • 1 ⁇ 107 cells were electroporated with 25 ⁇ g of a linearized targeting vector in 800 ⁇ l of HEPES-buffered saline using a Gene Pulser II electroporator (230 V, 500 ⁇ F) and plated onto three 10-cm dishes. The next day, puromycin selection (1 ⁇ g ml-1) was initiated. Resulting colonies were picked and screened by PCR. Targeted clones were expanded and further verified by Southern blot analysis. Correctly targeted clones were then subjected to transposon excision.
  • 2 ⁇ 106 cells were electroporated with 40 ⁇ g of pCMV-hyPBase in 800 ⁇ l of HEPES-buffered saline using a Gene Pulser II electroporator (230 V, 500 ⁇ F) and plated onto one well of a 6-well plate. After passage once, cells were replated on day 4 at 5 ⁇ 105 cells per 10-cm dish. On the following day, FIAU (0.2 ⁇ M) selection was initiated. On day 5 of selection, FIAU was withdrawn. Resulting colonies were picked at day 7 and screened by PCR. Primer sequences to detect homologous recombination are shown in Table 7.
  • the cell suspension was transferred to a cuvette and electroporated using the Amaxa Nucleofector device (Lonza) with program A23.
  • the electroporated cells were plated onto one or two 10-cm feeder dishes in MEF-conditioned hESC medium containing 10 ⁇ M Y-27632. hESC medium without any drug was used for daily medium change between day 1-3.
  • puromycin selection (1 ⁇ g ml-1) was started.
  • medium was changed to MEF-conditioned hESC medium containing 0.5 ⁇ g ml-1 puromycin, which was used for medium change at every other day until picking colonies. Resulting colonies were picked on day 13-17. Colonies was cut into 2 pieces.
  • One half was transferred onto a well of 24-well feeder plate and the other half was lysed and used for PCR-genotyping. PCR-positive clones were further expanded and homologous recombination was verified by Southern blot analysis.
  • Homozygously targeted clones (B-16, C-G4, SeV-1-C3 and SeV-3-G5) were used for transposon removal.
  • Line A-derived clones were omitted because this line displayed a lower capability of differentiating into endodermal lineages.
  • Cells were prepared as described above. 2 ⁇ 106 cells were mixed with 10 ⁇ g of the hyperactive piggyBac transposase expression vector (pCMV-hyPBase8) in 100 ⁇ l of hESC solution 1 and electroporated using the Nucleofector device with the program A23.
  • Electroporated cells were plated onto 6-well plate in 1:2, 1:3 and 1:6 dilutions in MEF-conditioned hESC medium containing 10 ⁇ M Y-27632. Note that ROCK inhibitor was added to the culture medium until day 6 in this experiment. On day 2, cells with ⁇ 80% confluency were passaged using Accutase at a split ratio 1:2, 1:3 and 1:6 into 6-well plates. On day 4, cells with ⁇ 80% confluency were washed with PBS, detached with Accutase, suspended in hESC medium and pelleted. Cells were resuspended in hESC medium into single-cell level and counted.
  • 1 ⁇ 104 cells were then plated onto one 10-cm dish in hESC medium containing 10 ⁇ M Y-27632. 16-18 hrs after plating (day 5), medium was changed to hESC medium containing 0.25 ⁇ M FIAU and 10 ⁇ M Y-27632. On day 6, medium was changed to hESC medium containing 0.25 ⁇ M FIAU and then medium was changed every other day. Genotype and deletion of the piggyBac transposon were analyzed by PCR and further verified by Southern blot analysis.
  • An Illumina HumanCytoSNP-12 SNP array was used following the manufacturer's protocol. Genotype calls were performed by Illumina's GenomeStudio. B allele frequency and log R ratio were analyzed by KaryoStudio. CNVpatition v2.4.4 bundled in KaryoStudio was used for copy number analysis.
  • ZFNs were designed against a region containing Z mutation in the A1AT gene (see FIGS. 2 a, b ) and assembled as previously described 30 .
  • the amino acid residues at positions ‘ ⁇ 1’ to ‘+6’ of the recognition alpha helix 31,32 of each of the zinc finger DNA-binding domain for each DNA triplet target are shown in Table 4.
  • the ZFNs were linked to wild type FokI catalytic domain.
  • the activity of the ZFN at the endogenous target site was determined using the Surveyor Nuclease assay as previously described 33 .
  • the corrected iPSC line, B-16-C2, and its parental fibroblasts were analyzed. Exome sequencing and analysis were performed as described previously34 with minor modification. Exome pulldown was performed using an Agilent SureSelect Human All Exon 50 Mb Kit according to the manufacturer's instructions. Enriched DNA was sequenced on an Illumina HiSeq2000 (75-bp paired-end sequencing). 90.32% (Fibroblast-B) and 90.72% (B-16-C2) of total targeted regions were covered with more than 10 ⁇ sequencing depth, covering 93.01% and 93.35% of CCDS exons, respectively. Substitutions in the coding sequence were called as positions with at least 20% of reads reporting a different base with respect the reference human sequence (GRCh37).
  • somatic mutations were identified by comparing the sequence with the control fibroblasts, and removing the common polymorphisms described in dbSNP and in the 1000 Genomes Project35. Small insertions and deletions were identified using samtools, as the ones not present in the control cell line and that had at least 20 ⁇ of coverage and 20% of the reads reporting the mutation. Validation of mutations was carried out by Sanger capillary sequencing on parental Fibroblast-B, A1ATD-hIPSC line B, the homozygously targeted B-16 cells and the piggyBac-excised B-16-C2 cells.
  • the primary hIPSC line C also showed a slightly higher ratio in the affected region than in the adjacent region whereas line A and B, both carrying normal chromosome 20 based on the SNP analysis, had same ratios.
  • line C is heterogeneous with a minor population of cells with amplified 20q11.21.
  • the mean log2 ratio of this region in line C is 0.208, which indicates 2.3 copies per diploid genome ( FIG. 7 d ).
  • line C consists of 2 cell types (2-copy normal and 3-copy abnormal cells), 30% of the population carries 3 copies of 20q11.21.

Abstract

The present disclosure includes methods for manipulation of the genome, to products obtained or obtainable from such methods, and uses of these products.

Description

  • This application is a continuation in part of PCT/GB2011/001275, filed Aug. 25, 2011, which claims the benefit of GB Application No. 1014169.5, filed Aug. 25, 2010, both of which are herein incorporated by reference in its entirety.
  • BACKGROUND
  • A number of different techniques are available for genome modification. However, in many such methods the modification process leaves a residual sequence in the targeted genome. For example, Soldner et al (Cell, 146, 318-331, July 2011) disclose 4 targeting strategies for genome modification in an iPS cell. Two selection based strategies leave residual DNA (a IoxP site) in the genome. The non-selection based approaches have the obvious disadvantage that they are less efficient than selection based approaches.
  • SUMMARY OF DISCLOSURE
  • A method of modifying the genome of a cell, the method comprising recombining into the genome of the cell an exogenous nucleic acid, the nucleic acid comprising
  • (i) a modifying sequence and
  • (ii) a selection element flanked by transposon inverted repeats,
  • the method further comprising the steps of:
  • selection of insertion of the nucleic acid into the genome using the selection element, and
  • use of a transposase to excise the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence
  • A method of correcting or introducing a mutation in a genome of a cell, the method comprising
  • i recombining into the genome of the cell a nucleic acid comprising (i) a modifying sequence which corrects or introduces the mutation and (ii) a selection element flanked by transposon inverted repeats,
  • ii selection of insertion of the nucleic acid into the genome using the selection element, and
  • iii use of a transposase to excise the selection element.
  • A cell comprising an exogenous nucleic acid, the exogenous comprising (i) a modifying sequence and
  • (ii) a selection element flanked by transposon inverted repeats
  • A cell comprising a genome which has been modified by insertion of a modifying sequence using a method according to claim 1.
  • A modified cell comprising a modifying nucleic acid sequence, the cell having been selected for the insertion of the modifying nucleic acid sequence using a selectable element, and wherein the selectable element is then removed from the genome, such that genome of the modified cell is identical to the genome of the original starting cell except for the modifying nucleic acid.
  • A collection of cells, such as an organ or tissue, wherein the genomes of all, or a proportion, of the cells have been modified by insertion of a modifying sequence using a method as described above.
  • A collection of cells, such as an organ or tissue, wherein a mutation in the genomes of all, or a proportion, of the cells has been corrected by insertion of a modifying sequence using a method as described above.
  • A method for treating a disease in a patient, the method comprising modifying the genome of a cell using a method as described above to correct a mutation associated with the disease in the cell, and using the cell, or cells derived or differentiated therefrom, in disease treatment.
  • Use of a cell as described above in treatment of disease. A cell as described above for use in the treatment of disease.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Correction of the G290T mutation in the Tyr gene in mIPSCs.
  • a, The strategy for precise genome modification using the piggyBac transposon. Top line, structure of the Tyr gene; red line, 5′ external probe for Southern blot analysis; open arrow, piggyBac transposon carrying a PGK-puroΔtk cassette; P1, P2 and P3, PCR primers; B, BamHI; E, EcoNI. b, c, Southern blot (b) and PCR analyses (c) showing insertion (c/PB) and excision (c/Rev) of the piggyBac transposon. ES, mouse ESCs as a control. d, e, Sequence analyses revealed correction of the G290T mutation (d) and seamless excision of the piggyBac transposon (e). Note that two silent mutations (A and T indicated by arrowheads) introduced near the TTAA site were also detected. f, A chimeric mouse generated by injecting corrected Tyr c/Rev mIPSCs (left) displays black coat color. Right, a non-injected albino mouse.
  • FIG. 2. Correction of the Z mutation in A1ATD-hIPSCs.
  • a, The strategy for precise genome modification using ZFNs and the piggyBac transposon. Top line, structure of the A1AT gene; blue lines, Southern blot probes; thin and thick boxes, non-coding and coding exons, respectively; open arrow, piggyBac transposon; B, BamHI; A, AflIII. b, Sequences of wild-type (Reference), Z, PB, and Rev alleles. Amino acid position 342 (blue), recognition sites for ZFNs (green), piggyBac excision site (red) are shown. Sequence changes in Rev allele from Z allele were indicated by asterisks. c, Surveyor nuclease assay showing the cleavage of Z mutation in ZFNs-transfected K562 cells. Non-transfected cells were used as a control. d, Southern blot analysis showing bi-allelic piggyBac insertion (B-16) and bi-allelic excision (B-16-C2, -C3 and -C6) during correction of the A1ATD-hIPSCs line B. Genomic DNA was digested by BamHI (5′ and PB probes) or AlfIII (3′ probe). Genotype: ZZ, homozygous for Z allele; PP, homozygous for insertion of piggyBac; RR, homozygous for reverted allele. e, Sequence analysis showing correction of Z mutation in 3 corrected hIPSC lines. Wild-type sequence (top line) and A1ATD-hIPSC (second line).
  • FIG. 3. Functional analysis of restored A1AT in c-hIPSCs-derived hepatocyte-like cells.
  • a, Immunofluorescence showing the absence of polymeric A1AT protein in hepatocyte-like cells generated from c-hIPSCs. All forms of A1AT (left panels) and misfolded polymeric A1AT (middle panels). b, c, ELISA to assess the intracellular (b) and secreted (c) levels of polymeric A1AT protein in hepatocyte-like cells derived from A1ATD-hIPSCs (ZZ), c-hIPSCs (RR) and control hIPSCs (++). d, Endoglycosidase H (E) and peptide:N-glycosidase (P) digestion of A1AT immunoprecipitated from uncorrected (ZZ), corrected (RR) and control (++) hIPSC-derived hepatocyte-like cells (upper panels) and corresponding culture medium (lower panels). e, Chymotrypsin ELISA showing that corrected cells (RR) have A1AT enzymatic inhibitory activity that is superior to uncorrected cells (ZZ) and close to adult hepatocytes. f, g, Immunofluorescence of transplanted liver sections detecting human albumin (f) and A1AT (g). DNA was counterstained with DAPI. h, ELISA read-out of human albumin in the mouse serum longitudinally followed for each mouse. Asterisk, the mouse was subjected to histology analysis. Scale bars, 100 μm. Data in b, c and e are shown as mean±s.d. (n=3). Student's t-test was performed. NS, not significant.
  • FIG. 4. Southern blot analyses of bi-allelic targeting of the A1AT gene in 3 independent A1ATD-iPSC lines. a, The structures of the Z allele and the targeted PB allele. Thin and thick open boxes, non-coding and coding exon, respectively; B, BamHI. b, The result obtained with A1ATD-iPSCs from patient B. Both alleles in line B-16 were correctly targeted while line B-17 carried one correctly targeted allele and one abnormal allele. c-e, Similar results were obtained with A1ATD-iPSCs generated from patient A (c) and from patient C (d, e). Genomic DNA was digested by BamHI and hybridized with the 5′ external probe. The first lane on each panel is DNA from the parental A1ATD-iPSC line. Correct homozygous clones are highlighted in red.
  • FIG. 5. Characterization of genome modification in A1ATD-IPSC line C.
  • a, b, Southern blot (a) and sequence (b) analyses showing bi-allelic correction of Z mutation in A1ATD-iPSCs generated from patient C. See FIG. 2 for details.
  • FIG. 6. Pluripotency of corrected iPSCs.
  • a, Immunofluorescence showing the expression of pluripotency markers OCT4, SOX2, NANOG and TRA1-60 in corrected iPSCs grown for 20 passages after correction. DNAs were stained with DAPI (lower panels). b, Immunofluorescence of in vitro differentiated corrected iPSCs showing maintenance of differentiation capacity into three germ layers. Scale bars, 200 μm
  • FIG. 7. Detailed analysis of 20q11.21 amplification
  • a, aCGH result of the homozygous clone, C-B6. Top, an ideogram of chromosome 20; middle, genes in the region pointed by red in the chromosome ideogram; bottom, the log2 ratio plot of C-B6. The amplified region is highlighted in red. The log2 ratios in the adjacent region highlighted in gray are used as a normal copy number control for a comparison shown in c. b, SNP data showing B allele frequency (x axis on top) and smoothed log R ratio (x axis on bottom) of chromosome 20 of indicated cell lines. The 20q11.21 is highlighted in gray. c, Box plots showing log2 ratios of the affected (red) and the adjacent normal (gray) regions. Bars within the box plots represent median values. The ends of bars indicate the 25th and 75th percentiles, and the 10th and 90th percentiles are represented by error bars. Student' s t-test was performed. NS, not significant. d, Copy number and normalized copy number of the 20q11.21 region per diploid genome. Copy numbers were calculated from mean values of log2 ratio in the region. Copy numbers in the homozygous clones were further normalized with the calculated copy number of A1ATD-iPSC line C
  • FIG. 8. SNP analyses. a, b, SNP array data showing entire chromosome 14 (a) and 830-kb region around the A1AT (SERPINA1) gene (b). Data for fibroblasts and correctly targeted homozygous clones from each patient are shown. Blue dots represent B allele frequency (left y axis) of each SNP and red lines represent smoothed Log R ratio (right y axis). Note that heterozygosity is maintained in all 6 targeted clones.
  • FIG. 9. Differentiation of corrected iPSCs into hepatocyte-like cells.
  • a, Quantitative RT-PCR analyses showing the expression of markers of liver development during corrected iPSC differentiation. Values are normalized to expression on day 0. b, Immunostaining showing the expression of specific proteins marking key stages of hepatocyte development during corrected iPSC differentiation (Day 3, endoderm; Day 10, hepatic endoderm; Day 18 and 20, hepatic progenitor; Day 25, hepatocyte-like cells). c, Flow cytometric analyses showing that 80% of cells are positive for CXCR4 at 3 days of differentiation (endoderm stage) and 80% of cells are positive for Albumin at 25 days of differentiation. d-g, Corrected iPSC-derived hepatocyte-like cells display functional activity characteristic of primary human hepatocytes including presence of intracellular glycogen storage as shown by periodic acid Schiff staining (d), uptake of DiL-LDL (e), albumin secretion (f) and CytP450 metabolism (g). Scale bar, 100 μm
  • FIG. 10. Characterization of integration-free A1ATD-iPSC.
  • a, b, Pluripotency marker expression by RT-PCR (a) and immunostaining (b) analyses. Note that Sendai virus (SeV) genome were not detected in established iPSC lines. RV, retrovirus. c, Bisulfite sequencing of OCT4 and NANOG promoter regions. d, Southern blot analysis of the key pluripotency gene, OCT4, showing an evidence of integration-free iPSCs. Genomic DNA was digested with PstI. The OCT4 exon1 was used as a probe. Arrowhead, endogenous OCT4; asterisk, provirus-derived OCT4. e, CGH plot of primary A1ATD-iPSC line 3, showing no copy number change. f, Immunostaining of in vitro differentiated cells, showing maintenance of differentiation capacity. Scale bars, 500 μm (b), 200 μm(f)
  • FIG. 11. Correction of the Z mutation in the integration-free A1ATD-iPSC line B.
  • a, Validation of genetic modification by Southern blot analysis. See FIG. 2 for the detail. b, Sequencing analysis of gene correction. Note that the Z mutation (A) is corrected to wild-type sequence (G), while the piggyBac excisions produced TTAA as designed. c, CGH analysis showing that the 3-G5-A7 line kept the intact genome compared to the paretal fibroblasts. d, Immunostaining of 3-G5-A7-derived hepatocyte-like cells, showing that the corrected cells express normal A1AT porteins. Left, all forms of A1AT; middle, polymeric A1AT; right, DNA staining by DAPI. Scale bar, 50 μm.
  • DETAILED DESCRIPTION
  • The present disclosure generally relates to method of modifying the genome of a cell, the method comprising recombining into the genome of the cell an exogenous nucleic acid, the nucleic acid comprising
  • (i) a modifying sequence and
  • (ii) a selection element flanked by transposon inverted repeats,
  • the method further comprising the steps of:
  • (a) selection of insertion of the nucleic acid into the genome using the selection element, and
  • (b) use of a transposase to excise the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence.
  • Recombination of the exogenous DNA sequence into the cell suitably occurs by homologous recombination. As such the exogenous nucleic acid is suitably in a form that can be incorporated into the genome of the cell by homologous recombination and the nucleic acid is provided to the cell under conditions that allow homologous recombination to occur. These conditions are well known in the art. In one aspect the exogenous nucleic acid is DNA, which may be provided as a circular plasmid or linear nucleic acid.
  • Homologous recombination is well known and understood. For example, the exogenous nucleic acid to be inserted suitably comprises two regions the same or a very similar sequence to the genome of the cell to be modified, such that homologous recombination between the homologous regions results in insertion of the exogenous nucleic acid into the chromosome, replacing the equivalent genomic sequence.
  • The exogenous nucleic acid to be inserted comprises a selectable marker and the insertion event can therefore be selected for by selection of the selection element under appropriate selection conditions.
  • In a further aspect of the invention the efficiency of targeting can be increased using Zinc Finger Nucleases (ZFNs) or Transcription activator like effector nucleases (TALENS) to affect double stranded breaks which can be repaired by, for example, homology directed DNA repair pathways using an exogenous donor plasmid or other exogenous nucleic acid sequence as a template. These approaches may be particularly useful in human cells where homologous recombination can be relatively inefficient. Thus the methods disclosed herein include those in which homologous recombination is used in combination with a technique for increasing the efficiency of the homologous recombination event. For example, the method of the invention also comprises providing to the cell a zinc finger nuclease or Transcription activator like effector nucleases (TALES), or nucleic acid encoding the same, to allow expression of the ZFN or TALES in the cell whose genome is to be modified.
  • By way of non-limiting example, we disclose herein that a combination of zinc finger nucleases (ZFNs)7 and piggyBac8,9 technology in hIPSCs can achieve bi-allelic correction of a point mutation (Glu342Lys) in the α1antitrypsin (A1AT, also called SERPINA1) gene that is responsible for α1-antitrypsin deficiency (A1ATD). Genetic correction of hIPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene targeting technology that is currently available and prevents contamination of the host genome with residual non-human sequences. These results provide the first proof of principle for the potential of combining hIPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.
  • In one aspect the cell to be modified is a human cell or a non-human animal cell. In one aspect the cell is a pluripotent cell, such as a totipotent non-human cell, or a pluripotent human or non-human cell. The cell may also be an iPS cell, such as a human iPS cell or non-human iPS cell. An iPS cell may originate from a fibroblast cell, adipose tissue, bone marrow or other suitable somatic cell. iPS cells may be generated using methods well known in the art, such as expression of Yamanaka factors as disclosed in EP1970446 or as disclosed in Wang et at www.pnas.org/cgi/doi/10.1073/pnas.1100893108
  • iPS cells may be derived from individuals with diseases caused by genetic mutation, for example diseases affecting the epithelial, endothelial, or interstitial compartments of the lung, such as cystic fibrosis, a-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease.
  • In one aspect the cell to be modified is an iPS cell is derived from an individual in whom it is desired to re-transplant the iPS cell, or cells derived or differentiated therefrom.
  • In one aspect an iPS cell is reprogrammed using an integration-free method, for example as disclosed herein using Sendai virus vectors (ref 25). Suitably the reprogramming of an iPS cell does not leave any residual DNA in the genome of the iPS cell. Another example of suitable methodology includes that disclosed in Somers et al, Stem cells 2010;28:1728-1740. Another example of suitable integration free technology includes providing mRNA or DNA minicircle as disclosed in https://www.stemgent.com/applications/reprogramming.
  • In one aspect the iPS cell modified by the method of the invention maintains expression of pluripotency markers for at least 10, at least 15, or at least 20 passages and or the ability to differentiate into cells expressing markers of three germ layers.
  • The modifying sequence is the sequence of exogenous nucleic acid which is not the selection element flanked by the transposase inverted repeat sequences. This sequence of the exogenous nucleic acid generally remains in the genome after transposase excision.
  • The modifying sequence may comprise a nucleic acid sequence which is substantially identical to a part of the genome of the cell to be modified but which has an addition, substitution or deletion of nucleotides with respect to that genome. The modifying nucleic acid may be isogenic with the genome with the exception of the addition, substitution or deletion. The modifying nucleic acid may be isogenic with the genome with the exception of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides, for example, or even with the exception of up to 50 nucleotides, e.g. up to 100 nucleotides, up to 200 nucleotides, up to 500 nucleotides or up to 1 kb, or 2 kb.
  • Alternatively the modifying sequence may be a sequence which is only partly isogenic with the genomic DNA, for example at the ends of the nucleic acid sequence to allow for homologous recombination, but may comprise a region of non-isogenic DNA of some length, such as more than 20, 30 ,40 or 50 nucleotides, such as a gene from a species different to that of the cell to be modified.
  • In one aspect the modification causes a change in function of the cell, or the tissue or organism derivable from development of the cell. The modification need not be associated with a disease phenotype.
  • In one aspect the modification is correction of a mutation associated with disease, such as correction of a point mutation. A point mutation may be changed to a nucleotide that is not associated with a disease phenotype, for example, changing of the Lys 342 to Glu in the α1-antitrypsin (A1AT, also called SERPINA1) gene that is responsible for α 1-antitrypsin deficiency (A1ATD). There are many other diseases associated with mutations, such as point mutations, that can be addressed using the present methods, including Cystic fibrosis, Tay—Sachs disease, Sickle Cell Anemia and Neurofibromatosis.
  • In one aspect the modification restores an enzymatic function of the cell.
  • In one aspect the modification is a correction of one or both alleles of a gene. The modification may be a correction of one or both alleles of a point mutation associated with a disease phenotype.
  • As disclosed above, the method can be, for example:
  • A method of correcting a mutation in a genome of a cell, the method comprising
  • 1) recombining into the genome of the cell an exogenous nucleic acid comprising (i) a modifying sequence which corrects the mutation and (ii) a selection element flanked by transposon inverted repeats,
  • 2) selection of insertion of the nucleic acid into the genome using the selection element, and
  • 3) use of a transposase to excise the selection element,
  • wherein after the excision process the genome of the corrected cell is identical to the genome of the original cell with the only exception being mono or biallelic correction of the mutation.
  • In such a method the modifying sequence comprises the wild type nucleic acid sequence that would be found in individuals without disease which is inserted into the genome to replace the genome region comprising the mutation.
  • It will be appreciated that the modifying sequence can also be used to introduce a mutation into a cell, and thus the invention also relates to a method of correcting or introducing a mutation into a genome of a cell, as disclosed herein.
  • It will also be appreciated that cells accumulate mutations every time they divide such that no two cells are alike. Accordingly in one aspect, after the excision process, the genome of the corrected cell is identical to the genome of the original cell with the only exception being mono or biallelic correction of the mutation or mono or biallelic introduction of a mutation—such as a point mutation—but wherein, in one aspect, there may be additional mutations in the genome that occur due to the natural mutation processes that occur in during cell division.
  • Thus the invention relates to a method of correcting or introducing a modification into the genome of a cell as disclosed herein, wherein after the transposon mediated excision process the genome of the corrected cell comprises the desired specific modification and may comprise a random mutation(s) but comprises no other nucleic acid sequence alteration, for example, no nucleic acid sequence alteration that might arise from insertion of, or recombination with, of a selectable element.
  • In one aspect, the exogenous nucleic acid may comprise a selection element located within transposon inverted repeats close to the site of the mutation to be corrected, for example the inverted repeat sequence may be between 3-100 nucleotides away from the site of the mutation to be corrected, such as 5-20 nucleotides, such as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 nucleotides. The exogenous nucleic acid may be engineered to contain a transposon insertion site for insertion of a transposon carrying a selection element, to allow the selection element to be located close to the site of the mutation to be corrected. Alternatively the nucleic acid may be synthesized to contain a suitable site for insertion of a transposon carrying a selection element. Where the exogenous nucleic acid has been engineered to contain a site for transposon integration then in one aspect, which can be preferred, insertion of the transposon site does not change the protein encoded by the nucleic acid into which the site is engineered. In one aspect the site is the TTAA piggyBac insertion sequence.
  • Therefore, in another aspect, the method disclosed herein comprises use of an exogenous nucleic acid capable of recombination into the host genome, the nucleic acid comprising a transposon insertion site which is not found in the same relative location in the genome of the cell to be modified and which site does not affect any amino acid coding potential of the nucleic acid, such that—after transposase mediated excision—the genome of the cell comprises a mutation that has been corrected or introduced and additionally comprises a transposon insertion site.
  • As such, the insertion of the modifying sequence into the genome and subsequent excision of the selection element may result in a genome which has the native genomic sequence at the point of excision of the selection element, or may result in a genome which has a non-native genomic sequence at the point of excision of the selection element, suitably a nucleic acid sequence which does not change the sequence of any protein encoded by the nucleic acid.
  • In one aspect the inverted repeat sequences flanking the selectable marker are piggyBac inverted repeat sequences that can be excised by the action of the piggyBac transposase. The inverted repeat sequences are, in one aspect, about 200-350 base pairs, such as 250 or 300 bp, and may be unequal lengths, such as 300 bp for 5′ side and 250 bp for 3′ side, which are typical repeat length for piggyBac vector. In one aspect the minimla inverted repeat size is 311 and 236 base pairs for the 5′ and 3′ respectively. Other transposase sequences may be used. The action of a suitable transposase enzyme may provide for complete excision of the sequence comprised between those repeats to leave only a single transposon target site and leave no nucleotide footprint remaining in the genome, or may leave a single transposon target site which alters the nucleotide sequence of the genome of the cell but not the protein(s) encoded by that genome, for example.
  • In the present method, for example, an exogenous nucleic acid may be provided comprising a selection element located within piggyBac transposon inverted repeats, inserted into a piggyBac target site (TTAA). Insertion of the nucleic acid into the genome is followed by removal of the selectable element from the genome by action of a transposase such as the piggyBac transposase on the transposon inverted repeats, which will leave a TTAA remaining in the genome which TTAA could be either the wild type sequence or a variant of the genome if there is no naturally occurring TTAA sequence at that point in the genome. Transposase action may be provided by, for example, expression of the transposase gene from a plasmid within the cell.
  • Thus the method of the invention also comprises providing to the cell an expression vector comprising an expressable transposase. Expression vectors may be co-delivered with the exogenous DNA, for example by co-electroporation.
  • The invention is not limited to the inverted repeats of the piggyBac transposon and extends to any suitable transposon sequences that can be excised without leaving a footprint. In one aspect the transposase has a TTAA target site. In one aspect the transposon elements are from the superfamily of TTAA-specific mobile elements.
  • In one aspect the transposase for use in the method is the piggyBac transposase, which optionally may be engineered to have increased activity with respect to the wild type sequence, and may be a hyperactive piggyBac transposase as disclosed herein.
  • In one aspect the genome is modified on one allele of a gene after use of the method. In one aspect the genome is modified on both alleles of a gene after use of the method.
  • In one aspect the method can achieve at least, or greater than, 30%, 40%, 45% or 50% efficiency in targeting on one allele.
  • In one aspect the method is capable of simultaneous biallelic modification. In one aspect the method can achieve at least or greater than 1%, 2%, 3%, 4% biallelic targeting.
  • In the examples disclosed herein we demonstrate a very high efficiency of biallelic targeting which allows for the rapid isolation of cells with homozygous gene modification.
  • Any suitable selection element can be employed within the cell to select for insertion of the exogenous nucleic acid into the genome of the cell, and suitable selection systems are well known in the art. In one aspect the selectable element is a puro delta tk cassette, which can be selected by FIAU (1-(-2-deoxy-2-fluoro-1-b-D-arabinofuranosyl)-5-iodouracil (FIAU)).
  • Thus in one aspect the method is a method of modification of the genome of a cell comprising:
  • 1 Delivering to a cell an exogenous nucleic acid comprising:
  • (i) a modifying sequence and
  • (ii) a selection element flanked by transposon inverted repeats
  • 2 optionally delivering to the cell a zinc finger nuclease or TALES to enhance the efficiency of homologous recombination of the nucleic acid into the genome of the cell;
  • 3 selection of the insertion of the nucleic acid into the genome using the selection marker
  • 4 providing expression of a transposase within the cell to remove the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence.
  • In another aspect the method is a method of modification of the genome of an iPS cell, the genome of which has a mutation associated with a disease phenotype, the method comprising delivering to the cell:
  • 1 an exogenous nucleic acid comprising:
  • (i) a modifying sequence and
  • (ii) a selection element flanked by piggyBac inverted repeats; and
  • 2 a zinc finger nuclease or TALENS to enhance the efficiency of homologous recombination of the nucleic acid into the genome of the cell;
  • wherein delivery is followed by selection for the insertion of the nucleic acid into the genome using the selection marker; and then
  • providing expression of a piggyBac transposase within the cell to remove the selection element.
  • Suitably after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence.
  • In a further step the genome of the ipS cell can then be analysed to confirm that no changes have been introduced into the genome of the cell other than those intended.
  • In all methods of the invention, where a pluripotent call line is used, the cell may be differentiated after removal of the selection element using methods known in the art.
  • In one aspect of the methods of the invention there is no transposase integrated into the genome of the cell. The transposase may be provided to the cell on an expression plasmid, for example. Suitable plasmid expression systems are well known in the art.
  • Also disclosed herein is a cell comprising an exogenous nucleic acid, the nucleic acid comprising (i) a modifying sequence and (ii) a selection element flanked by transposon inverted repeats. This cell represents the intermediate of the process described in more detail above.
  • Also disclosed is a cell comprising a genome which has been modified by insertion of a modifying sequence using a method according to claim 1. Such a cell represents the product of the method of the invention.
  • Also disclosed herein is a pair of cells or cell lines, one of which has been modified from the other as disclosed herein, such that the cells differ from one another only by one genetic modification, for example by a point mutation, insertion or deletion. An example includes for example a wild type cell and one with a disease causing mutation. In one aspect pairs of cells are isogenic.
  • Further disclosed is a modified cell comprising a modifying nucleic acid sequence, the cell having been selected for the insertion of the modifying nucleic acid sequence using a selectable element, and wherein the selectable element is then removed from the genome, such that the genome of the modified cell is identical to the genome of the original starting cell except for the modifying nucleic acid. Such a cell includes, for example, a cell selected for correction of a mutation and in which no other genetic changes have been made to the genome other than the correction event.
  • Also disclosed herein is a collection of cells, such as an organ or tissue, wherein the genomes of all, or a proportion, of the cells has been modified by insertion of a modifying sequence using a method as described above.
  • Also disclosed herein is a method for treating a disease in a patient, the method comprising modifying the genome of a cell using a method as described above to correct a mutation associated with the disease in the cell, and using the cell, or cells derived or differentiated therefrom, in disease treatment.
  • In particular the method for treatment of disease may include isolation of a cell from a individual, conversion of that cell to an iPS cell, modification of the genome of the cell as disclosed herein to effect a desired change to the genome of the cell, and then either delivery of that cell to the individual or differentiation of the cell to form a different cell type followed by delivery of the differentiated cell to the individual.
  • Also disclosed is a modified cell as described herein in treatment of disease, and use of a cell in the preparation of a medicament for the treatment of disease.
  • In one aspect the treatment is an autologous cell based therapy.
  • Cells of the invention may be used in combination with pharmaceutically acceptable excipients and/or carriers, such as buffers, examples of which are well known in the art.
  • By way of example, iPS and stem cell therapies have been suggested or tried in the following disease areas, all of which may be applicable for the present methods; brain degeneration, such as in Parkinson's and Alzheimer's disease, cancer, Cystic fibrosis, cystic fibrosis, a-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease.
  • The method of the invention can also be used ex-vivo. i.e. directly on cells isolated from a patient and then returned in their original differentiation state.
  • The method of the invention can also be used to directly modify the genome of cells in a tissue. The method of the invention therefore can comprise in vivo delivery of DNA and zinc-finger nucleases or TALENS to a tissue where the DNA is taken up, either as naked DNA or by an appropriate formulation, such as being packaged in a virus.
  • The present invention provides cells or tissues, such as iPS cell or tissues differentiated therefrom, into which specific modifications have been introduced, such as modifications that correct mutations.
  • Therefore in a further aspect the cells or tissues of the invention may be used as models for understanding disease, and the invention relates to uses of the modified cells of the invention or cells or tissues derived therefrom, optionally used in combination with the original unmodified cell, in the assessment of any of the following:
  • the effect of a therapeutic agent (e.g. drug screening);
  • the identification of targets for drug treatment;
  • the toxicological effect of a therapeutic agent;
  • the safety of a therapeutic agent;
  • the effect of an individual modification or modifications on drug activity;
  • the effect of an individual modification or modifications on likely patient responsiveness the effect of an individual modification or modifications on likely patient resistance.
  • The use of the modified cells of the invention, optionally used in combination with the original unmodified cell, may also be used to predict which patient sub-groups will respond to currently-available and future drug treatments.
  • The present invention provides pairs of cells or cell lines or tissues that differ only by a defined modification or modifications, and comparison of such cells or cell lines or tissues allows the precise effect of the modification(s) to be assessed.
  • It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine study, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims.
  • All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
  • The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.” The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.” Throughout this application, the term “about” is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
  • As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps. In one aspect such open ended terms also comprise within their scope a restricted or closed definition, for example such as “consisting essentially of”, or “consisting of”.
  • The term “or combinations thereof” as used herein refers to all permutations and combinations of the listed items preceding the term. For example, “A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
  • All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • All documents referred to herein are incorporated by reference to the fullest extent permissible.
  • Any element of a disclosure is explicitly contemplated in combination with any other element of a disclosure, unless otherwise apparent from the context of the application.
  • The present invention is further described by reference to the following examples, not limiting upon the present invention.
  • The present invention is now illustrated in the following, non limiting example.
  • EXAMPLE 1
  • Human induced pluripotent stem cells (hIPSCs) represent a unique opportunity for regenerative medicine since they offer the prospect of generating unlimited quantities of cells for autologous transplantation as a novel treatment for a broad range of disorders1,23,4. However, the use of hIPSCs in the context of genetically inherited human disease will require correction of disease-causing mutations in a manner that is fully compatible with clinical applications3,5. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome6. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of hIPSCs. Here, we show that a combination of zinc finger nucleases (ZFNs)7 and piggyBac8,9 technology in hIPSCs can achieve bi-allelic correction of a point mutation (Glu342Lys) in the α1antitrypsin (A1AT, also called SERPINA1) gene that is responsible for α1-antitrypsin deficiency (A1ATD). Genetic correction of hIPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle for the potential of combining hIPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.
  • Currently available methods for gene targeting rely on positive selection to isolate rare clones that have undergone homologous recombination. To remove the unwanted selection cassettes, Cre/IoxP or Flp/FRT recombination systems are used, which leave behind single IoxP or FRT sites10,11. These small ectopic sequences have the potential to interfere with transcriptional regulatory elements of surrounding genes12, most of which are not fully characterized in the human genome. An alternative method to remove selection cassettes is to convert them into transposons. The most suitable transposon for this purpose is piggyBac, a moth-derived DNA transposon, which can transpose efficiently in mammalian cells including human embryonic stem cells (hESCs)9,13. A remarkable feature of this mobile element is seamless excision, which enables removal of transgenes flanked by piggyBac inverted repeats without leaving any residual sequences9,14.
  • To explore the use of piggyBac for the correction of point mutations, we designed a vector to correct an albino mutation (G290T substitution in the Tyr gene) in mouse induced pluripotent stem cells (mIPSCs) isolated from fibroblasts of the C57B16-Tyrc-Brdstrain15. The targeting vector was constructed, carrying a wild-type 290G sequence and a PGK-puroΔtk cassette flanked by piggyBac repeats into the TTAA site (FIG. 1 a). Following isolation of targeted clones, the selection cassette was excised from the mIPSCs genome by transient expression of the piggyBac transposase and subsequent FIAU selection. Genomic modification was verified by Southern blot and PCR analyses (FIGS. 1 b, c). The correction of the G290T mutation and seamless piggyBac excision were confirmed by sequence analyses (FIGS. 1 d, e). Two introduced silent mutations were observed, confirming that the T290G substitution was mediated by gene correction, not by spontaneous reversion (FIG. 1 e). The function of the reverted allele was tested by injecting the corrected mIPSCs into albino mouse blastocysts. The resulting chimeric mice displayed a black coat color, indicating phenotypic correction of the albino mutation (FIG. 1 f). These results collectively demonstrate that the piggyBac transposon can be used as a versatile tool for highly precise modification (e.g. correction or mutation) of the mammalian genome at a single base-pair level.
  • We next explored whether this approach could be used to correct a mutation in hIPSCs derived from individuals with α1-antitrypsin deficiency (A1ATD)16. A1ATD is an autosomal recessive disorder found in 1 out of 2000 individuals of North European descent and represents the most common inherited metabolic disease of the liver17,18. It results from a single point mutation in the MAT gene (the Z allele; Glu342Lys) that causes the protein to form ordered polymers within the endoplasmic reticulum of hepatocytes17,18. The resulting inclusions cause cirrhosis for which the only current therapy is liver transplantation. The increasing shortage of donors and harmful effects of immunosuppressive treatments impose major limitations on organ transplantation, making the potential of hIPSC-based therapy highly attractive. Since homologous recombination is relatively inefficient in hESCs6, we employed ZFN technology, which stimulates gene targeting in hESCs as well as hIPSCs7,10,19. ZFN pairs were designed to specifically cleave the site of the Z mutation (FIGS. 2 a-c, Table 1). Amino acid sequences of left (L) and right (R) ZFNs are shown. Zinc finger protein regions are underlined.
  • ZFN-L
    (SEQ ID NO: 1)
    MDYKDHDGDYKDHDIDYKDDDDKMAPKKKRKVGIHGVPAAMAERPFQCRICMRNFSQSGSLTRHIRTHTG
    EKPFACDICGRKFAQSADRTKHTKIHTGSQKPFQCRICMRNFSRSDHLSTHIRTHTGEKPFACDICGRKF
    AQSAHRITHTKIHLRGSQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNSTQDRILEMKVMEFFMKVY
    GYRGKHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMQRYVEENQTRNKHINPNEWWK
    VYPSSVTEFKFLFVSGHFKGNYKAQLTRLNHITNCNGAVLSVEELLIGGEMIKAGTLTLEEVRRKFNNGE
    INFRS
    ZFN-R
    (SEQ ID NO: 2)
    MDYKDHDGDYKDHDIDYKDDDDKMAPKKKRKVGIHGVPAAMAERPFQCRICMRKFAQSSDLRRHTKIHTG
    EKPFQCRICMRNFSQSSDLSRHIRTHTGEKPFACDICGRKFAQSGNLARHTKIHTPNPHRRTDPSHKPFQ
    CRICMRNFSQSGHLARHIRTHTGEKPFACDICGRKFARLDNRTAHTKIHLRGSQLVKSELEEKKSELRHK
    LKYVPHEYIELIEIARNSTQDRILEMKVMEFFMKVYGYRGKHLGGSRKPDGAIYTVGSPIDYGVIVDTKA
    YSGGYNLPIGQADEMQRYVEENQTRNKHINPNEWWKVYPSSVTEFKFLFVSGHFKGNYKAQLTRLNHITN
    CNGAVLSVEELLIGGEMIKAGTLTLEEVRRKFNNGEINFRS
  • A targeting vector was constructed from isogenic DNA with piggyBac repeats flanking the PGK-puroΔtk cassette (FIG. 2 a). To minimize the distance between the mutation and the piggyBac transposon, a CTG leucine codon, 10 bp upstream of the mutation, was altered to a TTA leucine codon, generating the TTAA sequence, which would be left in the genome following piggyBac excision (FIG. 2 b).
  • SUPPLEMENTARY TABLE 1
    ZFN design
    Target sequence a Triplet subsites Finger designs b
    ZFN name (5′-3′) (5′-3′) −1123456
    ZFN-L gtCGATGGTCAGCAca CGA QSAHRIT F4
    TGG RSDHLST F3
    TCA QSADRTK F2
    GCA QSGSLTR F1
    ZFN-R gaAAGGGActGAAGCTGCTgg AAG RLDNRTA F5
    GGA QSGHLAR F4
    GAA QSGNLAR F3
    GCT QSSDLSR F2
    GCT QSSDLRR F1
    a The core DNA target sequences (capital letters) and 2 flanking bases on each side (small letters) are shown.
    b The amino acid residues at positions ‘−1’ to ‘+6’ of the recognition alpha helix31,32 of each of the zinc finger DNA-binding domain for each DNA triplet target are shown.
  • Puromycin-resistant hIPSC colonies obtained after co-electroporation of ZFN expression vectors and the targeting vector were screened for targeted clones by PCR. A1ATD-hIPSC lines derived from 3 different patients yielded targeted clones (Table 2). Remarkably, 54% of the puromycin-resistant colonies were targeted on one allele, while 4% were the result of simultaneous targeting of both alleles (FIG. 4).
  • TABLE 2
    Summary of PCR genotyping of ZFN-stimulated gene targeting
    Het. + Homo./Hemi. +
    A1ATD- Clones Homo./ additonal additonal
    iPSC line analyzed Het.a Hemi.b integrationsc integrationsc Non-targetedd
    A 84 45 3 23 8 5
    B 18 10 2 3 3 0
    Ce 216 112 9 52 21 22
    Mean frequency [%] 54 6 23 12 5
  • To remove the piggyBac-flanked selection cassette from these modified clones, we transiently transfected two homozygously targeted clones (B-16 and C-G4) with a hyperactive form of the piggyBac transposase8 and subjected them to FIAU selection. The genotype of the resulting FIAU-resistant colonies was analyzed by PCR and confirmed by Southern blot (FIG. 2 d and FIG. 5 a). Bi-allelic excision was observed in 11% of FIAU-resistant colonies (Table 3). Sequence analyses demonstrated that the Z mutation was corrected on both alleles and that transposon excision yielded a TTAA sequence as initially planned (FIG. 2 b, e and FIG. 5 b). The resulting corrected A1ATD-hIPSC (c-hIPSC) lines maintained the expression of pluripotency markers for more than 20 passages and their abilities to differentiate into cells expressing markers of the three germ layers (FIG. 6), indicating that genome modification did not alter the pluripotency of c-hIPSCs.
  • TABLE 3
    Frequencies of bi-allelic piggyBac excision
    Bi-allelic excision Bi-allelic excision
    w/o re-integration w/re-integration
    Cell No. of Frequency No. of Frequency
    line analyzed clones [%] clones [%]
    B-16 88 15 17 33 38
    C-G4 94 5 5 19 20
    Mean frequency [%] 11 29
  • Genomic instability is known to be associated with prolonged culture of hESCs20,21 and those arising during genome modification would be another concern for clinical application of hIPSCs. Therefore, we analyzed the genomic integrity of the hIPSC lines using comparative genomic hybridization (CGH) (Table 4a-c). Two out of three A1ATD-hIPSC primary lines differed from their parental fibroblasts, showing amplifications or deletions ranging from 20 kb to 1.3 Mb, including a gain of 20q11.21, a frequently amplified region in hESCs22,23 (see FIG. 7). Line A retained a normal genome content compared to its parental fibroblast. Reassuringly, we found that after ZFN-stimulated targeting, four out of six homozygous clones had unaltered genomes compared to their parental hIPSC lines. Sixteen cell lines with bi-allelic piggyBac excision were compared with their corresponding primary hIPSCs and 12 had unaltered genomes. We also analyzed the hIPSC lines by SNP arrays to check for loss of heterozygosity and found that all lines analyzed retained heterozygosity throughout their genome (FIG. 8). This observation demonstrates that bi-allelic gene correction was the result of simultaneous homologous recombination followed by simultaneous excision at both alleles and that mitotic recombination was not involved in this process.
  • SUPPLEMENTARY TABLE 2
    aCGH analyses of primary A1ATD-hIPSCs and their derivatives
    A1AT Reference No. of affected
    Cell line genotype Genome Abnormality Chr. Band Size Mean log2 ratio genes
    a. Primary A1ATD-hIPSCs
    A Z/Z A-Fibroblast
    B Z/Z B-Fibroblast Gain 1 p36.11  300 kb 0.60 8
    Loss 8 q22.2   20 kb −0.93 1
    C Z/Z C-Fibroblast Loss 2 p22.3  207 kb −0.96 1
    Loss 17 q23.1  142 kb −0.76 3
    Gain* 20 q11.21 1.34 Mb 0.21 30
    b. Homozygously targeted clones
    A-A9 PB/PB A
    A-G6 PB/PB A Loss 14 q23.13   23 kb −0.87 0
    B-16 PB/PB B
    C-B6 PB/PB C Gain* 20 q11.21 1.34 Mb 0.71 30
    C-C11 PB/PB C
    C-G4 PB/PB C
    c. Clones with bi-allelic piggyBac excision
    B-16-C2 Rev/Rev B
    B-16-C3 Rev/Rev B
    B-16-C6 Rev/Rev B
    B-16-D2 Rev/Rev B
    B-16-E4 Rev/Rev B Gain 22 q11.23   47 kb 0.89 1
    B-16-E5 Rev/Rev B Gain 15 q26.2  4.7 kb 1.15 1
    B-16-F4 Rev/Rev B
    B-16-F10 Rev/Rev B Gain 17 q23.3-q24.2  4.7 Mb 0.62 56
    B-16-G8 Rev/Rev B
    B-16-G10 Rev/Rev B Gain 4 q31.3  262 kb 0.57 1
    B-16-H4 Rev/Rev B
    C-G4-B3 Rev/Rev C
    C-G4-C2 Rev/Rev C
    C-G4-C10 Rev/Rev C
    C-G4-D7 Rev/Rev C
    C-G4-E9 Rev/Rev C
    d. in vitro differentiated hepatocytes
    B-16-C2-hep. Rev/Rev B-16-C2
    *See Supplementary FIG. 5 and Supplementary Analysis for more detail.
  • ZFN off-target cleavage and imprecise excision after multiple piggyBac transposition might introduce mutations into the genome. In order to investigate these possibilities at a single basepair resolution, we sequenced exomes of the corrected B-16-C2 line and its parental fibroblast. Comparison of these exomes identified 29 mutations (Table 5). The genesis of these mutations was determined by analysis of the primary hIPSC line and the homozygously targeted intermediate. Twenty-four point mutations and one 1-bp deletion were detected in the primary hIPSC line and four mutations arose during genetic correction: one during targeting and three during piggyBac excision. These mutations appeared to arise during culture since their genomic signatures were inconsistent with ZFN off-target sites or piggyBac integration sites. Taken together, we conclude that the combination of ZFNs with piggyBac provides a new method for rapid and clean correction of a point mutation in hIPSCs without affecting their basic characteristics.
  • SUPPLEMENTARY TABLE 3
    Validated mutations in protein coding regions indentified by exome sequencing
    Wt Mut cDNA Protein Detected in SIFT functional Expression in
    Chr Position Allele Allele Gene_ID Transcript_ID Mutation type annotation annotation Fibro-B iPSC-B B-16 B-16-C2 prediction hepatocytes***
    Chr1 35223302 G A GJB5 ENST00000338513_r62 MISSENSE c.371G > A p.R124Q
    Figure US20130209426A1-20130815-C00001
    TOLERATED
    Chr1 173499158 C A SLC9A11 ENST00000367714_r62 SILENT c.2199G > T p.V733V
    Figure US20130209426A1-20130815-C00002
    N/A
    Chr1 233314872 C A PCNXL2 ENST00000258229_r62 MISSENSE c.3116G > T p.G1039V
    Figure US20130209426A1-20130815-C00003
    DAMAGING
    Chr3 42705381 C T ZBTB47 ENST00000457842_r62 SILENT c.702C > T p.G234G
    Figure US20130209426A1-20130815-C00004
    N/A +
    Chr3 100962755 C T IMPG2 ENST00000193391_r62 MISSENSE c.2420G > A p.R807K
    Figure US20130209426A1-20130815-C00005
    TOLERATED
    Chr3 189586471 G A TP63 ENST00000264731_r62 SILENT c.1095G > A p.S365S
    Figure US20130209426A1-20130815-C00006
    N/A
    Chr3 189586472 G A TP63 ENST00000264731_r62 MISSENSE c.1096G > A p.D366N
    Figure US20130209426A1-20130815-C00007
    TOLERATED
    Chr4 2831347 G A SH3BP2 ENST00000452765_r62 SILENT c.714G > A p.P238P
    Figure US20130209426A1-20130815-C00008
    N/A +
    Chr4 57182264 C A KIAAI211 ENST00000264229_r62 MISSENSE c.2596C > A p.Q866K
    Figure US20130209426A1-20130815-C00009
    TOLERATED
    Chr4 122254096 G T QRFPR ENST00000394427_r62 MISSENSE c.677C > A p.P226H
    Figure US20130209426A1-20130815-C00010
    DAMAGING
    Chr5 14692993 C T FAM105B ENST00000284274_r62 MISSENSE c.895C > T p.R299C
    Figure US20130209426A1-20130815-C00011
    TOLERATED +
    Chr5 33936907 T C RXFP3 ENST00000330120_r62 MISSENSE c.62T > C p.L21P
    Figure US20130209426A1-20130815-C00012
    DAMAGING*
    Chr6 166883328 C A RPS6KA2 ENST00000265678_r62 SPLICE Exon11 + 1G > T N/A
    Figure US20130209426A1-20130815-C00013
    N/A +
    Chr7 5567522 T C ACTB ENST00000331789_r62 MISSENSE c.985A > G p.I329V
    Figure US20130209426A1-20130815-C00014
    DAMAGING* +
    Chr9 124083640 A C GSN ENST00000373823_r62 MISSENSE c.1286A > C p.Y429S
    Figure US20130209426A1-20130815-C00015
    DAMAGING +
    Chr9 125424828 C T OR1L1 ENST00000373686_r62 SILENT c.984C > T p.T328T
    Figure US20130209426A1-20130815-C00016
    N/A
    Chr11 4143385 G A RRM1 ENST00000300738_r62 MISSENSE c.1053G > A p.M351I
    Figure US20130209426A1-20130815-C00017
    DAMAGING +
    Chr11 28119199 C T KIF18A ENST00000263181_r62 MISSENSE c.296G > A p.R99H
    Figure US20130209426A1-20130815-C00018
    DAMAGING
    Chr11 76909568 C A MYO7A ENST00000409709_r62 SILENT c.4470C > A p.I1490I
    Figure US20130209426A1-20130815-C00019
    N/A +
    Chr12 129160382 C T TMEM132C ENST00000315208_r62 MISSENSE c.511C > T p.R171C
    Figure US20130209426A1-20130815-C00020
    DAMAGING
    Chr13 99361826 G A SLC15A1 ENST00000376503_r62 MISSENSE c.1067C > T p.T356I
    Figure US20130209426A1-20130815-C00021
    DAMAGING +
    Chr14 69521906 A G DCAF5 ENST00000341516_r62 MISSENSE c.1597T > C p.S533P
    Figure US20130209426A1-20130815-C00022
    DAMAGING +
    Chr14 94844957 C T SERPINA1** ENST00000440909_r62 SILENT c.1086G > A p.L362L
    Figure US20130209426A1-20130815-C00023
    N/A +
    Chr14 94844959 G A SERPINA1** ENST00000440909_r62 SILENT c.1084C > T p.L362L
    Figure US20130209426A1-20130815-C00024
    N/A
    Chr14 106378125 G T IGHD5-5 ENST00000390588_r62 MISSENSE c.11C > A p.A4D
    Figure US20130209426A1-20130815-C00025
    Not scored
    Chr15 68497606 C T CALML4 ENST00000448060_r62 MISSENSE c.109G > A p.G37S
    Figure US20130209426A1-20130815-C00026
    TOLERATED +
    Chr15 86225404 A C AKAP13 ENST00000361243_r62 MISSENSE c.5129A > C p.H1710P
    Figure US20130209426A1-20130815-C00027
    DAMAGING +
    Chr17 79428053 C G BAHCC1 ENST00000307745_r62 MISSENSE c.6364C > G p.L2122V
    Figure US20130209426A1-20130815-C00028
    TOLERATED
    Chr18 52899761 G TCF4 ENST00000356073_r62 FRAMESHIFT c.1628 C > − p.S543fs*1
    Figure US20130209426A1-20130815-C00029
    N/A +
    ChrX 12938569 C A TLR8 ENST00000218032_r62 MISSENSE c.1410C > A p.F470L
    Figure US20130209426A1-20130815-C00030
    TOLERATED
    ChrX 107823778 C A COL4A5 ENST00000328300_r62 SILENT c.796C > A p.R266R
    Figure US20130209426A1-20130815-C00031
    N/A
    N/A, not appricable
    *Low confidence
    **These two silent mutations were intensionally introduced though genetic correction.
    ***Expression in hepatocyte was analyzed using control samples in GSE 11942. Genes with ‘present’ call in more than 50% of samples are considered as expressed.
    Figure US20130209426A1-20130815-C00032
    Figure US20130209426A1-20130815-C00033
  • To confirm that the genetic correction of hIPSCs resulted in the expected phenotypic correction, hIPSCs were differentiated in vitro into hepatocyte-like cells, the main cell type affected by the disease A1ATD. Differentiation of the corrected lines occurred as expected, resulting in a near homogenous population of hepatocyte-like cells (FIGS. 9 a-c). Remarkably, CGH analysis of differentiated cells showed that hepatic differentiation neither increases the number of genetic abnormalities nor selects for cells with abnormal karyotype (Table 4d). The resulting cells shared key functional attributes of their in vivo counterparts including glycogen storage, LDL-cholesterol uptake, albumin secretion and Cytochrome P450 activity (FIGS. 9 d-g). Importantly, immunofluorescence and ELISA both confirmed the absence of mutant polymeric A1AT in c-hIPSCs-derived hepatocyte-like cells that instead efficiently secreted normal endoglycosidase-H-insensitive monomeric A1AT (FIGS. 3 a-d). In addition, secreted A1AT displayed an enzymatic inhibitory activity that was comparable to that obtained from normal adult hepatocytes (FIG. 3 e), thereby suggesting that physiological restoration of enzyme inhibitory activity could be achieved.
  • Finally, the in vivo function of c-hIPSCs-derived hepatocyte-like cells (B-C16-2 line) was assessed following transplantation into the liver of Alb-uPA+/+;Rag2−/−;II2rg−/−mice via intra-splenic injection. Livers harvested 14 days after injection were colonized by human cells identified using antibodies specific to human albumin and A1AT (FIGS. 3 f, g). These human hepatocyte-like cells were distributed throughout the liver lobes and were seen to be integrated into the existing mouse parenchyma (FIGS. 3 f, g). In addition, human albumin was detected in the serum of transplanted animals for at least 5 weeks (FIG. 3 h), while no tumor formation was detected in any mice. Therefore, c-hIPSCs-derived hepatocyte-like cells were able to colonize the liver in vivo and display functional activities characteristic of their human ESC-derived counterparts24. Collectively these analyses demonstrate that genetic correction of the Z mutation resulted in functional restoration of A1AT in patient-derived cells.
  • All experimental evidence above strongly support the applicability of genetic correction in patient-specific iPSCs for cell-based therapy of A1ATD. We therefore repeated the genetic correction in more clinically relevant cells using patient-specific iPSCs reprogrammed from fibroblasts with Sendaiviral vectors, an integration-free method25 (FIGS. 10 a-f). One primary hIPSC line with an intact genome by CGH analysis (FIG. 10 e and Table 6) was corrected by the method described above. The final product, iPSC-3-G5-A7, had the corrected A1AT, had an intact genome compared to the parental fibroblast, and expressed normal A1AT protein when differentiated to hepatocyte-like cells (FIG. 11 and Table 6). This is the first demonstration of the generation of mutation-corrected patient-specific iPSCs, which could realize the therapeutic promise of hIPSCs.
  • SUPPLEMENTARY TABLE 4
    aCGH analyses of integration-free iPSCs and their derivatives.
    A1AT Reference No. of affected
    Cell line genotype Genome Abnormality Chr. Band Size Mean log2 ratio genes
    a. Primary factor-free A1ATD-hIPSCs
    SeV-iPSC1 Z/Z Fibroblast-B Gain 6 q22.1 271 kb 0.52 2
    SeV-iPSC2 Z/Z Fibroblast-B Loss 2 q23.3  18 kb −0.93 1
    SeV-iPSC3 Z/Z Fibroblast-B
    SeV-iPSC4 Z/Z Fibroblast-B Gain 10 q21.1  1.1 Mb 0.52 2
    Gain 16 p13.12  76 kb 0.61 0
    Gain 22 q13.33  86 kb 0.63 3
    SeV-iPSC5 Z/Z Fibroblast-B Gain 18 p11.31-p11.23 951 kb 0.6 5
    SeV-iPSC6 Z/Z Fibroblast-B Loss 22 q13.1  10 kb −0.81 1
    b. Homozygously targeted clones
    iPSC1-C3* PB/PB Fibroblast-B
    iPSC3-G5 PB/PB Fibroblast-B
    c. Clones with bi-allelic piggyBac excision
    iPSC1-C3-A8* Rev/Rev Fibroblast-B
    iPSC1-C3-A9* Rev/Rev Fibroblast-B
    iPSC3-G5-A7 Rev/Rev Fibroblast-B
    iPSC3-G5-C8 Rev/Rev Fibroblast-B Loss 20 p12.1 421 kb −1.00 1
    *A deletion on Chr.6 found in the parental line is not shown.
  • In the present study, we demonstrate that ZFNs and piggyBac transposon enable simultaneous bi-allelic correction of diseased hIPSCs. No residual ectopic sequences remain at the site of correction and the genome appears to be undisturbed elsewhere. Although we could readily obtain cell lines without large genomic alterations during genetic modification, the resulting corrected hIPSCs carry 29 mutations in protein coding exons, of which 22 were non-synonymous or splice site mutations. The likely impact of this mutation load needs to be considered in the context of their likely functional impact, taking into account the normal germ-line load, accumulated somatic variation, the presence of compensating normal gene copies and the requirement for the gene product in the derived differentiated cells. From this point of view, only eight mutations might affect gene functions in hepatocyte-like cells (Table 5). Nevertheless, the corrected iPSCs could efficiently differentiate to hepatocyte-like cells and engraft into the animal model for liver injury without tumor formation. Therefore, limited genomic abnormalities might have restricted biological consequences. Careful screening of primary and corrected hiPSCs using deep sequencing analyses would contribute to the safe use of hIPSCs in clinical applications.
  • hIPSCs derived from different patients were effectively corrected, demonstrating that this method could be applied to a large number of A1ATD-hIPSC lines. Since the bi-allelic correction could be carried out in less than 4 months, our approach may be compatible with large-scale production of corrected patient-specific hIPSCs not only for A1ATD but also for other monogenic disorders.
  • Method Summary
  • A1ATD-hIPSCs were described previously16. 2×106 hIPSCs were co-transfected with ZFN expression vectors and the donor template, and subjected to puromycin selection (1 μg ml−1) initiated 4 days after transfection. For transposon excision, targeted cells were transfected with pCMV-hyPBase8, cultured for 4 days, replated and selected in 250 nM 1-(2-Deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-indouracil (FIAU). To increase clonogenicity, cells were treated with ROCK inhibitor26, Y-27632 (10 μM) 4 hours prior to dissociation and 24 hours post plating. Resulting colonies were picked 2 weeks later, analyzed by PCR and further verified by Southern blot analysis. Primer sequences are listed in Supplementary Table 7.
  • SUPPLEMENTARY TABLE 5
    Primer sequences
    Construction of pPB-R1R2-NP attR1-F CTAGCTAGCACAAGTTTGTACAAAAAAGCTGAAC
    attR1-R CCCAAGCTTGAATTCGGATCCCATAGTGACTGGATATGTTGTGTT
    attR2-F CCGCTCGAGTCTAGACATAGTGACTGGATATGTTGTGTT
    attR2-R GACTAGTACCACTTTGTACAAGAAAGCTGAA
    EM7neo-F CCCAAGCTTGTTGACAATTAATCATCGGCATAG
    EM7neo-R CCGCTCGAGTCAGAAGAACTCGTCAAGAAGGCG
    Construction of the Tyr-mini Left arm-F GGCGCGCCATGGAACAGTGAAGTTCTCATCCCCAG
    targeting vector Left arm-R TTTAACGTACGTCACAATATGATTATCTTTCTAGGGTTAATGATATCAACATCTACGAC
    CTCTTTGTATG
    Right arm-F AAGAATGCATGCGTCAATTTTACGCAGACTATCTTTCTAGGGTTAAACATAGGTGTTGA
    TCCATTGTTCATTTGGCCATA
    Right arm-R CCTTAATTAAGCGGAAACTGTAAGTTTGGATTTG
    Construction of the Tyr- Left arm-F CCGCTCGAGTGCCCATGCCTAAACACAGATGTA
    retrieving vector Left arm-R CCCAAGCTTTTTTGGATTCGGATTCTGAGGCT
    Right arm-F CCCAAGCTTGTACAAGATTTTCATATGTAAGC
    Right arm-R AGGCGCGCCAGGAATTCATGCCCCAGTTGACA
    Homologous recombination F GTTGGCGCCTACCGGTGGATGTGGAATGTG
    detection at Tyr R GAAATCTCTTCAAGCTGGGAAAACCTAAAG
    Tyr genotyping P1 TCCTCGAGCCTGTGCCTCCTCTAAG
    P2 CCCAAGTACTCATCTGTGCAAATGTC
    P3 GCGACGGATTCGCGCTATTTAGAAAG
    Tyr
     5′ probe F ACTCATTATTCCAGGATACTTGAGTG
    R ATTTACCACATGCCCAAACTAATAAC
    A1AT 2-KB fragment F GTGGTGGGTCCCAGAAGAACAAGAGGATGC
    R CATAGCTGAGGAGTCCTTGCAATGGCCTTCC
    Sequencing primers of the Seq1 AGGGGCCGAGGGAAACAAATGAAGA
    A1AT 2-kb region Seq2 GCTAAAGATGACACTTATTTTGGAA
    Seq3 ATGTGACAGGGAGGGAGAGGATGTG
    Seq4 GAAAAGTGGTGAATCCCACCCAAA
    Seq5 GCCCATCTGTTTCTGGAGGGCTCCA
    Seq6 TCACCTAACCAGACTCGGGCCCTGC
    Seq7 TACCAGGGTGCAACAAGGTCGTCAG
    Construction of the A1AT-donor Left arm-F GGCGCGCCTCTGCACGACAGGTCTGCCAGCTTAC
    template vector Left arm-R AAGAATGCATGCGTCAATTTTACGCAGACTATCTTTCTAGGGTTAACACAGCCTTATGC
    ACGGCCTGGAGGGGAG
    Right arm-F TTAACGTACGTCACAATATGATTATCTTTCTAGGGTTAACCATCGACGAGAAAGGGACT
    GAAGCTGCTGG
    Right arm-R CCTTAATTAATGGTCTTCTGGGGCCTGCTGGGGC
    Genotyping after ZFN- V4 TGGAGTGACGATGCTCTTCCCTGTTC
    mediated targeting L1 GGTCAATGGGTGATGTGCTTCCTCTC
    PB5P2 CGTCAATTTTACGCATGATTATCTTTAAC
    Detection of backbone F ACACAGGAAACAGCTATGACCATGATTACG
    integration R CGTCAATTTTACGCATGATTATCTTTAAC
    Genotyping after transposon U4 TGGAGTGACGATGCTCTTCCCTGTTC
    excision L2 GCAGTTATTTTTGGGTGGGATTCAC
    PB-P2 GCGACGGATTCGCGCTATTTAGAAAG
    Detection of transposon re- F CTGCTGCAACTTACCTCCGGGATG
    integration R CCAATCCTCCCCCTTGCTGTCCTG
    A1AT
     5′ probe F GATTCCCCAACCTGAGGGTGACCAAG
    R AAGTGACAGAGAAACGCAAGCCTTC
    A1AT
     3′ probe F CGAGGGTCACGCTAAACTTCTGCAG
    R CTCTGGGAGGTTATGCTAGTTTGAA
    Surveyor Nuclease assay F AGGAGCAAGGCCTATGTGA
    R GAGGAGCGAGAGGCAGTTAT
  • a Het., clones heterozygous for PB allele. b Homo./Hemi., clones homozygous or hemizygous for PB allele. Cells with one targeted allele and deletion of the other allele are undistinguishable from correctly targeted homozygous clones by PCR. Such cells are designated as hemizygotes. c Vector backbone integration was analyzed by PCR. d Clones showing incorrect PCR bands are included. e A sum of 2 independent experiments.
  • Methods
  • Plasmid Construction
  • Gateway-adapted piggyBac transposon vectors: A destination vector pPB-R1R2-NP was constructed as follows. The attR1 and attR2 sites were PCR-generated and digested by NheI/HindIII and XhoI/SpeI, respectively. EM7-neo was PCR-generated and digested by HindIII/XhoI. These 3 fragments were then cloned into the NheI-SpeI site of pPB-LR527, resulting in pPB-R1R2-Neo. An EcoRI-XbaI fragment containing PheS was excised from pR6K-R1R2-ZP28, blunt-ended and cloned into the blunted XhoI site of pPB-R1R2-Neo, resulting in pPB-R1R2-NP. An entry vector pENTR-PGKpuroΔtk was constructed by cloning a KpnI-NotI PGK-puroitk fragment into the KpnI-NotI site of pENTR-2B.
  • A targeting vector for Tyr: The targeting vector was constructed using BAC recombineering. A BAC clone RP24-221M7 was introduced into Escherichia coli
  • strain EL35029. A mini targeting vector was first constructed to modify the Tyr gene on the BAC. Left and right homology arms were PCR-generated and digested by AscI/BsiWI and NsiI/PacI, respectively. The transposon fragment was excised from pPB-R1R2-NP by NsiI/BsiWI digestion. These 3 fragments were then cloned into AscI/Pact site of pMCS, resulting in pMCS-Tyr-NP. An AscI-PacI fragment was excised from pMSC-Tyr-NP and used for BAC targeting. A retrieving vector was constructed by cloning PCR-generated left and right homology arm into the XhoI/AscI site of pMSC-DTA, following AscI/HindIII and XhoI/HindIII digestion of the left and right arm, respectively. The retrieving vector was linearized by HindIII digestion and used to retrieve 3.0-kb 5′ arm, the transposon and 6.5-kb 3′ arm. Finally, the Neo-PheS cassette was replaced with the PGK-puroΔtk cassette by Gateway cloning, resulting in pDTA-TyrPB. The targeting vector was linearized by AscI prior to electroporation into the albino mIPSCs. A donor template vector for A1AT: A 2-kb fragment, which contained 1 kb at both side of Z mutation, was first PCR-amplified using genomic DNA from A1ATD-hIPSC line B as a template and cloned into pCR4-blunt-TOPO (Invitrogen), resulting in pCR4-AAT_Z. To construct a donor template with corrected sequence and a piggyBac transposon, the 5′ arm and 3′ arm were PCR-amplified and digested with AscI/NsiI and BsiWI/PacI, respectively. The NsiI-BsiWI fragment containing a piggyBac transposon with the Neo-PheS cassette was excised from pPB-R1R2-NP. The digested fragments were cloned into the AscI-Pact site of pMCS, resulting in pMCS-AAT-PB:NP. The Neo-PheS cassette was subsequently replaced with a PGK-puroΔtk cassette by Gateway cloning, resulting in the final donor vector, pMCS-AAT-PB:PGKpuroΔtk.
  • The plasmids (pPB-R1R2-NP, pENTR-PGKpuroΔtk, pMCS-AAT-PB:PGKpuroΔtk) have been deposited in the Wellcome Trust Sanger Institute Archives and available upon request (http://www.sanger.ac.uk/technology/clonerequests/).
  • Cell Culture
  • Appropriate ethical approval and patient consent have been obtained (Ethics reference no. 08/H0311/201; R&D no. A091485). A1ATD-hIPSCs (ref. 16; A, patient 2 line 1; B, patient 1 line1; C, patient 3 line1) were cultured on MEF-feeder layers in hESC medium: DMEM/F12 supplemented with 20% knockout serum replacement, 1 mM GlutaMax, 0.1 mM 2-mercaptoethanol, 1× non-essential amino acid and 4 ng ml-1 FGF2 (Invitrogen). Subculture was performed every 5-7 days by detaching hIPSCs by incubation in 0.5 mg ml-1 dispase and 0.5 mg ml-1 collagenase type IV for 1 hr at 37 ° C., collecting detached hIPSC colonies, breaking down into small clamps and plating them onto new feeder plates. Mouse embryonic fibroblasts (CF1 or B6129F1) were cultured in DMEM containing 10% FCS, 2 mM Glutamine, 0.1 mM 2-melcaptoethanol and 1× non-essential amino acid. mIPSCs (iPS25A1; ref. 15) were cultured on MEF-feeder layers in mESC medium: KO-DMEM supplemented with 15% FBS, 1 mM GlutaMax, 0.1 mM 2-mercaptoethanol, 1× non-essential amino acid and 1000 unit ml-1 LIF (Millipore).
  • Gene Targeting and Transposon Excision in Mouse IPSCs
  • 1×107 cells were electroporated with 25 μg of a linearized targeting vector in 800 μl of HEPES-buffered saline using a Gene Pulser II electroporator (230 V, 500 μF) and plated onto three 10-cm dishes. The next day, puromycin selection (1 μg ml-1) was initiated. Resulting colonies were picked and screened by PCR. Targeted clones were expanded and further verified by Southern blot analysis. Correctly targeted clones were then subjected to transposon excision. 2×106 cells were electroporated with 40 μg of pCMV-hyPBase in 800 μl of HEPES-buffered saline using a Gene Pulser II electroporator (230 V, 500 μF) and plated onto one well of a 6-well plate. After passage once, cells were replated on day 4 at 5×105 cells per 10-cm dish. On the following day, FIAU (0.2 μM) selection was initiated. On day 5 of selection, FIAU was withdrawn. Resulting colonies were picked at day 7 and screened by PCR. Primer sequences to detect homologous recombination are shown in Table 7.
  • ZFNs-Mediated Gene Targeting in A1ATD-hIPSCs.
  • On the day of electropolation (day 0), near-confluent cells were pre-treated with a ROCK inhibitor26 (Y-27632, Sigma) at 10 μM for 3-4 hrs prior to electroporation. Cells were then washed with PBS once, detached by Accutase (Millipore; 10 min at 37° C.) and mixed with DMEM/F12 containing 10% FCS. Cells were dissociated into single-cell suspension by vigorous pipetting and counted. 2×106 cells were pelleted and mixed with 5 μg of a 5′-ZFN expression vector, 5 μg of a 3′-ZFN expression vector and 2 μg of the donor template in 100 μl of hESC solution 1 (Lonza). The cell suspension was transferred to a cuvette and electroporated using the Amaxa Nucleofector device (Lonza) with program A23. The electroporated cells were plated onto one or two 10-cm feeder dishes in MEF-conditioned hESC medium containing 10 μM Y-27632. hESC medium without any drug was used for daily medium change between day 1-3. On day 4, puromycin selection (1 μg ml-1) was started. On day 6, medium was changed to MEF-conditioned hESC medium containing 0.5 μg ml-1 puromycin, which was used for medium change at every other day until picking colonies. Resulting colonies were picked on day 13-17. Colonies was cut into 2 pieces. One half was transferred onto a well of 24-well feeder plate and the other half was lysed and used for PCR-genotyping. PCR-positive clones were further expanded and homologous recombination was verified by Southern blot analysis.
  • Transposon Excision in Homozygously Targeted hIPSCs.
  • Homozygously targeted clones (B-16, C-G4, SeV-1-C3 and SeV-3-G5) were used for transposon removal. Line A-derived clones were omitted because this line displayed a lower capability of differentiating into endodermal lineages. Cells were prepared as described above. 2×106 cells were mixed with 10 μg of the hyperactive piggyBac transposase expression vector (pCMV-hyPBase8) in 100 μl of hESC solution 1 and electroporated using the Nucleofector device with the program A23. Electroporated cells were plated onto 6-well plate in 1:2, 1:3 and 1:6 dilutions in MEF-conditioned hESC medium containing 10 μM Y-27632. Note that ROCK inhibitor was added to the culture medium until day 6 in this experiment. On day 2, cells with ˜80% confluency were passaged using Accutase at a split ratio 1:2, 1:3 and 1:6 into 6-well plates. On day 4, cells with ˜80% confluency were washed with PBS, detached with Accutase, suspended in hESC medium and pelleted. Cells were resuspended in hESC medium into single-cell level and counted. 1×104 cells were then plated onto one 10-cm dish in hESC medium containing 10 μM Y-27632. 16-18 hrs after plating (day 5), medium was changed to hESC medium containing 0.25 μM FIAU and 10 μM Y-27632. On day 6, medium was changed to hESC medium containing 0.25 μM FIAU and then medium was changed every other day. Genotype and deletion of the piggyBac transposon were analyzed by PCR and further verified by Southern blot analysis.
  • CGH Analysis
  • Genomic DNA was extracted using a DNeasy kit (Qiagen). Agilent 244K human genome arrays were used following the manufacturer's protocol. The arrays were scanned with an Agilent microarray scanner and data were generated by Agilent Feature Extraction software. CGH calls were made with Agilent's DNA analytics software using the ADM2 algorithm (6.0 threshold) with a minimum of 5 probes in the region as a filter.
  • SNP Analysis
  • An Illumina HumanCytoSNP-12 SNP array was used following the manufacturer's protocol. Genotype calls were performed by Illumina's GenomeStudio. B allele frequency and log R ratio were analyzed by KaryoStudio. CNVpatition v2.4.4 bundled in KaryoStudio was used for copy number analysis.
  • ZFN Design
  • ZFNs were designed against a region containing Z mutation in the A1AT gene (see FIGS. 2 a, b) and assembled as previously described30. The amino acid residues at positions ‘−1’ to ‘+6’ of the recognition alpha helix31,32 of each of the zinc finger DNA-binding domain for each DNA triplet target are shown in Table 4. The ZFNs were linked to wild type FokI catalytic domain. The activity of the ZFN at the endogenous target site was determined using the Surveyor Nuclease assay as previously described33.
  • hIPSCs-derived hepatocyte-like cell transplantation in immunodeficient uPA transgenic mice
  • All mice were housed in pathogen-free conditions and animal studies were approved by the committee on animal experimentation of the Institut Pasteur and by the French Ministry of Agriculture. Differentiated cells (5×105 cells per animal in 50 μl DMEM) were injected into the spleens of 3- to 4-week-old Alb-uPA+/+;Rag2−/−;II2rg−/− mice (n=7). The recipient mouse was sacrificed 2 weeks after transplantation for histological analysis. Blood samples were collected and human albumin in plasma was quantified by ELISA (Bethy Laboratories). Frozen liver sections were analyzed by immunofluorecence with human albumin (Dako) or human A1AT (Dako) specific antibodies. Non-transplanted mice were used as controls.
  • Exome Sequencing
  • The corrected iPSC line, B-16-C2, and its parental fibroblasts were analyzed. Exome sequencing and analysis were performed as described previously34 with minor modification. Exome pulldown was performed using an Agilent SureSelect Human All Exon 50 Mb Kit according to the manufacturer's instructions. Enriched DNA was sequenced on an Illumina HiSeq2000 (75-bp paired-end sequencing). 90.32% (Fibroblast-B) and 90.72% (B-16-C2) of total targeted regions were covered with more than 10× sequencing depth, covering 93.01% and 93.35% of CCDS exons, respectively. Substitutions in the coding sequence were called as positions with at least 20% of reads reporting a different base with respect the reference human sequence (GRCh37). Additionally, somatic mutations were identified by comparing the sequence with the control fibroblasts, and removing the common polymorphisms described in dbSNP and in the 1000 Genomes Project35. Small insertions and deletions were identified using samtools, as the ones not present in the control cell line and that had at least 20× of coverage and 20% of the reads reporting the mutation. Validation of mutations was carried out by Sanger capillary sequencing on parental Fibroblast-B, A1ATD-hIPSC line B, the homozygously targeted B-16 cells and the piggyBac-excised B-16-C2 cells.
  • Sendaiviral regrogramming, RT-PCR, quantitative RT-PCR, bisulfite sequencing, immunostaining, flow cytometric analysis, ELISA and EndoH analysis
  • These experiments were performed as described previously see refs 16,24,25,36
  • 20q11.21 Amplification
  • In the initial analysis of CGH data, we found amplification of 20q11.21 in one homozygous clone, C-B6 (FIG. 7 a). Through analysis of SNP array data, however, this amplification was found in all lines derived from A1ATD-iPSC line C (FIG. 7 b). This region is found to be frequently amplified following prolonged human ESC culture37,38. We individually analyzed CGH data of the 20q11.21 region in 3 primary A1ATD-iPSC lines and 3 homozygous clones derived from line C. We first compared log2 ratios in the affected region (position 29,297,270-30,638,579; 167 probes) and those in the adjacent normal copy number region (position 30,642,607-32,056,016) that contains the same number of probes (FIG. 7 a). Consistent with the SNP array data, all three homozygous clones showed significantly higher ratios in the affected region than in the adjacent region (FIG. 7 c). However, the ratios in C-C11 and C-G4 were lower than the cut-off used for the initial CGH data analysis. C-B6 showed a much higher ratio than C-C11 and C-G4, suggesting that C-B6 carried more copies of the 20q11.21 region than the others. Surprisingly, the primary hIPSC line C also showed a slightly higher ratio in the affected region than in the adjacent region whereas line A and B, both carrying normal chromosome 20 based on the SNP analysis, had same ratios. This result strongly suggests that line C is heterogeneous with a minor population of cells with amplified 20q11.21. The mean log2 ratio of this region in line C is 0.208, which indicates 2.3 copies per diploid genome (FIG. 7 d). On the assumption that line C consists of 2 cell types (2-copy normal and 3-copy abnormal cells), 30% of the population carries 3 copies of 20q11.21. Since we used genomic DNA from line C as a reference for subsequent CGH analyses of the line C-derived cell lines, the log2 ratio of the 20q11.21 region in C-C11 and C-G4 was lower than a ratio representing single-copy gain (0.58) and thus this region was not called as a “Gain”. We recalculated the copy number for 3 homozygous clones using the corrected copy number for the line C. Normalized copy numbers of 20q11.21 were 3 copies for C-C11 and C-G4, and 3.8 for C-B6 (FIG. 7 d), which is consistent with the SNP array data. Thus, in C-B6, the 20q11.21 region was further amplified. The ZFN-stimulated gene targeting in line C was conducted several passages after CGH analysis. The abnormal cells might become dominant at the time of gene targeting. As a result all derivative lines carried 20q11.21 amplification.
  • Analysis on One Point Mutation Introduced During ZFN-Stimulated Targeting
  • As ZFN-induced mutations are primarily InDels, the one observed single base substitution is unlikely to be the result of off-target ZFN cleavage. To support this conclusion we scanned the human genome (hg19) for potential ZFN cleavage sites and calculated the distance between maximum-likelihood off-target sites and the mutation. Both heterodimers and homodimers of either site with 5 or 6bp between individual ZFN sites and up to 6 mismatches from the intended binding sites were allowed; this returned 495 potential cleavage sites. The smallest distance between any of these 495 potential cleavage sites and any of the mutation was 110,407 bp. Thus, it is extremely unlikely that ZFN cleavage at any of these putative off-target sites caused the observed mutation.
  • REFERENCES
      • 1. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861-872 (2007).
      • 2 Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920 (2007).
      • 3 Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and applications. Genes Dev 24, 2239-2263 (2010).
      • 4 Hanna, J. et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 318, 1920-1923 (2007).
      • 5 Fairchild, P. J. The challenge of immunogenicity in the quest for induced pluripotency. Nat Rev Immunol 10, 868-875 (2010).
      • 6 Tenzen, T., Zembowicz, F. & Cowan, C. A. Genome modification in human embryonic stem cells. J Cell Physiol 222, 278-281 (2010).
      • 7 Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S. & Gregory, P. D. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 11, 636-646 (2010).
      • 8 Yusa, K., Zhou, L., Li, M. A., Bradley, A. & Craig, N. L. A hyperactive piggyBac transposase for mammalian applications. Proc Natl Acad Sci U S A 108, 1531-1536 (2011). 9 Wang, W. et al. Chromosomal transposition of PiggyBac in mouse embryonic stem cells. Proc Natl Acad Sci U S A 105, 9290-9295 (2008).
      • 10 Hockemeyer, D. et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol 27, 851-857 (2009).
      • 11 van der Weyden, L., Adams, D. J. & Bradley, A. Tools for targeted manipulation of the mouse genome. Physiol Genomics 11, 133-164 (2002).
      • 12 Meier, I. D. et al. Short DNA sequences inserted for gene targeting can accidentally interfere with off-target gene expression. FASEB J 24, 1714-1724 (2010).
      • 13 Lacoste, A., Berenshteyn, F. & Brivanlou, A. H. An efficient and reversible transposable system for gene delivery and lineage-specific differentiation in human embryonic stem cells. Cell Stem Cell 5, 332-342 (2009).
      • 14 Fraser, M. J., Ciszczon, T., Elick, T. & Bauser, C. Precise excision of TTAA-specific lepidopteran transposons piggyBac (IFP2) and tagalong (TFP3) from the baculovirus genome in cell lines from two species of Lepidoptera. Insect Mol Biol 5, 141-151 (1996).
      • 15 Yusa, K., Rad, R., Takeda, J. & Bradley, A. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods 6, 363-369 (2009).
      • 16 Rashid, S. T. et al. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest 120, 3127-3136 (2010).
      • 17 Perlmutter, D. H. Autophagic disposal of the aggregation-prone protein that causes liver inflammation and carcinogenesis in alpha-1-antitrypsin deficiency. Cell Death Differ 16, 39-45 (2009).
      • 18 Gooptu, B. & Lomas, D. A. Conformational pathology of the serpins: themes, variations, and therapeutic strategies. Annu Rev Biochem 78, 147-176 (2009).
      • 19 Zou, J. et al. Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell 5, 97-110 (2009).
      • 20 Mitalipova, M. M. et al. Preserving the genetic integrity of human embryonic stem cells. Nat Biotechnol 23, 19-20 (2005).
      • 21 Baker, D. E. et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat Biotechnol 25, 207-215 (2007).
      • 22 Lefort, N. et al. Human embryonic stem cells reveal recurrent genomic instability at 20q11.21. Nat Biotechnol 26, 1364-1366 (2008).
      • 23 Spits, C. et al. Recurrent chromosomal abnormalities in human embryonic stem cells. Nat Biotechnol 26, 1361-1363 (2008).
      • 24 Touboul, T. et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 51, 1754-1765 (2010).
      • 25 Fusaki, N., Ban, H., Nishiyama, A., Saeki, K. & Hasegawa, M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc Jpn Acad Ser B Phys Biol Sci 85, 348-362 (2009).
      • 26 Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol 25, 681-686 (2007).
      • 27 Cadinanos, J. & Bradley, A. Generation of an inducible and optimized piggyBac transposon system. Nucleic Acids Res 35, e87 (2007).
      • 28 Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337-342 (2011).
      • 29 Liu, P., Jenkins, N. A. & Copeland, N. G. A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res 13, 476-484 (2003).
      • 30 Urnov, F. D. et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 435, 646-651 (2005).
      • 31 Beerli, R. R. & Barbas, C. F., 3rd. Engineering polydactyl zinc-finger transcription factors. Nat Biotechnol 20, 135-141 (2002).
      • 32 Pavletich, N. P. & Pabo, C. O. Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science 252, 809-817 (1991).
      • 33 Guschin, D. Y. et al. A rapid and general assay for monitoring endogenous gene modification. Methods Mol Biol 649, 247-256 (2010).
      • 34 Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539-542 (2011).
      • 35 Consortium, T. G. P. A map of human genome variation from population-scale sequencing. Nature 467, 1061-1073 (2010).
      • 36 Seki, T. et al. Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells. Cell Stem Cell 7, 11-14 (2010).
      • 37 Lefort, N. et al. Human embryonic stem cells reveal recurrent genomic instability at 20q11.21. Nat Biotechnol 26, 1364-1366 (2008).
      • 38 Spits, C. et al. Recurrent chromosomal abnormalities in human embryonic stem cells. Nat Biotechnol 26, 1361-1363 (2008).

Claims (17)

1. A method .of modifying the genome of a cell, the method comprising recombining into the genome of the cell a nucleic acid, the exogenous nucleic acid comprising
(i) a modifying sequence and
(ii) a selection element flanked by transposon inverted repeats,
the method further comprising the steps of:
(a) selection of insertion of the nucleic acid into the genome using the selection element, and
(b) use of a transposase to excise the selection element, such that after transposase mediated excision the only exogenous nucleic acid remaining in the genome is the modifying sequence
2. A method of modifying according to claim 1 wherein the modification is correcting or introducing a mutation in a genome of a cell, wherein the modifying sequence comprises nucleic acid which corrects or introduces the mutation, respectively, and wherein after the transposon mediated excision process the genome of the corrected cell is identical to the genome of the original cell with the only exceptions being the mono or biallelic correction of the mutation or mono or biallelic introduction of the mutation.
3. A method according to claim 1 or 2 comprising an additional step of providing to the cell a Zinc Finger Nucleases (ZFNs) or Transcription activator like effector nuclease (TALES) to increase efficiency of the homologous recombination of the nucleic acid into the cell genome.
4. A method according to any preceding claim wherein the transposon sequences are from the piggyback transposon.
5. A method according to any preceding claim 1 wherein the transposase used to excise the nucleic acid comprising the selectable element is a piggyBac transposase.
6. A method according to any preceding claim wherein the cell is an iPS cell from an individual having a genomic mutation associated with disease.
7. A method according to any preceding claim wherein the modification is the correction of a point mutation.
8. A method according to any preceding claim wherein modification is of one allele of a gene.
9. A method according to any preceding claim wherein modification is simultaneous modification of both alleles of a gene.
10. A method according to any preceding claim wherein the method provides at least 40% efficiency in targeting on one allele, determined as number of single allele modifications for cells having selected insertion of a selectable marker.
11. A method according to any preceding claim wherein the method provides an efficiency of at least 3% biallelic targeting, determined as number of biallelic modifications for cells having selected insertion of a selectable marker.
12. A method according to any preceding claim which is a method of modification of the genome of an iPS cell, the genome of which has a mutation associated with a disease phenotype, the method comprising delivering to the cell:
(a) an exogenous nucleic acid comprising:
(i) a modifying sequence and
(ii) a selection element flanked by piggyBac inverted repeats; and
(b) a zinc finger nuclease or TALES to enhance the efficiency of homologous recombination of the nucleic acid into the genome of the cell;
wherein delivery is followed by selection for the insertion of the nucleic acid into the genome using the selection element; and then
providing expression of a piggyBac transposase within the cell to remove the selection element, optionally followed by differentiation of the modified cell.
13. A cell comprising a genome which has been modified by insertion of a modifying sequence using a method according to claim 1-12.
14. A collection of cells, such as an organ or tissue, wherein the genomes of all, or a proportion, of the cells has been modified by insertion of a modifying sequence using a method according to claim 1-12.
15. A pair of cells, or cell lines or tissues differentiated therefrom, the genome of one of the pair having been modified from the other according to the method of any of claims 1-12.
16. A cell, or cell line or tissues differentiated therefrom, made according to any of claims 1-12, optionally in combination with the original unmodified cell, for use in the assessment of any of the following:
the effect of a therapeutic agent (e.g.drug screening);
the identification of targets for drug treatment;
the toxicological effect of a therapeutic agent;
the safety of a therapeutic agent;
the effect of an individual modification or modifications on drug activity;
the effect of an individual modification or modifications on likely patient responsiveness the effect of an individual modification or modifications on likely patient resistance;
in the assessment of which patient sub-groups will respond to drug treatments.
17. A method for treating a disease in an individual, the method comprising modifying the genome of a cell using the method of claims 1-12 to correct a mutation associated with the disease in the cell, and using the cell, or cells derived or differentiated therefrom, in disease treatment.
US13/594,053 2010-08-25 2012-08-24 Method for genome modification Abandoned US20130209426A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB1014169.5A GB201014169D0 (en) 2010-08-25 2010-08-25 In vitro hepatic differentiation
GB1014169.5 2010-08-25
PCT/GB2011/001275 WO2012025725A1 (en) 2010-08-25 2011-08-25 In vitro hepatic differentiation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/001275 Continuation-In-Part WO2012025725A1 (en) 2010-08-25 2011-08-25 In vitro hepatic differentiation

Publications (1)

Publication Number Publication Date
US20130209426A1 true US20130209426A1 (en) 2013-08-15

Family

ID=42984593

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/818,515 Active US9284576B2 (en) 2010-08-25 2011-08-25 In vitro hepatic differentiation
US13/594,053 Abandoned US20130209426A1 (en) 2010-08-25 2012-08-24 Method for genome modification

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/818,515 Active US9284576B2 (en) 2010-08-25 2011-08-25 In vitro hepatic differentiation

Country Status (10)

Country Link
US (2) US9284576B2 (en)
EP (1) EP2609192B1 (en)
JP (1) JP6063383B2 (en)
CN (1) CN103180436B (en)
AU (1) AU2011294869B2 (en)
CA (1) CA2841641C (en)
DK (1) DK2609192T3 (en)
GB (1) GB201014169D0 (en)
SG (2) SG10201506704XA (en)
WO (1) WO2012025725A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140335063A1 (en) * 2013-05-10 2014-11-13 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
WO2018039440A1 (en) * 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
WO2020123327A1 (en) 2018-12-10 2020-06-18 Amgen Inc. Mutated piggybac transposase
US10920242B2 (en) 2011-02-25 2021-02-16 Recombinetics, Inc. Non-meiotic allele introgression
US10959414B2 (en) 2013-08-27 2021-03-30 Recombinetics, Inc. Efficient non-meiotic allele introgression
US11339409B2 (en) * 2012-11-01 2022-05-24 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US11827900B2 (en) 2016-08-24 2023-11-28 Sangamo Therapeutics, Inc. Engineered target specific nucleases
US11851653B2 (en) * 2015-12-01 2023-12-26 Crispr Therapeutics Ag Materials and methods for treatment of alpha-1 antitrypsin deficiency

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201216796D0 (en) * 2012-09-20 2012-11-07 Cambridge Entpr Ltd In vitro pancreatic differentiation
USD1009775S1 (en) 2014-10-15 2024-01-02 Maxeon Solar Pte. Ltd. Solar panel
USD933584S1 (en) 2012-11-08 2021-10-19 Sunpower Corporation Solar panel
WO2014165663A1 (en) * 2013-04-03 2014-10-09 Cellular Dynamics International, Inc. Methods and compositions for culturing endoderm progenitor cells in suspension
US20170029778A1 (en) 2013-06-11 2017-02-02 President And Fellows Of Harvard College Sc-beta cells and compositions and methods for generating the same
GB201317869D0 (en) 2013-10-09 2013-11-20 Cambridge Entpr Ltd In vitro production of foregut stem cells
US20150166952A1 (en) * 2013-12-16 2015-06-18 Industrial Technology Research Institute Cultured hepatocyte and method for preparing the same
US9512406B2 (en) * 2013-12-20 2016-12-06 The J. David Gladstone Institute, a testamentary trust established under the Will of J. David Gladstone Generating hepatocytes
US11066649B2 (en) * 2014-03-19 2021-07-20 Institut National De La Sante Et De La Recherche Medicale (Inserm) Method for inducing human cholangiocyte differentiation
US10597633B2 (en) 2014-05-16 2020-03-24 Koninklijke Nederlandse Akademie Van Wetenschappen Culture method for organoids
CN105154386B (en) * 2014-05-30 2018-04-24 中国人民解放军第二军医大学东方肝胆外科医院 Human liver cell maintains for a long time and the special culture media and cultural method of propagation secondary culture
USD933585S1 (en) 2014-10-15 2021-10-19 Sunpower Corporation Solar panel
USD913210S1 (en) 2014-10-15 2021-03-16 Sunpower Corporation Solar panel
USD896747S1 (en) 2014-10-15 2020-09-22 Sunpower Corporation Solar panel
USD999723S1 (en) 2014-10-15 2023-09-26 Sunpower Corporation Solar panel
GB201421092D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
US10443042B2 (en) 2014-12-18 2019-10-15 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
WO2016100930A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College Methods for generating stem cell-derived b cells and methods of use thereof
WO2016100909A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College METHODS FOR GENERATING STEM CELL-DERIVED β CELLS AND USES THEREOF
EP3239293A4 (en) * 2014-12-24 2018-08-08 Kyoto University Endodermal cell production method, liver cell production method, pancreatic cell production method, endodermal cell induction promoter, liver cell induction promoting kit, pancreatic cell induction promoting kit, and micro fluid device
CN106434535B (en) * 2015-08-11 2021-05-14 中国科学院广州生物医药与健康研究院 Method for controlling stem cell differentiation
CN106754636B (en) * 2015-11-19 2019-08-30 中国人民解放军第二军医大学 External evoked primary hepatocyte bile ductization and long-term cultivation, amplification and the method and its application of differentiation
JP6756493B2 (en) 2016-02-29 2020-09-16 米満 吉和 High-performance hepatocytes and their utilization
GB201603569D0 (en) * 2016-03-01 2016-04-13 Koninklijke Nederlandse Akademie Van Wetenschappen Improved differentiation method
CN106479957B (en) * 2016-08-10 2019-11-19 上海爱萨尔生物科技有限公司 Hepatocyte cultures liquid and cultural method
US11530388B2 (en) 2017-02-14 2022-12-20 University of Pittsburgh—of the Commonwealth System of Higher Education Methods of engineering human induced pluripotent stem cells to produce liver tissue
WO2019073951A1 (en) * 2017-10-12 2019-04-18 国立大学法人東京工業大学 Method for inducing differentiation of pluripotent stem cells into hepatocytes
BR112020009275A2 (en) 2017-11-15 2020-10-27 Semma Therapeutics, Inc. islet cell manufacturing compositions and methods of use
JP2019201584A (en) * 2018-05-23 2019-11-28 国立大学法人神戸大学 Diabetes preventive agent or therapeutic agent screening method, and cell used in the method
AU2019320072A1 (en) 2018-08-10 2021-02-25 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
CN111073843A (en) * 2018-10-22 2020-04-28 立沃生物科技(深圳)有限公司 Method for maturing and amplifying liver-like cells
CN111500525B (en) * 2019-01-30 2023-05-02 中国科学院广州生物医药与健康研究院 Composition and application thereof
CN115551994A (en) 2020-03-11 2022-12-30 倍特博有限公司 Method for producing hepatocytes

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040199938A1 (en) * 2001-06-11 2004-10-07 Pascual Perez Method for obtaining a monocotyledon plant containing a gene of interest free of foreign ancillary sequence

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060270032A1 (en) 2005-05-24 2006-11-30 The Regents Of The University Of California Microscale micropatterened engineered in vitro tissue
WO2007050043A2 (en) 2005-10-24 2007-05-03 Agency For Science, Technology And Research Methods of specifying mesodermal, endodermal and mesoendodermal cell fates
EP3957716A1 (en) 2007-07-18 2022-02-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
EP2195417A1 (en) * 2007-08-24 2010-06-16 University Court of the University of Edinburgh Regionalised endoderm cells and uses thereof
WO2009090882A1 (en) 2008-01-18 2009-07-23 The University Of Tokyo Diagnosis method for non-alcoholic fatty liver disease
US20110195056A1 (en) * 2008-07-31 2011-08-11 The General Hospital Corporation Compositions comprising hepatocyte-like cells and uses thereof
WO2010049752A1 (en) 2008-10-31 2010-05-06 Katholieke Universiteit Leuven Optimized methods for differentiation of cells into cells with hepatocyte and hepatocyte progenitor phenotypes, cells produced by the methods, and methods for using the cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040199938A1 (en) * 2001-06-11 2004-10-07 Pascual Perez Method for obtaining a monocotyledon plant containing a gene of interest free of foreign ancillary sequence

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Keng et al 2009, Genetics 183:1565-157. *
Li et al., 2009, J. Microbiol. Methods 79:220-226. *
Maeder et al 2008, Molecular Cell 31: 294-301. *
VandenDriessche et al 2009 May, Blood 114:1461-1468. *
Woltjen et al., 2009, Nature 458:766-770. *
Yusa et al., 2009, Nature Methods 6:363-369. *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10920242B2 (en) 2011-02-25 2021-02-16 Recombinetics, Inc. Non-meiotic allele introgression
US11339409B2 (en) * 2012-11-01 2022-05-24 Factor Bioscience Inc. Methods and products for expressing proteins in cells
US20140335063A1 (en) * 2013-05-10 2014-11-13 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US10604771B2 (en) * 2013-05-10 2020-03-31 Sangamo Therapeutics, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US10959414B2 (en) 2013-08-27 2021-03-30 Recombinetics, Inc. Efficient non-meiotic allele introgression
US11477969B2 (en) 2013-08-27 2022-10-25 Recombinetics, Inc. Efficient non-meiotic allele introgression in livestock
US11851653B2 (en) * 2015-12-01 2023-12-26 Crispr Therapeutics Ag Materials and methods for treatment of alpha-1 antitrypsin deficiency
WO2018039440A1 (en) * 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
US11827900B2 (en) 2016-08-24 2023-11-28 Sangamo Therapeutics, Inc. Engineered target specific nucleases
US11845965B2 (en) 2016-08-24 2023-12-19 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
WO2020123327A1 (en) 2018-12-10 2020-06-18 Amgen Inc. Mutated piggybac transposase

Also Published As

Publication number Publication date
JP2013535980A (en) 2013-09-19
EP2609192A1 (en) 2013-07-03
GB201014169D0 (en) 2010-10-06
AU2011294869B2 (en) 2016-12-15
CN103180436B (en) 2017-09-22
US9284576B2 (en) 2016-03-15
CN103180436A (en) 2013-06-26
AU2011294869A1 (en) 2013-03-21
CA2841641C (en) 2019-12-03
JP6063383B2 (en) 2017-01-18
WO2012025725A1 (en) 2012-03-01
CA2841641A1 (en) 2012-03-01
DK2609192T3 (en) 2018-03-05
US20130156743A1 (en) 2013-06-20
EP2609192B1 (en) 2017-12-20
SG188288A1 (en) 2013-04-30
SG10201506704XA (en) 2015-10-29

Similar Documents

Publication Publication Date Title
US20130209426A1 (en) Method for genome modification
US11674158B2 (en) Methods and compositions for genome editing in non-dividing cells
Xie et al. Seamless gene correction of β-thalassemia mutations in patient-specific iPSCs using CRISPR/Cas9 and piggyBac
Aida et al. Cloning-free CRISPR/Cas system facilitates functional cassette knock-in in mice
US20190134227A1 (en) Generation of genetically engineered animals by crispr/cas9 genome editing in spermatogonial stem cells
US20190185850A1 (en) Single guide rna/crispr/cas9 systems, and methods of use thereof
KR101773782B1 (en) Methods and compositions for the targeted modification of a genome
Sato et al. Genome editing in mouse spermatogonial stem cell lines using TALEN and double-nicking CRISPR/Cas9
Yumlu et al. Gene editing and clonal isolation of human induced pluripotent stem cells using CRISPR/Cas9
CN105658805B (en) RNA-guided gene editing and gene regulation
US8598328B2 (en) Tol1 factor transposase and DNA introduction system using the same
Wu et al. Generation of healthy mice from gene-corrected disease-specific induced pluripotent stem cells
KR20190140950A (en) Human genetic correction
US20230193321A1 (en) Methods for increasing the efficiency of homology directed repair (hdr) in the cellular genome
WO2010117464A1 (en) Targeted integration into stem cells
Chenouard et al. Advances in genome editing and application to the generation of genetically modified rat models
JP2020527938A (en) Methods and means for genetically modifying the genome using designer DNA recombinant enzymes
Brandes et al. DGK and DZHK position paper on genome editing: basic science applications and future perspective
EP3666898A1 (en) Gene knockout method
EP3792347A1 (en) Method for producing homozygous cells
US11959094B2 (en) Methods and compositions for genome editing in non-dividing cells
Suzuki et al. Seamless Gene Correction in the Human Cystic Fibrosis Transmembrane Conductance Regulator Locus by Vector Replacement and Vector Insertion Events
US20220154184A1 (en) Increasing genome stability and reprogramming efficiency of induced pluripotent stem cells
Yusa et al. Targeted gene correction of α1-antitrypsin deficiency
Kouskoff et al. OPEN ACCESS EDITED AND REVIEWED BY

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENOME RESEARCH LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRADLEY, ALLAN;YUSA, YOSUKE;REEL/FRAME:030383/0625

Effective date: 20130321

Owner name: CAMBRIDGE ENTERPRISE LTD, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RASHID, SHEIK TAMIR;REEL/FRAME:030383/0630

Effective date: 20130411

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION