WO2009092042A1 - Reprogrammation de cellules progénitrices ou somatiques différenciées au moyen d'une recombinaison homologue - Google Patents

Reprogrammation de cellules progénitrices ou somatiques différenciées au moyen d'une recombinaison homologue Download PDF

Info

Publication number
WO2009092042A1
WO2009092042A1 PCT/US2009/031334 US2009031334W WO2009092042A1 WO 2009092042 A1 WO2009092042 A1 WO 2009092042A1 US 2009031334 W US2009031334 W US 2009031334W WO 2009092042 A1 WO2009092042 A1 WO 2009092042A1
Authority
WO
WIPO (PCT)
Prior art keywords
gene
cell
nucleic acid
acid construct
polynucleotide sequence
Prior art date
Application number
PCT/US2009/031334
Other languages
English (en)
Inventor
Yupo Ma
Original Assignee
Nevada Cancer Institute
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 Nevada Cancer Institute filed Critical Nevada Cancer Institute
Publication of WO2009092042A1 publication Critical patent/WO2009092042A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • 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
    • C12N2501/60Transcription factors
    • C12N2501/602Sox-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
    • C12N2501/60Transcription factors
    • C12N2501/603Oct-3/4
    • 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
    • C12N2501/60Transcription factors
    • C12N2501/604Klf-4
    • 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
    • C12N2501/60Transcription factors
    • C12N2501/606Transcription factors c-Myc
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES
    • C12N2840/206Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES having multiple IRES

Definitions

  • the invention relates generally to the genetic and epigenetic reprogramming of a differentiated cell using homologous recombination, and more specifically to reprogramming cells to confer a phenotype similar to progenitor cells of a given lineage or embryonic stem cells.
  • iPS induced pluripotent stem
  • mice displaying a phenotype similar to human sickle cell anemia were cured of the disease through somatic cell reprogramming and directed differentiation into blood cell progenitor populations.
  • Current studies have used retroviral delivery of the reprogramming genes into the genomic DNA, which may have deleterious effects because retroviral delivery causes random insertion of the reprogramming genes into the genome, raising the possibility that this delivery could insert into the coding sequence of a vital gene, blocking its expression.
  • the invention relates generally to the reprogramming of a differentiated or incompletely differentiated cell to a phenotype that is more primitive than that of the initial cell using homologous recombination.
  • the invention contemplates a method for directing insertion of the gene or genes responsible for reprogramming the somatic cell by homologous recombination such that the site of insertion within the genome is a pre-determined insertion site and such that the insertion event does not have an adverse effect upon the recipient cell.
  • the invention provides a nucleic acid construct for targeted delivery of genes capable of inducing pluripotency in a somatic cell through homologous recombination with the genome of the somatic cell, such that the nucleic acid is directed to a pre-determined insertion site in the genome that will not result in adverse effects upon the recipient somatic cell.
  • the nucleic acid construct includes, in 5' to 3' orientation, a first polynucleotide sequence capable of homologous recombination with a first region of a target polynucleotide sequence, a second polynucleotide sequence encoding an expression cassette including at least one gene that induces pluripotency, and a third polynucleotide sequence capable of homologous recombination with a second region of the target polynucleotide sequence.
  • the expression cassette further includes in operable linkage to the gene that induces pluripotency a promoter and a translation initiation site, hi various aspects, the expression cassette further includes a selectable marker, such as a lethal gene.
  • the gene or genes capable of inducing pluripotency may be one or more of a SOX family gene, a KLF family gene, a MYC family gene, SALL4, OCT4, NANOG, LIN28, NOBOX, STELLA, Esrrb or a STAT family gene.
  • STA T family members may include, for example STATl, STAT2, STATS, STAT4, STAT5 (STAT5A and STAT5B), and STAT6 .
  • the cassette includes four genes capable of inducing pluripotency, such as OCT4, SOX2, KLF4 and C-MYC, wherein a translation initiation site is spaced between each of the genes.
  • the invention provides a vector including a nucleic acid construct for targeted delivery of genes capable of inducing pluripotency in a somatic cell through homologous recombination with the genome of the somatic cell.
  • the nucleic acid construct includes, in 5' to 3' orientation, a first polynucleotide sequence capable of homologous recombination with a first region of a target polynucleotide sequence, a second polynucleotide sequence encoding an expression cassette including at least one gene that induces pluripotency, and a third polynucleotide sequence capable of homologous recombination with a second region of the target polynucleotide sequence.
  • the expression cassette further includes in operable linkage a promoter and a translation initiation site.
  • the expression cassette further includes a selectable marker, such as a lethal gene.
  • the invention provides a method of generating an induced pluripotent stem (iPS) cell.
  • the method includes introducing a nucleic acid construct of the present invention into a somatic cell.
  • Introduction of the construct into the somatic cell allows integration of the construct into the somatic cell genome through homologous recombination and expression of at least one gene that induces pluripotency, thereby reprogramming the somatic cell and generating an induced pluripotent stem (iPS) cell.
  • the introduction and integration of the nucleic acid construct into the somatic cell is performed using a non-viral based transfection technique.
  • integration of the construct results from targeted homologous recombination with introduction of the construct into the genome of the host cell using a non- viral-mediated transfer technique, such as, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magnetofection, liposome mediated transfer, microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer, or cell fusion.
  • a non- viral-mediated transfer technique such as, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magnetofection, liposome mediated transfer, microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer, or cell fusion.
  • the present invention provides an induced pluripotent stem (iPS) cell produced using the methods described herein.
  • iPS induced pluripotent stem
  • the present invention provides a population of induced pluripotent stem (iPS) cells produced using the methods described herein.
  • iPS induced pluripotent stem
  • the present invention provides a method of treating a subject with induced pluripotent stem (iPS) cells.
  • the method includes obtaining a somatic cell from a subject, reprogramming the somatic cell into an induced pluripotent stem (iPS) cell using the methods described herein, culturing the iPS cell under condtions that allow the iPS cell to differentiate into a desired cell type suitable for treating a condition, and introducing into the subject the differentiated cell, thereby treating the condition.
  • iPS induced pluripotent stem
  • Figure 1 is an illustrative drawing of a nucleic acid construct including a single gene of interest that may be introduced via targeted homologous recombination into the genome of a somatic cell.
  • the construct includes an expression cassette including a promoter, the gene of interest, and a drug resistance gene.
  • FIG. 2 is an illustrative drawing of a homologous reprogramming cassette for somatic cell reprogramming.
  • the cassette is configured for targeted integration into genomic DNA (gDNA) at the SALL4 locus by incorporation of flanking gDNA sequences capable of homologous recombination and integration at the SALL4 locus target.
  • the construct includes a cytolomegalo virus (CMV) promoter which is a constitutively active promoter in most cell types and used to regulate transcription of SALL4.
  • CMV cytolomegalo virus
  • the construct further includes a translation initiation site (an internal ribosome entry site or IRES) which is used to regulate expression of the drug resistance gene (Neomycin) from the CMV promoter.
  • IRES internal ribosome entry site
  • Figure 3 is an illustrative drawing of a nucleic acid construct including multiple genes of interest that may be introduced via targeted homologous recombination into the genome of a somatic cell.
  • the construct includes an expression cassette including multiple genes of interest under the control of a promoter.
  • Figure 4 is an illustrative drawing of the cloning strategy used for construction of a targeting vector using the Gateway® Cloning System. The system allows three different vectors to be used in a recombination reaction that correctly and specifically orients each arm in the final targeting vector.
  • Figure 5 is an illustrative drawing of a nucleic acid construct generated to reprogram somatic cells using a homologous recombination approach for targeted integration at the OCT4 loci of the host cell genome.
  • the present invention is based on innovative nucleic acid constructs and an approach involving homologous recombination to reprogram differentiated or semi- differentiated cells to a phenotype that is more primitive than that of the initial cell.
  • the present invention provides an approach involving homologous recombination to reprogram differentiated or incompletely differentiated cells to a phenotype that is more primitive than that of the initial cell without requiring retroviral delivery.
  • This may include, but is not limited to, the introduction of promoter regions (be they activating, inducible, or inhibiting) upstream of endogenous genes, the introduction of drug selection cassettes, or the introduction of entire expression cassettes that include not only promoter regions but also the coding sequences for one or more genes using homologous recombination for the purpose of reprogramming cells to confer a phenotype similar to progenitor cells of a given lineage (as a non-limiting example, hematopoietic stem cells) or of embryonic stem cells.
  • the present invention is based on the innovative concept of reprogramming somatic or progenitor cells into iPS cells using homologous recombination.
  • recombination it is possible to introduce reprogramming genes into defined regions on the chromosomes, avoiding random insertions.
  • the recombination sites can also be sequenced to validate their exact genomic location and thus provide a much safer avenue for in vivo use.
  • differentiation into specific tissues is then possible for a variety of therapeutic purposes.
  • pluripotent cells include cells that have the potential to divide in vitro for an extended period of time (greater than one year) and have the unique ability to differentiate into cells derived from all three embryonic germ layers, namely endoderm, mesoderm and ectoderm.
  • Somatic cells for use with the present invention may be primary cells or immortalized cells.
  • Such cells may be primary cells (non-immortalized cells), such as those freshly isolated from an animal, or may be derived from a cell line (immortalized cells).
  • the somatic cells are mammalian cells, such as, for example, human cells or mouse cells. They may be obtained by well-known methods, from different organs, such as, but not limited to skin, lung, pancreas, liver, stomach, intestine, heart, reproductive organs, bladder, kidney, urethra and other urinary organs, or generally from any organ or tissue containing living somatic cells, or from blood cells.
  • Mammalian somatic cells useful in the present invention include, by way of example, adult stem cells, Sertoli cells, endothelial cells, granulosa epithelial cells, neurons, pancreatic islet cells, epidermal cells, epithelial cells, hepatocytes, hair follicle cells, keratinocytes, hematopoietic cells, melanocytes, chondrocytes, lymphocytes (B and T lymphocytes), erythrocytes, macrophages, monocytes, mononuclear cells, fibroblasts, cardiac muscle cells, other known muscle cells, and generally any live somatic cells.
  • fibroblasts are used.
  • somatic cell is also intended to include adult stem cells.
  • An adult stem cell is a cell that is capable of giving rise to all cell types of a particular tissue.
  • Exemplary adult stem cells include hematopoietic stem cells, neural stem cells, and mesenchymal stem cells.
  • Homologous recombination itself is a rather common occurrence during the process of meiosis in eukaryotic systems.
  • the process involves the alignment of highly similar DNA sequences in chromosomes, and the exchange of DNA sequences between the DNA in each of the sister chromosomes.
  • the complex series of molecular interactions is simply defined as "cross-over". When these sequences are aligned, breaks in the double strand of DNA can facilitate the swapping of genetic material.
  • iPS induced pluripotent stem
  • iPS cells successful generation of iPS cells is possible through introduction of an expression cassette consisting of a promoter, a gene or genes that induce(s) pluripotency, and a selectable marker, such as a drug resistance gene.
  • the gene of interest and drug resistance gene are preferably separated by a translation initiation site (TIS), such as for example, an internal ribosome entry site (IRES), to allow for expression of both genes to be preferably controlled by the same promoter ( Figure 1).
  • TIS translation initiation site
  • IRS internal ribosome entry site
  • Figure 1 By expressing the gene of interest from the cassette it is possible to reprogram the somatic cell into an iPS cell.
  • SALL4 loci Figure 2 of a somatic cell genome an expression cassette consisting of a CMV promoter, the coding sequence for the gene SALL4 (a gene associated with pluripotency and somatic cell reprogramming), and a gene encoding resistance to the drug neomycin.
  • SALL4 a gene associated with pluripotency and somatic cell reprogramming
  • various isoforms of SALL4 are included in the invention. These include but are not limited to SALLl, SALL2, SALL3, and SALL4 as well as SALL4 mRNA spliced forms, SALUA and SALL4B.
  • Another alternative would be to construct a single insertion cassette of multiple genes and selection markers for homologous recombination.
  • a promoter would drive expression of the string of genes of interest separated by translation initiation sites as shown in the construct of Figure 3.
  • the advantages of this system are that it allows the construction of a cassette containing several coding sequences that can be inserted into the genome in a correctly oriented and specific site, and that requires only one homologous recombination event and therefore only one drug selection.
  • the invention provides a nucleic acid construct for targeted delivery of genes capable of inducing pluripotency in a somatic cell through homologous recombination with the genome of the somatic cell.
  • the nucleic acid construct includes in 5' to 3' orientation, a first polynucleotide sequence capable of homologous recombination with a first region of a target polynucleotide sequence, a second polynucleotide sequence encoding an expression cassette including at least one gene that induces pluripotency, and a third polynucleotide sequence capable of homologous recombination with a second region of the target polynucleotide sequence.
  • the expression cassette further includes in operable linkage a promoter, at least one gene that induces pluripotency, and a translation initiation site.
  • operatively linked means that two or more molecules are positioned with respect to each other such that they act as a single unit and effect a function attributable to one or both molecules or a combination thereof.
  • a polynucleotide encoding a gene can be operatively linked to a transcriptional or translational regulatory element, in which case the element confers its regulatory effect on the polynucleotide similar to the way in which the regulatory element would effect a polynucleotide sequence with which it normally is associated with in a cell.
  • polynucleotide or “nucleotide sequence” or “nucleic acid molecule” is used broadly herein to mean a sequence of two or more deoxyribonucleotides or ribonucleotides that are linked together by a phosphodiester bond.
  • the terms include RNA and DNA, which can be a gene or a portion thereof, a cDNA, a synthetic polydeoxyribonucleic acid sequence, or the like, and can be single stranded or double stranded, as well as a DNA/RNA hybrid.
  • nucleic acid molecules which can be isolated from a cell
  • synthetic polynucleotides which can be prepared, for example, by methods of chemical synthesis or by enzymatic methods such as by the polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the nucleotides comprising a polynucleotide are naturally occurring deoxyribonucleotides, such as adenine, cytosine, guanine or thymine linked to 2'- deoxyribose, or ribonucleotides such as adenine, cytosine, guanine or uracil linked to ribose.
  • a polynucleotide also can contain nucleotide analogs, including non-naturally occurring synthetic nucleotides or modified naturally occurring nucleotides.
  • Nucleotide analogs are well known in the art and commercially available, as are polynucleotides containing such nucleotide analogs.
  • the covalent bond linking the nucleotides of a polynucleotide generally is a phosphodiester bond.
  • the covalent bond also can be any of numerous other bonds, including a thiodiester bond, a phosphorothioate bond, a peptide-like bond or any other bond known to those in the art as useful for linking nucleotides to produce synthetic polynucleotides.
  • a polynucleotide comprising naturally occurring nucleotides and phosphodiester bonds can be chemically synthesized or can be produced using recombinant DNA methods, using an appropriate polynucleotide as a template.
  • a polynucleotide comprising nucleotide analogs or covalent bonds other than phosphodiester bonds generally will be chemically synthesized, although an enzyme such as T7 polymerase can incorporate certain types of nucleotide analogs into a polynucleotide and, therefore, can be used to produce such a polynucleotide recombinantly from an appropriate template.
  • genes that induce pluripotency are utilized to reprogram differentiated or incompletely differentiated cells to a phenotype that is more primitive than that of the initial cell, such as the phenotype of an iPS cell.
  • Such genes are capable of generating an iPS cell from a somatic cell upon expression of one or more such genes having been integrated into the genome of the somatic cell.
  • a gene that induces pluripotency is intended to refer to a gene that is associated with pluripotency and capable of generating a less differentiated cell, such as an iPS cell from a somatic cell upon integration and expression of the gene.
  • the expression of a pluripotency gene is typically restricted to pluripotent stem cells, and is crucial for the functional identity of pluripotent stem cells.
  • genes have been found to be associated with pluripotency and suitable for use with the present invention.
  • Such genes include, by way of example, SOX family genes (SOXl, SOX2, SOX3, SOXl 5, SOXl 8), KLF family genes (KLFl, KLF2, KLF4, KLF5), MYC family genes (C-MYC, L-MYC, N-MYC), SALL4, OCT4, NANOG, LIN28, STELLA ,NOBOX, Esrrb or a ST ⁇ r family gene.
  • STAT family members may include for example STATl, STAT2, STAT3, STAT4, STATS (STAT5A and STAT5B), and STAT6, FoxD3, UTFl, Rexl, ZNF206, Mybl2, DPPA2, ESGl, Otx2 and combinations thereof. While in some instances, use of only one gene to induce pluripotency may be possible, in general, expression of more than one gene is required to induce pluripotency. For example, two, three, four or more genes may be simultaneously integrated into the somatic cell genome as a polycistronic construct to allow simultaneous expression of such genes. In an illustrative aspect, four genes are utilized to induce pluripotency including OCT4, SOX2, KLF4 and C- MYC.
  • the potency-determining factor may be a transcription factor and may include other factors known in the art.
  • nucleic acid construct or "recombinant nucleic acid molecule” is used herein to refer to a polynucleotide that is manipulated by human intervention.
  • a recombinant nucleic acid molecule can contain two or more nucleotide sequences that are linked in a manner such that the product is not found in a cell in nature.
  • the two or more nucleotide sequences can be operatively linked and, for example, can encode multiple genes, such as genes that induce pluripotency, along with regulatory elements for controlling expression of such genes.
  • one advantage of utilizing homologous recombination for integration of the engineered nucleic acid construct of the present invention is that homologous recombination allows for targeted integration of the construct.
  • Successful targeting of the insertion site can facilitate expression of the inserted genes under appropriate circumstances and/or avoid inactivation of a vital gene as a result of a random insertion event.
  • the nucleic acid construct includes polynucleotides homologous to the targeted region of the genome of the host cell to allow a "crossover" event to occur.
  • the nucleic acid construct of the present invention includes polynucleotide sequences flanking (i.e., upstream and downstream) the expression cassette including the genes that induce pluripotency, that allow for homologous recombination to occur.
  • the construct includes a first polynucleotide sequence (e.g., the 5' homology arm) capable of homologous recombination with a first region of a target polynucleotide sequence and a third polynucleotide sequence (e.g., the 3' homology arm) capable of homologous recombination with a second region of the target polynucleotide sequence.
  • the first and third polynucleotide sequences are homologous to a first and second region of the target polynucleotide sequence, such as a region in a somatic cell genome.
  • the sequences can include a nucleotide sequence of somatic cell genomic DNA (gDNA) that is sufficient to undergo homologous recombination with somatic cell genomic DNA, for example, a nucleotide sequence comprising about 400 to 5000 or more substantially contiguous nucleotides of somatic cell genomic DNA.
  • the nucleic acid construct may be configured for homologous recombination with any locus or loci within a somatic cell genome, such as the OCT4 and/or SALL4 locus or locis.
  • the second polynucleotide encoding the expression cassette of the nucleic acid construct of the present invention further includes a selectable marker, such as, a lethal gene.
  • the expression cassette includes one or more genes that induce pluripotency in operable linkage with a selectable marker.
  • the one or more genes that induce pluripotency may be co-expressed with the selectable marker.
  • cells that are reprogrammed as a result of expression of the genes that induce pluripotency will also express a selectable phenotype determined by the selectable marker employed.
  • the selectable marker may be a gene that confers drug resistance.
  • reprogrammed cells may be easily identified by their selectable marker and may be selectively grown and proliferated while non-reprogrammed cells will be eliminated.
  • a selectable marker is a marker that, when expressed, confers upon a cell a selectable phenotype, such as, but not limited to, antibiotic resistance, resistance to a cytotoxic agent, nutritional prototrophy or expression of a surface protein. Co- expression of the selectable marker and one or more genes that induce pluripotency make it possible to identify and select reprogrammed cells in which the integrated pluripotency genes are expressed.
  • selectable marker genes are suitable for use with the present invention including, but not limited to, the neomycin resistance gene, puromycin resistance gene, guanine phosphoribosyl transferase, dihydrofolate reductase, adenosine deaminase, puromycin-N-acetyltransferase, hygromycin resistance gene, multi-drug resistance gene, and hisD gene.
  • the second polynucleotide encoding the expression cassette of the nucleic acid construct of the present invention further includes one or more promoters.
  • a promoter is intended mean a polynucleotide sequence capable of facilitating transcription of genes in operable linkage with the promoter. While several types of promoters are well known in the art and suitable for use with the present invention, in an exemplary aspect the promoter is a constitutive promoter that allows for unregulated expression in mammalian cells, such as the cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • the exogenously introduced genes that induce pluripotency may be expressed from one or more inducible promoters.
  • An inducible promoter is a promoter that, in the absence of an inducer (such as a chemical and/or biological agent), does not direct expression, or directs low levels of expression of an operably linked gene (including cDNA), and, in response to an inducer, its ability to direct expression is enhanced.
  • exemplary inducible promoters include, for example, promoters that respond to heavy metals, to thermal shocks, to hormones, and those that respond to chemical agents, such as glucose, lactose, galactose or antibiotic.
  • the second polynucleotide encoding the expression cassette of the nucleic acid construct of the present invention further includes one or more TIS (e.g., IRES).
  • TIS e.g., IRES
  • a TIS is ideally positioned between each gene to allow each gene to be driven off of a single upstream promoter.
  • a nucleic acid construct useful in a method of the invention can be contained in a vector.
  • One potential drawback of generating the nucleic acid constructs of the present invention is the construction of the targeting vector.
  • the homologous recombination step requires flanking DNA that is identical in sequence to the targeted locus, and a positive selection marker (e.g., antibiotic resistance).
  • the vector can be any vector useful for introducing a nucleic acid construct of the present invention into a somatic cell.
  • Gateway® cloning technology developed by Invitrogen Inc., enables the orienting and insertion of multiple polynucleotide fragments into a target vector in one step which is suitable for homologous recombination ( Figure 4).
  • the present invention further provides a method of generating an iPS cell.
  • the method includes introducing a nucleic acid construct of the present invention into a somatic cell to allow for integration and expression of genes that induce pluripotency to reprogram the somatic cell to an undifferentiated or less differentiated state.
  • Introduction of the construct into the somatic cell allows integration of the construct into the somatic cell genome through homologous recombination and expression of the at least one gene that induces pluripotency, thereby reprogramming the somatic cell and generating an iPS cell.
  • the introduction and integration of the nucleic acid construct into the somatic cell is performed using a non-viral based technique.
  • the method incorporates targeted integration of the nucleic acid construct of the present invention via homologous recombination with the host genome.
  • the nucleic acid construct of the present invention may be introduced into a cell using a variety of well known techniques, such as non- viral based transfection of the cell.
  • the construct is incorporated into a vector and introduced into the cell to allow homologous recombination.
  • Introduction into the cell may be performed by any non- viral based transfection known in the art, such as, but not limited to, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magnetofection, liposome mediated transfer, microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer (DEAE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like) or cell fusion.
  • Other methods of transfection include proprietary transfection reagents such as Lipofectamine , Dojindo Hilymax , Fugene JetPEI , Effectene TM and DreamFect TM.
  • reprogramming is intended to refer to a process that alters or reverses the differentiation status of a somatic cell that is either partially or terminally differentiated.
  • Reprogramming of a somatic cell may be a partial or complete reversion of the differentiation status of the somatic cell.
  • reprogramming is complete wherein a somatic cell is reprogrammed into an induced pluripotent stem cell.
  • reprogramming may be partial, such as reversion into any less differentiated state. For example, reverting a terminally differentiated cell into a cell of a less differentiated state, such as a multipotent cell.
  • the methods of the present invention further include detecting the selectable marker.
  • Reprogrammed cells may be easily identified by their selectable marker and may be selectively grown and proliferated while non-reprogrammed cells will perish.
  • Further analysis may be performed to assess the pluripotent characteristics of a reprogrammed cell.
  • the cells may be analyzed for different growth characteristics and embryonic stem cell like morphology.
  • cells may be differentiated in vitro by adding certain growth factors known to drive differentiation into specific cell types.
  • Reprogrammed cells capable of forming only a few cell types of the body are multipotent, while reprogrammed cells capable of forming any cell type of the body are pluripotent.
  • Expression profiling of reprogrammed somatic cells to assess their pluripotency characteristics may also be conducted. Expression of individual genes associated with pluripotency may also be examined. Additionally, expression of embryonic stem cell surface markers may be analyzed.
  • Detection and analysis of a variety of genes known in the art to be associated with pluripotent stem cells may include analysis of genes such as, but not limited to OCT4, NANOG, SALL4, SSEA-I, SSEA-3, SSEA-4, TRA-I -60, TRA-I -8 J, or a combination thereof.
  • the invention further provides iPS cells produced using the methods described herein, as well as populations of such cells.
  • the reprogrammed cells of the present invention capable of differentiation into a variety of cell types, have a variety of applications and therapeutic uses.
  • the basic properties of stem cells, the capability to infinitely self-renew and the ability to differentiate into every cell type in the body make them ideal for therapeutic uses.
  • the present invention further provides a method of treatment or prevention of a disorder and/or condition in a subject using iPS cells generated using the methods described herein.
  • the method includes obtaining a somatic cell from a subject and reprogramming the somatic cell into an iPS cell using the methods described herein.
  • the cell is then cultured under suitable conditions to differentiate the cell into a desired cell type suitable for treating the condition.
  • the differentiated cell may then be introduced into the subject to treat or prevent the condition.
  • One advantage of the present invention is that it provides an essentially limitless supply of isogenic or syngenic human cells suitable for transplantation.
  • the iPS cells are tailored specifically to the patient, avoiding immune rejection. Therefore, it will obviate the significant problem associated with current transplantation methods, such as, rejection of the transplanted tissue which may occur because of host versus graft or graft versus host rejection.
  • iPS cells of the present invention in bone marrow transplants, will circumvent the requirement of providing heavy immune suppression with drugs that have potentially adverse side effects to avoid rejection.
  • the iPS cells of the present invention may be differentiated into a number of different cell types to treat a variety of disorders by methods known in the art.
  • iPS cells may be induced to differentiate into hematopoetic stem cells, muscle cells, cardiac muscle cells, liver cells, cartilage cells, epithelial cells, urinary tract cells, neuronal cells, and the like.
  • the differentiated cells may then be transplanted back into the patient's body to prevent or treat a condition.
  • the methods of the present invention can also be used in the treatment or prevention of neurological diseases.
  • neurological diseases include, for example, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), lysosomal storage diseases, multiple sclerosis, spinal cord injuries and the like.
  • ALS amyotrophic lateral sclerosis
  • lysosomal storage diseases multiple sclerosis, spinal cord injuries and the like.
  • the methods of the present invention can also be used to correct mutations of single genes. These mutations account for diseases such as cystic fibrosis, hemophilia, and various cancers such as those associated with the BRCAl and BRCA2 mutations with high risk of development of breast and ovarian cancers.
  • the cells produced in the methods of the invention can be utilized for repairing or regenerating a tissue or differentiated cell lineage in a subject.
  • the method includes obtaining the reprogrammed cell as described herein and administering the cell to a subject (e.g., a subject having a myocardial infarction, congestive heart failure, stroke, ischemia, peripheral vascular disease, alcoholic liver disease, cirrhosis, Parkinson's disease, Alzheimer's disease, diabetes, cancer, arthritis, wound healing, immunodeficiency, aplastic anemia, anemia, and genetic disorders) and similar diseases, where an increase or replacement of a particular cell type/ tissue or cellular de-differentiation is desirable.
  • a subject e.g., a subject having a myocardial infarction, congestive heart failure, stroke, ischemia, peripheral vascular disease, alcoholic liver disease, cirrhosis, Parkinson's disease, Alzheimer's disease, diabetes, cancer, arthritis, wound healing, immunodeficiency, a
  • the subject has damage to the tissue or organ, and the administering provides a dose of cells sufficient to increase a biological function of the tissue or organ or to increase the number of cell present in the tissue or organ.
  • the subject has a disease, disorder, or condition, and wherein the administering provides a dose of cells sufficient to ameliorate or stabilize the disease, disorder, or condition.
  • the subject has a deficiency of a particular cell type, such as a circulating blood cell type and wherein the administering restores such circulating blood cells.
  • a single gene is used to effect cell reprogramming to ease the clinical transition of iPS cells.
  • the single gene is SALL4.
  • the genetic integration of a single gene into the host genome significantly reduces the complications associated with genetic reactivation and/or insertional mutagenesis currently encountered in the field.
  • This example illustrates the generation of a polycistronic vector construct including four genes that induce pluripotency suitable for targeted integration into a somatic cell genome via homologous recombination.
  • the present example illustrates the design and execution of homologous recombination based cellular retrodifferentation for therapeutic purposes.
  • the genes OCT4, SOX2, KLF4, and c-MYC are able to reprogram fetal fibroblast cells to confer a stem cell-like phenotype.
  • other genes may also be utilized to reprogram somatic and progenitor cells using a similar vector design.
  • Classical cloning techniques were used to design and create a fragment of these four genes driven by the cytomegalovirus (CMV) promoter and separated by an internal ribosomal entry site (IRES).
  • CMV cytomegalovirus
  • IVS internal ribosomal entry site
  • the CMV promoter drives expression of nearly any gene of interest in eukaryotic systems while IRES allows for translation of multiple proteins driven from one promoter by serving as a type of translation initiation site.
  • This method was utilized primarily because it only requires the insertion of exogenous sequence into one loci, and therefore, requires only one drug selection (in this case hygromycin). Selecting the endogenous loci just downstream of the transcription start site of OCT4, present in the genomic DNA, for our targeted insertion point, both the 5'-homology arm, at a length of 3.5kb, and the 3'-homology arm, at 2.6kb, were successfully cloned.
  • the final target vector construct was made using the 4-way recombination mediated by the Gateway® Cloning System as shown in Figure 4.
  • This example illustrates the reprogramming of somatic cells by integration of a construct expressing endogenous SALL4 via homologous recombination.
  • the plasmid was electroporated into mouse tail tip fibroblasts expressing SALL4 -GFP promoter-reporter construct.
  • a SALL4 expression cassette was integrated into the SALL4 locus of the genomic DNA using homologous recombination because heterozygous SALL4 mice have no obvious phenotype.
  • ES-like clones expressed very low level of green fluorescent protein (GFP) indicating incomplete reprogramming at this stage.
  • GFP green fluorescent protein
  • Surrounding fibroblasts did not express GFP serving as the negative control.
  • the phase contrast images showed fibroblast cells and a potential iPS cell colony.
  • SALL4 within the potential iPS cell colony is suggestive of pluripotency. After 22 days post transfection (15 days in ES media), ES-like clones highly expressed GFP, indicating complete reprogramming at this stage. The phase contrast images allowed identification of fibroblast cells under different magnifications and a potential iPS cell colony expressing GFP under different magnifications. Surrounding fibroblasts did not express GFP serving as the negative control. The results indicate that SALL4 alone may be capable of reprogramming mouse fibroblast cells to pluripotency via introduction by homologous recombination.

Abstract

La présente invention a pour objet des procédés et des compositions pour la reprogrammation de cellules somatiques à un état plus primitif, comme des cellules souches pluripotentes induites, au moyen d'une recombinaison homologue. Les cellules souches pluripotentes induites, produites selon les procédés de la présente invention, sont utiles dans une variété d'applications thérapeutiques dans le traitement et la prévention de maladies et de troubles.
PCT/US2009/031334 2008-01-18 2009-01-16 Reprogrammation de cellules progénitrices ou somatiques différenciées au moyen d'une recombinaison homologue WO2009092042A1 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US2219408P 2008-01-18 2008-01-18
US61/022,194 2008-01-18
US10467108P 2008-10-10 2008-10-10
US61/104,671 2008-10-10
US11692008P 2008-11-21 2008-11-21
US61/116,920 2008-11-21

Publications (1)

Publication Number Publication Date
WO2009092042A1 true WO2009092042A1 (fr) 2009-07-23

Family

ID=40885672

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/031334 WO2009092042A1 (fr) 2008-01-18 2009-01-16 Reprogrammation de cellules progénitrices ou somatiques différenciées au moyen d'une recombinaison homologue

Country Status (1)

Country Link
WO (1) WO2009092042A1 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010019569A1 (fr) * 2008-08-12 2010-02-18 Cellular Dynamics International. Inc. Procédés de production de cellules ips
WO2013137491A1 (fr) 2012-03-15 2013-09-19 国立大学法人京都大学 Procédé pour produire un mélange de cellules cardiaques et vasculaires à partir de cellules souches pluripotentes artificielles
WO2013140927A1 (fr) 2012-03-21 2013-09-26 国立大学法人京都大学 Procédé de criblage d'agents thérapeutique et/ou prophylactique pour la maladie d'alzheimer
WO2013151186A1 (fr) 2012-04-06 2013-10-10 国立大学法人京都大学 Méthode d'induction de cellules produisant de l'érythropoïétine
WO2015083582A1 (fr) 2013-12-02 2015-06-11 国立大学法人京都大学 Agent prophylactique et thérapeutique pour maladies fgfr3, et son procédé de dépistage
US9228204B2 (en) 2011-02-14 2016-01-05 University Of Utah Research Foundation Constructs for making induced pluripotent stem cells
US9328332B2 (en) 2008-06-04 2016-05-03 Cellular Dynamics International, Inc. Methods for the production of IPS cells using non-viral approach
US9499786B2 (en) 2007-03-23 2016-11-22 Wisconsin Alumni Research Foundation Enriched population of human pluripotent cells with Oct-4 and Sox2 integrated into their genome
WO2021117886A1 (fr) 2019-12-12 2021-06-17 国立大学法人千葉大学 Préparation lyophilisée contenant des mégacaryocytes et des plaquettes

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HANNAH ET AL.: "'Treatment of Sickle Cell Anemia Mouse Model with iPS Cells Generated from Autologous Skin", SCIENCE, vol. 318, 21 December 2007 (2007-12-21), pages 1920 - 1923 *
MASUI ET AL.: "Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells", NATURE CELL BIOLOGY, vol. 9, no. 6, June 2007 (2007-06-01), pages 625 - 635 *
QI ET AL.: "''The magic of four: induction of pluripotent stem cells'' from somatic cells by Oct4, Sox2, Myc and KIf4''.", CELL RESEARCH, vol. 17, 10 July 2007 (2007-07-10), pages 578 - 580 *
WU ET AL.: "'Correction of sickle cell disease by homologous recombination in embryonic stem cells", BLOOD, vol. 108, no. 4, 15 August 2006 (2006-08-15), pages 1183 - 1188 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9499786B2 (en) 2007-03-23 2016-11-22 Wisconsin Alumni Research Foundation Enriched population of human pluripotent cells with Oct-4 and Sox2 integrated into their genome
US11898162B2 (en) 2007-03-23 2024-02-13 Wisconsin Alumni Research Foundation Reprogramming somatic cells into pluripotent cells using a vector encoding Oct4 and Sox2
US10106772B2 (en) 2007-03-23 2018-10-23 Wisconsin Alumni Research Foundation Somatic cell reprogramming
US9644184B2 (en) 2008-06-04 2017-05-09 Cellular Dynamics International, Inc. Methods for the production of IPS cells using Epstein-Barr (EBV)-based reprogramming vectors
US9328332B2 (en) 2008-06-04 2016-05-03 Cellular Dynamics International, Inc. Methods for the production of IPS cells using non-viral approach
EP3330371A1 (fr) * 2008-08-12 2018-06-06 Cellular Dynamics International, Inc. Procédés de production de cellules ips
US9175268B2 (en) 2008-08-12 2015-11-03 Cellular Dynamics International, Inc. Methods for the production of iPS cells
WO2010019569A1 (fr) * 2008-08-12 2010-02-18 Cellular Dynamics International. Inc. Procédés de production de cellules ips
US9228204B2 (en) 2011-02-14 2016-01-05 University Of Utah Research Foundation Constructs for making induced pluripotent stem cells
WO2013137491A1 (fr) 2012-03-15 2013-09-19 国立大学法人京都大学 Procédé pour produire un mélange de cellules cardiaques et vasculaires à partir de cellules souches pluripotentes artificielles
WO2013140927A1 (fr) 2012-03-21 2013-09-26 国立大学法人京都大学 Procédé de criblage d'agents thérapeutique et/ou prophylactique pour la maladie d'alzheimer
WO2013151186A1 (fr) 2012-04-06 2013-10-10 国立大学法人京都大学 Méthode d'induction de cellules produisant de l'érythropoïétine
WO2015083582A1 (fr) 2013-12-02 2015-06-11 国立大学法人京都大学 Agent prophylactique et thérapeutique pour maladies fgfr3, et son procédé de dépistage
EP3513789A2 (fr) 2013-12-02 2019-07-24 Kyoto University Agents prophylactiques et thérapeutiques pour maladies fgfr3
WO2021117886A1 (fr) 2019-12-12 2021-06-17 国立大学法人千葉大学 Préparation lyophilisée contenant des mégacaryocytes et des plaquettes

Similar Documents

Publication Publication Date Title
US20090191171A1 (en) Reprogramming of Differentiated Progenitor or Somatic Cells Using Homologous Recombination
WO2009092042A1 (fr) Reprogrammation de cellules progénitrices ou somatiques différenciées au moyen d'une recombinaison homologue
US11008550B2 (en) Use of RNA for reprogramming somatic cells
Cho et al. Induction of pluripotent stem cells from adult somatic cells by protein-based reprogramming without genetic manipulation
Miyazaki et al. Emerging methods for preparing iPS cells
US20200291368A1 (en) Improved CRISPR-Cpf1 Genome Editing Tool
JP2018521642A (ja) 合成転写因子を用いる核リプログラミングのための方法
Karow et al. Site-specific recombinase strategy to create induced pluripotent stem cells efficiently with plasmid DNA
JP2019058176A (ja) 新規方法
US20200291369A1 (en) Improved CRISPR-Cas9 Genome Editing Tool
JP2019500910A (ja) ベクターを含まない人工多能性幹細胞を作製するための方法およびベクター
GB2596660A (en) A non-toxic Cas9 enzyme and application thereof
CN108949794B (zh) 一种tale表达载体及其快速构建方法和应用
WO2021181110A1 (fr) Procédé de génération de cellules hépatiques
JP2020537522A (ja) 体細胞の増強された再プログラミング
Becerra Colorado et al. Polyplex system versus nucleofection for human skin cell transfection and effect of internal ribosome entry site sequence
CN114402072A (zh) Pcbp1生成诱导多能干细胞同时抑制肿瘤发生的用途
US20210388322A1 (en) Method for producing induced pluripotent stem cells using rna nanoparticles for cell transformation
EP2739738B1 (fr) Utilisation d'une intégrase pour l'expression génique ciblée
US20220411826A1 (en) Co-opting regulatory bypass repair of genetic diseases
CN116004520A (zh) 带双安全开关的诱导多功能干细胞及其制备方法
KR20240022421A (ko) 전자기파에 반응하는 유전자 프로모터 및 이의 활용
WO2022170007A1 (fr) Protéines de fusion pour répression transcriptionnelle basée sur crispr
EP4118188A1 (fr) Procédé de génération de cellules hépatiques
TRAN Analysis of X Chromosome Reactivation during Reprogramming

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09702306

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09702306

Country of ref document: EP

Kind code of ref document: A1