EP2542676A1 - Procédés de clonage et de manipulation de génomes - Google Patents

Procédés de clonage et de manipulation de génomes

Info

Publication number
EP2542676A1
EP2542676A1 EP10726723A EP10726723A EP2542676A1 EP 2542676 A1 EP2542676 A1 EP 2542676A1 EP 10726723 A EP10726723 A EP 10726723A EP 10726723 A EP10726723 A EP 10726723A EP 2542676 A1 EP2542676 A1 EP 2542676A1
Authority
EP
European Patent Office
Prior art keywords
genome
donor
cell
host
genomes
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.)
Withdrawn
Application number
EP10726723A
Other languages
German (de)
English (en)
Inventor
Gwynedd A. Benders
John I. Glass
Clyde A. Hutchison
Carole Lartigue
Sanjay Vashee
Mikkel A. Algire
Hamilton O. Smith
Charles E. Merryman
Vladimir N. Noskov
Ray-Yuan Chuang
Daniel G. Gibson
J. Craig Venter
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.)
Viridos Inc
Original Assignee
Synthetic Genomics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/718,911 external-priority patent/US9273310B2/en
Application filed by Synthetic Genomics Inc filed Critical Synthetic Genomics Inc
Publication of EP2542676A1 publication Critical patent/EP2542676A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1079Screening libraries by altering the phenotype or phenotypic trait of the host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1031Mutagenizing nucleic acids mutagenesis by gene assembly, e.g. assembly by oligonucleotide extension PCR
    • 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
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • 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
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts

Definitions

  • nucleic acid molecules isolated from a variety of species allows for the manipulation of the isolated nucleic acid sequences in the host.
  • the ability to engineer organisms by cloning and modifying chromosomes and genomes in exogenous hosts is limited, however, by the size limitation on nucleic acid molecules that can be transferred to species such as yeast that have tractable genetics.
  • Nucleic acids cloned by conventional methods generally contain no more than a few genes, although larger nucleic acids (e.g., DNA) have been transferred into host cells.
  • nucleic acids e.g., DNA
  • the 16 kb mouse mitochondrial genome has been cloned in E. coli (Itaya et al, Nat Methods 5, 41 (2008); Yoon and Koob, Nucleic Acids Res 31, 1407 (2003)), Bacillus subtilis (Itaya et al. , Nat Methods 5, 41 (2008); Yoon and Koob, Nucleic Acids Res 31, 1407 (2003)), and yeast (Wheeler et al, Gene 198, 203 (1997)).
  • the 139 kb maize chloroplast genome has been cloned in yeast (Gupta and Hoo, Plant Mol Biol 17, 361 (1991), and the 135 kb rice chloroplast genome has been cloned in B. subtilis (Itaya et al., Nat Methods 5, 41 (2008)). About 10% of the 1.8 Mb Haemophilus influenzae genome has been cloned as episomal elements in E. coli (Smailus et al. , Syst Synth Biol; 1, 139 (2007)). The 3.5 Mb Synechocystis PCC6803 genome was inserted in three noncontiguous regions into the B.
  • 2005/0019924 describes nucleic acids and methods for introducing prokaryotic genomes into eukaryotic cells as circular molecules and conversion into artificial chromosomes.
  • WO 02/057437 describes YAC vectors containing cytomegalovirus (CMV) genomes.
  • CMV cytomegalovirus
  • U.S. Patent No. 7,083,971 describes a recombinatorial approach and system for cloning, manipulating, and delivering large nucleic acid segments.
  • U.S. Patent Application Publication No. 2005/0003511 and Bradshaw et al. Nucleic Acids Research, 23, 4850-56 (1995) describe yeast-bacterial shuttle vectors for cloning large regions of DNA by homologous recombination.
  • the disclosed cloning and manipulation methods are limited by the size of donor nucleic acids that can be transferred into a host cell, and do not provide for manipulating and/or transferring a nucleic acid molecule propagated in a host cell back into a recipient cell that is related to the donor, nor do they address incompatibility issues among different cell types used in cloning with regard to foreign nucleic acids.
  • Additional methods are needed for cloning large nucleic acids, such as chromosomes or genomes, into alternate heterelogous hosts, for manipulating the sequences of large nucleic acids in alternate hosts, and for transferring manipulated genomes back into recipient organisms that are similar to the donor organism, for example, organisms of the same genus (for example, from prokaryotic to eukaryotic cells and back).
  • nucleic acids such as chromosomes and genomes
  • transfer of the nucleic acids between species can be toxic to host, donor, and/or recipient cells.
  • Manipulation and propagation of nucleic acids in organisms of different species, genuses, or groups and from prokaryotic to eukaryotic cells and back can also cause instability of the nucleic acids and inhibit their activation, such as expression of genes from the nucleic acids.
  • nucleic acids Provided herein are methods, nucleic acids, and systems for transfer (cloning) of donor nucleic acids into host cells, for manipulation (e.g., modification) of donor nucleic acids, e.g., within host cells, and for transplantation of modified donor nucleic acids into recipient cells.
  • the provided methods and other compositions are useful in transfer and manipulation of nucleic acids across branches of life, such as for manipulation of prokaryotic nucleic acids in eukaryotic host cells and transplant of the nucleic acids back into prokaryotic recipients.
  • the methods are useful for manipulation of donor nucleic acids of organisms having poor genetic systems by transfer into hosts having strong, well-characterized genetic systems, such as yeast.
  • the methods, nucleic acids, and systems can be used for modifying nucleic acids of intractable organisms and to manipulate and engineer large nucleic acids, including genomes, for example, to produce synthetic genomes and cells, such as cells and genomes not previously in existence in the laboratory or in nature.
  • the provided methods are useful for cloning, modifying, and transplanting nucleic acids and genomes that are larger than 300 kilobases (kb), such as genomes, including whole genomes and at least minimal genomes, and cellular, viral, and organelle genomes.
  • Donor genomes can thereby be modified in host cells to produce modified donor genomes conferring one or more
  • Methods are particularly advantageous when such modified donor genomes are difficult to produce in the original cell type harboring the donor genome, or when synthetic genomes can be quickly assembled and modified in the host cell prior to transplanting the modified genome back into the original desired cell type for production of a phenotype or product of interest.
  • compositions and methods identified and described in the present application allow for new methods of transferring nucleic acid molecules and genomes from intractable donor cells into host cells where they can be modified to alter the genotype, and thereby the phenotype, to alter the nucleic acid molecule or genome.
  • the modifidied genomes can be modified in one or more ways using the host cell's genetic machinery.
  • the provided methods also provide for isolating a modified nucleic acid molecule or genome from a host cell.
  • the isolated modified nucleic acid molecule or genome can be methylated ex vivo.
  • a recipient cell can be treated with the methods described herein to allow for transfer of a modified nucleic acid molecule or genome into the cell.
  • a modified nucleic acid molecule or genome can then be further transferred into recipient cell, thereby altering the phenotype of the recipient cell to that of the modified nucleic acid molecule or genome.
  • a method for cloning a donor genome comprising: obtaining a donor genome from a donor cell or synthesizing a donor genome as one or more fragments; and introducing the donor genome and a host vector into a heterologous host cell, wherein the donor genome and the host vector are optionally joined prior to introduction into the host cell, thereby generating a host cell comprising the donor genome comprising the host vector, and further whereinthe donor genome is an essentially intact cellular, viral, or organelle genome that is at least a minimal genome, and is greater than about 300 kb in length.
  • the donor genome is an essentially whole cellular, viral or organelle genome.
  • a donor genome and a host vector can be introduced into the host cell simultaneously or sequentially. If the donor genome and host vector are introduced into the host cell sequentially, the introduction can be in either order.
  • a donor genome can be introduced into the host cell followed by introduction of a host vector.
  • a host vector can be introduced into the host cell followed by introduction of a donor genome.
  • a host vector is joined with the donor genome prior to introduction into the host cell by transforming the host vector into a donor cell containing the donor genome.
  • the donor genome can be a single molecule.
  • a nucleic acid molecule containing a donor genome and a host vector can exist as a circular centromeric plasmid.
  • the donor genome can exist as overlapping DNA fragments.
  • a donor genome can be linearized or fragmented prior to introduction into the host cell.
  • Certain embodiments of the provided methods include further recovering the donor genome and host vector from the host cell.
  • the methods further include introducing the donor genome into a recipient cell.
  • the provided methods further include degrading or removing the genome of the recipient cell.
  • Donor genomes contemplated herein include, but are not limited to, a fungal genome, an archea genome, a cyanobacterial genome, an algal genome, a viral genome, a bacteriophage genome, an organelle genome, a mitochondrial genome, a chloroplast genome (e.g., a maize chloroplast genome, or a rice chloroplast genome), an organelle genome or a synthetic genome.
  • a fungal genome an archea genome, a cyanobacterial genome, an algal genome, a viral genome, a bacteriophage genome, an organelle genome, a mitochondrial genome, a chloroplast genome (e.g., a maize chloroplast genome, or a rice chloroplast genome), an organelle genome or a synthetic genome.
  • Host cells contemplated herein a eukaryotic cell or a prokaryotic cell.
  • Host cells include, but are not limited to, a bacterial cell, a fungal cell, an insect cell, a plant cell or an algal cell.
  • Host cells also include yeast cells.
  • a host vector described herein can be a centromeric plasmid.
  • the host vector is a yeast centromeric plasmid and the host cell is a yeast cell.
  • a host vector described herein is a vector useful for homologous recombination.
  • Any of the methods described herein can further comprise modifying the donor genome within the host cell.
  • any of the methods described herein can further include recovering the donor genome comprising the host vector from the host cell.
  • the host vector can be removed from the donor genome.
  • the methods further include introducing the recovered donor genome into a recipient cell.
  • the methods described herein can further include degrading or removing the endogenous genome of the recipient cell.
  • the endogenous genome of the recipient cell is the native genome. Where multiple rounds of modifications occur (see, for example, Figures 1 and 16), the endogenous genome of the recipient cell may be a previously modified genome or a synthetic genome.
  • the provided methods further comprise modifying the donor genome in an iterative fashion.
  • a recovered donor genome may be methylated prior to introduction into a recipient cell by methylating one or more nucleotides in the donor genome.
  • a recipient cell can be, for example, a bacterial cell, a yeast cell, a fungal cell, an insect cell, a plant cell or an algal cell.
  • a restriction endonuclease function of a recipient cell is absent, removed, or inactivated prior to introduction of the donor genome.
  • a restriction modification enzyme can be mutated in the recipient cell to render it inactive.
  • the donor genome is derived from a prokaryotic cell (either natural or synthetic) and cloned in a eukaryotic cell where it may be optionally modified, and then recovered and introduced back into a prokaryotyic cell.
  • the donor genome is derived from a bacterial cell (either natural or synthetic) and cloned in a yeast cell where it may be optionally modified, and then recovered and introduced back into a bacterial cell.
  • the methods provided herein further include introducing a second donor genome into the host cell where the second donor genome is different from the first donor genome, thereby producing a host cell containing two different donor genomes.
  • Introducing the second donor genome can occur via mating the host cell containing the first donor genome with a second host cell containing the second donor genome.
  • Introduction of the recovered donor genome into the recipient cell can phenotypically transform the recipient cell to a phenotype corresponding to the donor genome, incorporating any modifications thereto.
  • a process for making a cell which exhibits a phenotype encoded by a donor genome comprising: (a) introducing into a host cell, the donor genome and a host vector suitable for cloning the donor genome in the host cell such that a product comprising the donor genome comprising the host vector is obtained; (b) recovering the product comprising the donor genome comprising the host vector obtained in step (a) from the host cell; (c) introducing the product of (b) into a recipient cell under conditions such that the recipient cell exhibits a phenotype encoded by the product; and (d) recovering the cell resulting from step (c); wherein the donor genome is an essentially intact cellular, viral, or organelle genome that is at least a minimal genome, and is greater than about 300 kb in length and comprises the minimal components of nucleic acid material necessary for the recipient cell to exhibit the phenotype encoded by the donor genome.
  • the method further comprises modifying the donor genome of (a) within the host cell. In another aspect, the method further comprises degrading or removing an endogenous genome of the recipient cell. In yet another aspect, the method further comprises modifying the donor genome of (a) within the host cell and degrading or removing an endogenous genome of the recipient cell. In certain embodiments, such processes may be automated. [0028] Provided herein is a cell which exhibits a desired phenotype encoded by a donor genome and not otherwise exhibited by the cell, wherein the cell is produced by the processes described herein.
  • a cell which comprises a donor genome and exhibits a desired phenotype encoded by the donor genome and not otherwise exhibited by the cell, wherein the donor genome comprises greater than 300 kb of foreign genomic nucleic acid material and the minimal components of a genome necessary for the cell to exhibit the desired phenotype.
  • Desired phenotypes may include the production of a expression product not native to the original cell, or modified (e.g, selective or regulated expression) or up- regulation of an existing expression product.
  • the provided methods also include cloning a plurality of genomes in a plurality of host cells.
  • the plurality of genomes can be genomic variants.
  • introducing the plurality of genomes into host cells comprises introducing host vectors and a plurality of variant overlapping fragments into the host cells, thereby generating a
  • the provided methods comprise introducing the donor genome into a recipient cell and the recipient cell supports gene expression from the donor genome to a greater extent than the host cell.
  • the provided methods comprise modifying the donor genome in a host cell; and modifying the donor genome comprises inducing one or more substitutions, one or more deletions, one or more insertions, one or more rearrangements, one or more recombinations, one or more homologous recombinations, or a combination thereof, into the donor genome.
  • the method comprises modifying the donor genome; and modification of the donor genome effects or improves a property of the donor genome compared to the donor genome prior to modification.
  • the provided methods also include transplantation into a recipient cell where transplantation can be carried out in the presence of polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • sizes of PEG that can be used in the present methods include, but are not limited to, sizes ranging from PEG 4,000 to PEG 20,000. In one embodiment, the size is PEG 8,000.
  • concentrations of PEG can be used in the disclosed methods such as, for example, from about 1% to about 20%. In one embodiment, PEG is used in a concentration of about 5%.
  • a vector for whole genome modification comprising a prokaryotic genome that is at least a minimal genome; a prokaryotic replication origin; a prokaryotic selection marker; a transposase and inverted repeats; one or more nucleic acid sequences capable of supporting segregation and replication in a eukaryotic cell; and a eukaryotic selection marker.
  • the eukaryotic cell is a yeast cell.
  • the prokaryotic genome, prokaryotic replication origin and selection marker can be bacterial.
  • the nucleic acid supporting segregation and replication in a eukaryotic cell comprises one or more of a CEN nucleic acid and an ARS nucleic acid.
  • the prokaryotic genome comprises at least at or about 300 kb in length.
  • the vector is stable in a eukaryotic and in a prokaryotic cell.
  • a combinatorial library containing a plurality of vectors, wherein the prokaryotic genomes can be genome variants.
  • a yeast nucleic acid construct for seamless modification of target region within a target nucleic acid comprising: a first portion of homology, containing homology to a portion of the target nucleic acid that is upstream or downstream of the target region along the length of the target nucleic acid; a nucleic acid encoding an endonuclease under the control of an inducible promotor; a nucleotide sequence recognized by the endonuclease; a yeast selectable marker; a second portion of homology, containing homology to a 5' portion of the target region; and a third portion of homology, containing homology to a 3' portion of the target region.
  • the second and third portions of homology flank the first portion of homology, the nucleic acid encoding the endonuclease, and the yeast selectable marker.
  • the endonuclease recognition site can be adjacent to the second or the third homologous portion and can be on the opposite terminus of the construct relative to the first portion of homology.
  • One or both of the second and third regions of homology comprises one or more substitutions, one or more deletions, one or more insertions, one or more rearrangements, one or more recombinations, one or more homologous recombinations, or one or more combinations thereof, compared to the homologous portion in the target nucleic acid.
  • a method for seamlessly introducing a modification in a target nucleic acid molecule comprising: introducing a mutagenesis construct and a host vector into a host cell whereby the host vector recombines with the mutagenesis construct in the host cell, wherein the mutagenesis construct contains a first portion of homology to a 5' portion of the target nucleic acid molecule upstream of the modification; an endonuclease recognition site, a promoter, a gene encoding the endonuclease, and a selectable marker; a second repeat portion of homology that is homologous to the sequence of the genome upstream of a target locus; and a third portion of homology that is homologous to a 3' portion of the target region downstream of the modification; and incubating the cells under conditions whereby recombination occurs between the first portion of homology and the upstream or downstream portion, thereby seamlessly removing a portion of the construct, that promote one or more double-strand break
  • Treatment to promote double-strand break cleavage can include expression of an endonuclease that cleaves the target nucleic acid molecule containing the construct at a recognition site, producing a double-strand break.
  • the provided methods further comprise performing a selection step, thereby selecting cells in which the yeast selectable marker has been removed from the target nucleic acid.
  • the provided methods comprise transplantation into a recipient cell where transplantation is carried out by isolating the donor genome in the presence of agarose;
  • Incubation with a methyltransferase can be incubation with a crude cell extract.
  • the provided methods can further include incubating the donor genome in the presence of a proteinase after incubation with the methytransferase, thereby removing proteins.
  • the donor genomes and the modified genomes contemplated herein are large nucleic acids.
  • the donor genome is at least at or about or greater than about 100 kb, about 150 kb, about 200 kb, about 250 kb, about 300 kb, about 350 kb, about 400 kb, about 450 kb, about 500 kb, about 550 kb, about 600 kb about 600 kb, about 650 kb, about 700 kb, about 750 kb, about 800 kb, about 850 kb, about 900 kb, about 1 megabase (MB), about 1.1 MB, about 1.2 MB, about 1.3 MB, about 1.4 MB, about 1.5 MB, about 1.6 MB, about 1.7 MB, about 1.8 MB, about 1.9 MB, about 2 MB, about 2.5 MB, about 3 MB, about 3.5 MB, about 4 MB, 4 about.5 MB, or greater in length, or any number therebetween.
  • MB megabase
  • Figure 1 illustrates various embodiments by which a donor genome and host vectors can be introduced (by transformation or cotransformation) into a host cell.
  • One or more genome modifications can be made in the host cell.
  • the modified genome can then be isolated and transplanted into a recipient cell.
  • Figures 2A-2C illustrate three methods for cloning bacterial genomes in yeast.
  • a host vector can be incorporated into a bacterial genome by transformation of the bacterium; the recombined genome and host vector can be isolated and used to transform a yeast host cell.
  • a whole genome and an optionally linearlized host vector can be cotransformed into a yeast host cell, where the yeast host cell combines the vector and the bacterial genome by homologous recombination genome.
  • a bacterial genome can be cloned by assembling multiple overlapping fragments and co-transforming the fragments into yeast host cell with a host vector where the bacterial genome fragments and host vector are combined by homologous recombination in the yeast host cell.
  • Figures 3A-3F illustrate yeast vector insertions in each of the M. genitalium, M. mycoides LC, and M. pneumoniae genomes, using the approach of Figure 2 A.
  • Figures 3A, 3B and 3E illustrate two shuttle vectors used in the experiments.
  • Figures 3C, 3D and 3F illustrate the location of the vector insertion in each genome.
  • Mycoplasma markers are the spiralin promoters, tetM, and lacZ; yeast vector features are CEN, ARS and HIS3; the E.
  • coli plasmid backbone is ampicillin resistance (ampR) and pUC19 origin (ori); BAC sequences are BAC; and transposon elements are IS256 outer inverted repeat (IR), IS256 inner inverted repeat (IR), and transposase (tnp).
  • ampR ampicillin resistance
  • ori pUC19 origin
  • BAC sequences are BAC
  • transposon elements are IS256 outer inverted repeat (IR), IS256 inner inverted repeat (IR), and transposase (tnp).
  • Figures 4A-4B show the analysis of whole Mycoplasma genome clones containing yeast vector sequences.
  • Figure 4A provides a map of the M. genitalium cll6-2 genome. The location of the yeast vector insertion is marked. Bars indicate position and numbers indicate size of PCR amplicons. Restriction fragments are numbered and their sizes provided in the legend; Eagl digest correspond to restriction fragment 1 and BssHlI digests correspond to restriction fragments 2-6.
  • Figure 4B provides a map of the M. pneumoniae genome. The location of the yeast vector insertion is marked. Bars indicate position and numbers indicate size of PCR amplicons. Restriction fragments are numbered and their sizes provided in the legend; Notl digests correspond to restriction fragments 1-4 and Sbf! correspond to restriction fragments 5 and 6.
  • Figure 5 provides a map of the M. mycoides LC ell .1 genome. Arrowheads represent IS 1296 elements. Bars indicate position and numbers indicate size of PCR amplicons.
  • FIGs. 6A-D illustrate targeted insertion of a yeast vector using homologous recombination as four alternate methods.
  • a yeast vector insertion was attempted with and without a double-stranded break at the insertion point.
  • Figure 6A Intact genome and linear vector.
  • Figure 6B Overlapping genome fragments and vector with homology internal to one of the fragments.
  • Figure 6C Genome cleaved at the integration target and linear vector.
  • Figure 6D Overlapping genome and vector fragments with homology to two of the fragments.
  • Figures 7A-7C demonstrate that crude extracts protected donor plasmid DNA from host restriction-modification system and increased efficiency of transformation, but inhibited genome transplantation.
  • Figure 7 A illustrates treatment of agarose plugs with a methylation step or the results of no treatment.
  • Figure 7B shows native genomic DNA treated in the absence of crude extracts (shown to allow transplant into recipient cells) displayed a punctate pattern (right panel), while endogenous genomic DNA treated in the presence of crude extracts (shown to inhibit transplant) formed large aggregates (left two panels).
  • Figure 7C shows that removal of the crude extracts by proteinase K treatment after incubation with the genomic DNA in agarose plugs restored the punctate pattern originally observed with the untreated genomic DNA.
  • Figure 8 shows three alternative whole genome transplantation methods that can be used.
  • the first approach (1) includes digestion of agarose plugs containing the genomic DNA ⁇ e.g., with ⁇ -agarase (melting step)), followed by transplantation directly into recipient cells.
  • the second approach (2) is identical to the first method, except that recipient cells had been modified to mutate the restriction enzyme genes (ARE).
  • ARE restriction enzyme genes
  • genomic DNA samples were methylated in vitro and subjected to a deproteinisation step (treatment with proteinase K), prior to the melting step ( ⁇ -agarase digestion) and
  • Figures 9A-9C illustrate problems with conventional modification methods with respect to unspecific deletions or rearrangements.
  • CEN6 round circles contained within constructs.
  • Figure 9A illustrates introduction of the wild-type fragment into yeast carrying the M. genitalium with URA3 insertion followed by selection on SD-HIS plates containing FOA, resulting in selection of two different types of recombination events (PI
  • Figures 10A-10D illustrate alternate seamless modification methods.
  • Figure 10A schematically illustrates generation of a diletto perfetto mutagenesis cassette: the cassette was introduced into the yeast strain containing the M. genitalium genome, using lithium acetate integrative transformation. Individual Ura + transformants were selected and analyzed by PCR, using diagnostic primers Seq-F and Seq-R (shown as small, single-head arrows flanking the insertion site. A fusion product was generated that contained the URA 3 marker and a 358 bp fragment ("repeat" fragment) homologous to a portion just upstream of the target locus (large arrow labeled as "repeat”.
  • the fusion product was PCR-reamplified: the resulting cassette contained, in the following order, 50 bp of homology to a 5' portion of the target region (upstream of the single-base deletion), the URA3 marker, the repeat cassette, and 50 bp of homology to a 3' portion of the target region.
  • the cassette was designed in this orientation so that upon transformation into the yeast host cells, replacement of a 450 base pair target region within the CDS 139 locus of the M. genitalium genome with this cassette (by HR) would result in a region in the genome containing two tandem repeat sequences (large arrows in labeled as "repeat") flanking the URA3 selection marker.
  • FIG. 1 a TREC (Tandem Repeat with Endonuclease Cleavage) mutagenesis construct was generated by fusing the (GALl/l-Scel)- URA3 fusion product with the 358 bp "repeat" fragment located upstream of the target locus.
  • the resulting TREC cassette contained, in the following order, 50 bp of homology to a 5' portion of the target region (upstream of the single-base deletion), a CORE cassette
  • the "repeat” (consisting of the 18 bp l-Scel recognition site, the GALl promoter, a gene encoding l-Scel endonuclease and the URA3 marker), the "repeat” (358 bp portion homologous to sequence of the genome just upstream of the target wt gene locus), and 50 bp of homology to a 3' portion of the target region (downstream of the single-base deletion being corrected).
  • the resulting LoxP-RE-GALl-Cxe-URA3-loxP- E mutagenesis cassette contained, in the following order, 50 bp of homology to a 5' portion of the target region (upstream of the single-base deletion), a first loxP site (loxP-RE), the GALl promoter, a Cre recombinase gene ORF, the URA3 marker, a second loxP site loxP-LE), and 50 bp of homology to a 3' portion of the target region (downstream of the single-base deletion).
  • Figure 11 illustrates generation of a Type III restriction enzyme deletion.
  • a linear DNA fragment, Knock-Out Cassette (KOC) was constructed by fusing two PCR products, CORE and tandem repeat sequence (TRS). This cassette was then transformed into yeast W303a strain harboring M. mycoides LC genome- YCp to replace the TypeR III ORF via the 50-bp homologous sequences to the target sites (Atypelllres: :URA3).
  • Galactose induction results in the expression of l-Scel endonuclease which cleaves the 18-bp l-Scel site (asterisk) to create a double strand break that promotes the homologous recombination between two tandem repeat sequences (red arrow). Recombination between the TRSs creates a seamless deletion of the typelllres gene (AtypefflR).
  • Figures 12 A - 12B illustrate engineering a point mutation at the MG259 locus of a synthetic M. genitalium genome.
  • Figure 12 A illustrates the scheme of mutation correction by two sequential homologous recombinations. Two primers (arrows), Seq-F and Seq-R, are separated by 0.4 kb in MG259 locus, and the insertion of a 1.1 kb URA3 marker results in the production of a 1.3 kb PCR DNA fragment.
  • Figure 12B illustrates possibilities of the URA3 marker loss from an M. genitalium YAC. 5-FOA resistant clones could be derived either from the replacement of the URA3 marker with the wild type DNA fragment (Rl) or from recombination between two repetitive sequences (R2). Size and locations of repeat sequences are schematic.
  • Figure 13 illustrates an outline of an exemplary TREC method.
  • the target region is replaced with a mutagenesis cassette that consists of a knock-out CORE (anl8-bp of l-Scel recognition site, I-Scelgene under the control of GAL1 promoter, and the URA3 gene) and a DNA fragment (shown in arrow) identical to the upstream of the target site.
  • the replacement generates tandem repeat sequences encompassing the CORE.
  • Galactose induces the expression of l-Scel that generates a double-strand break (DSB) at l-Scel site.
  • DSB promotes an intramolecular homologous recombination between the repeat sequences leading to an excision of the CORE.
  • Figures 14A and 14B illustrate two other methods to engineer the same locus and produce a point mutation or 450 bp deletion by the delitto perfetto method ( Figure 14A) or the tandem repeat pop-out method ( Figure 14B), respectively.
  • Figure 15 illustrates exemplary generation of final mutagenesis cassette constructs.
  • Figure 16 illustrates moving a donor genome into host cell, engineering it, and installing it back into a recipient by genome transplantation.
  • the repertoire of yeast genetic methods is used to create insertions, deletions, rearrangements, or any combination of modifications in the bacterial genome.
  • This engineered genome is then isolated and transplanted into a recipient cell to generate an engineered bacterium. Before transplantation back into the recipient cell it may be necessary to methylate the donor DNA in order to protect it from the recipient cell's restriction system(s). This cycle can be repeated starting from the newly engineered genome (dashed arrow).
  • Figure 17 provides the nucleic acid sequence of the seamless deletion region of the YCpMmycl . l-Atypelllres M. mycoides genome transplant verifying that the Type III restriction gene was removed as designed. Italicized sequence text corresponds to the "typelllmod" region of the genetic maps Figure 19; underlined sequence text corresponds to the "typelllres” region; and bold text corresponds to the "IGR" region. A small portion of the typelllres gene remained after the deletion because of the overlap between the typellrniod and typelllres genes.
  • Figures 18A-18C show the stability of the M. mycoides YCpMmycl .1 genome during propagation in yeast.
  • Figure 18A provides a schematic of the YCpMmycl.1 genome. The position of the integrated YCp is shown. The nine individual primer pairs used in the PCR amplifications are shown at their approximate locations in the genome and are numbered corresponding to the amplicons in Figure 18B.
  • Figure 18B Stability of the M. mycoides genome during propagation in yeast was tested by two methods. In the first, a yeast culture of a clone containing the genome was plated on solid synthetic media lacking histidine for two days and then individual colonies were patched onto a new plate.
  • a yeast culture of a clone containing the genome was grown to saturation, diluted to a 1/100 fraction and again grown to saturation. The culture was then plated on solid synthetic media lacking histidine for two days and then individual colonies were patched onto a new plate.
  • genomic DNA was isolated and used as template in multiplex PCR amplification using the nine individual primer pairs shown in A. The resulting amplicons were analyzed by gel electrophoresis. The numbers on the right side of the gel correspond to the individual primer pair amplicons as shown in A. Lane G is a positive control and lane N is the no-genome negative control. Molecular weight markers are in lane M. The results shown are representative of the 40 samples analyzed.
  • Figuer 18C provides a schematic of the M. mycoides YCpMmycl .1 genome; the position of the integrated YCp is shown.
  • the nine individual primer pairs used in the PCR amplifications are shown at their approximate locations in the genome and are numbered corresponding to the amplicons identified by Fig 18B. .
  • the diagonal line represents the missing amplicons in clone 3.
  • FIG. 19 illustrates the generation of Type III restriction enzyme deletions.
  • Type III restriction enzyme gene typefflres
  • iii a linear DNA fragment, knockout cassette, by fusing two PCR products, CORE and tandem repeat sequence (TR) was constructed (i).
  • This cassette was then transformed into a yeast W303a strain harboring the YCpMmycl.l M. mycoides genome (ii). Growth on (-)His (- )Ura medium selected for replacement of the Type III restriction enzyme open reading frame (ORF) by the cassette via the 50-base pair (bp) sequences homologous to the target sites (AtypeIIIres::URA3).
  • Galactose induction results in the expression of 1-SceI endonuclease, which cleaves the 18-bp l-Scel site (asterisk) to create a double-strand break that promotes homologous recombination between two tandem repeat sequences (TR) (unmarked line above constructs).
  • Figure 20 provides a schematic demonstrating the assembly of a synthetic M. mycoides genome in yeast.
  • a 1,077,947 bp synthetic M. mycoides genome was assembled from 1,078 overlapping DNA cassettes in three steps.
  • 1,080 bp cassettes (orange arrows), produced from overlapping synthetic oligonucleotides, were recombined in sets of 10 to produce one hundred nine ⁇ 10 kb assemblies (blue arrows). These were then recombined in sets of 10 to produce eleven ⁇ 100 kb assemblies (green arrows).
  • these eleven fragments were recombined into the complete genome (red circle).
  • assemblies were carried out by in vivo homologous recombination in yeast.
  • Major variations from the natural genome are shown as yellow circles. These include 4 watermarked regions (WM1-WM4), a 4 kb region that was intentionally deleted (94D), and elements for propagation in yeast and genome transplantation.
  • WM1-WM4 watermarked regions
  • 94D 4 kb region that was intentionally deleted
  • elements for propagation in yeast and genome transplantation there are 20 locations with nucleotide polymorphisms (asterisks). Coordinates of the genome are relative to the first nucleotide of the natural M. mycoides sequence. The locations of the Ascl and BssHII restriction sites are shown. Cassettes 1 and 800-810 were unnecessary and removed from the assembly strategy.
  • FIG. 21 provides a schematic illustrating error correction using PCR and in vitro recombination.
  • Primers BH pUC bckbn Fori and Revl
  • BH insert Fori and Revl Complements of backbone primers
  • Erlements of backbone primers (BH insert Fori and Revl) were used in conjunction with error correcting primers (Cassette Fix Fori and Revl) to produce amplicons with regions of homology to each other and the BH backbone amplicon.
  • the three PCR products were used in in vitro recombination to generate the corrected cassette.
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid means “one”, “at least one” or “one or more.”
  • nucleic acid RNA
  • nucleic acid molecule nucleic acid sequence
  • oligonucleotides oligonucleotides
  • polynucleotide are used interchangeably and include both ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) and modified nucleic acid molecules, such as peptide nucleic acids (PNA), locked nucleic acids (LNA), and other modified nucleic acid molecules, including, without limitation, cDNA, genomic DNA and m
  • a "restriction endonuclease site” refers to a target nucleic acid sequence that is recognized and cleaved by a restriction enzyme. Restriction enzymes are well known in the art.
  • genome includes whole (complete) genomes (e.g., whole cellular, viral, and organelle genomes), and also includes portions of whole genomes having nucleic acid sequences sufficient to effect and/or sustain viability of a cell (minimal cellular genome), viability, within a host cell, of an organism that depends on a host cell for viability (e.g., minimal viral genome), or organelle function within a host cell (minimal organelle genome), under at least one set of environmental conditions.
  • genome refers to whole genomes and portions thereof that are at least minimal genomes. The particular environmental conditions and property that is caused or sustained by the genome can be specified.
  • the environmental conditions can include the environment of a suitable and functional host cell.
  • genome encompasses minimal genomes and minimal replicative genomes, and genomes containing additional nucleic acid sequences beyond those found in such minimal genomes but not containing all the nucleic acid sequences present in a whole genome.
  • gene encompasses naturally-occurring genomes and synthetic genomes, and includes genetically engineered genomes, such as genomes not previously existing in nature or in a laboratory, including modified genomes and hybrid genomes that contain nucleic acids and/or portions of genomes from more than one species.
  • genome encompasses organelle genomes (e.g., mitochondrial and chloroplast genomes), genomes of self-replicating organisms (cellular genomes), including prokaryotic and eukaryotic organisms, fungi, yeast, bacteria (e.g., Mycoplasma), archeabacteria, vertebrates, mammals, and other organisms, and viral genomes and other genomes that depend on a host for propagation. Genomes further include those of organisms not falling into any known Linnean catergory and synthetic organisms. Exemplary genomes can be microorganism genomes, such as genomes of unicellular organisms including bacteria and yeast.
  • cellular genome refers to genomes containing nucleic acid sequences sufficient to cause and/or sustain viability of a cell.
  • nucleic acid sequences include those encoding molecules required for replication, transcription, translation, energy production, transport, production of membranes and cytoplasmic components, and cell division.
  • Cellular genomes include minimal cellular genomes, whole cellular genomes, and genomes having nucleic acids additional to minimal cellular genomes but not all the nucleic acids of whole cellular genomes. Cellular genomes differ from "viral genomes" and
  • organelle genomes at least in that a cellular genome contains the nucleic acids sufficient for replication and/or viability of a cell whereas viral and organelle genomes contain the nucleic acids necessary to sustain or replicate the virus or organelle, e.g., within a host cell, but not to sustain the viability or replication of the host cell.
  • minimal genome refers to a genome consisting of or consisting essentially of a minimal set of nucleic acids sufficient to effect and/or sustain viability of a cell (minimal cellular genome), viability, within a host cell, of an organism that depends on a host cell for viability (e.g., minimal viral genome), or organelle function within a host cell (minimal organelle genome), under at least one set of environmental conditions. It is understood that even whole organelle genomes do not necessarily encode all the proteins needed to perpetuate the organelle, and that some of the proteins are encoded by genes within the nucleus of the cell containing the organelle. Thus, minimal organelle genomes need only contain those genes necessary for organelle function within the environment of the cell.
  • minimal viral genomes are minimal genomes that, in addition to the minimal nucleic acid sequences sufficient for survival, further contain nucleic acid sequences sufficient for self replication of a cell or organism.
  • synthetic nucleic acid sequences including synthetic genomes, all or part of which have been constructed from genetic components that have been chemically synthesized in vitro or copies of such components. The copies may have been produced by any of a number of methods as are known in the art, including cloning and amplification by in vivo or in vitro methods.
  • a completely synthetic nucleic acid sequence or genome is one in which the entire nucleic acid or genome has been chemically synthesized in vitro or has been produced or assembled from copies of such in vitro chemically synthesized nucleic acids.
  • a "semi-synthetic" genome refers to a partially synthetic nucleic acid sequence or genome is a synthetic genome in which some of the genetic components are naturally- occurring, including nucleic acids cloned from naturally-occurring nucleic acids.
  • a foreign or heterologous genome or nucleic acid sequence is a genome or nucleic acid sequence that is present in a host cell but is derived from a donor organism that is of a different species than the host cell.
  • the donor organism can be of a different genus, order, kingdom, or other genetic classification, or can simply be of a different species in the same genus.
  • a "target nucleic acid sequence” refers to a nucleic acid sequence that is targeted for modification, for example, by the modification methods described herein and known in the art.
  • One or more modifications of a target nucleic acid sequence includes introduction of one or more mutations, one or more deletions, one or more substitutions and/or one or more insertions into the target nucleic acid sequence.
  • Target regions are particular regions of the target nucleic acid sequences, such as a single gene locus, multiple gene loci, or portions thereof that are the subject of modification.
  • the target region includes the region of the target nucleic acid sequence that is replaced with another nucleic acid sequence such as, for example, by homologous recombination.
  • modification of the target nucleic acid sequence it is not necessary that the entire target region in the modified nucleic acid sequence be modified compared to the original target region.
  • modification of the target region can encompass a single insertion, deletion or substitution at a target position/residue within the target region, or can encompass modification of a number of positions/residues within one or more target portions of the target region.
  • nucleic acids, methods and systems for introducing donor genomes and other donor nucleic acid sequences into heterologous host cells for modifying the donor genomes and nucleic acid sequences within host cells, recovering donor genomes and nucleic acid sequences from the host cells, and introducing the recovered donor genomes and nucleic acid sequences into recipient cells.
  • FIG. 1 An exemplary embodiment of the provided methods is illustrated in Figure 1, in which the methods are used to transform a bacterial donor genome into a yeast host cell, by joining it with a yeast host vector, modifying the donor genome within the yeast host cell, and transplanting the modified donor genome into a bacterial recipient cell, thereby generating an engineered bacterium.
  • the modified genome present in that engineered bacterium can be isolated and serve as a donor genome in a subsequent round of the methods in an iterative fashion.
  • a number of variations of this embodiment are contemplated and within the scope of this application, as described herein.
  • FIG. 16 Another exemplary method of the provided methods is illustrated in Figure 16 in which the methods are used to insert a host yeast vector into a bacterial genome, isolate the genome with the integrated yeast vector, transform a yeast host cell with the bacterial genome/yeast vector, modify the bacterial genome, isolate the modified genome, optionally methylate the genome, and transplant the donor genome into a recipient cell.
  • Example 5 describes the successful combination of the provided methods to generate a new bacterial organism by transforming a donor bacterial genome into yeast host cells, modifying the donor genome within the yeast cells, and then transplanting the resulting modified donor genomes into recipient cells, whereupon gene expression from the modified donor genomes was induced. The results were verified by negative controls and sequencing of genomes in the recipient cells.
  • This study demonstrated successful generation of a M mycoides LC genome, containing a seamless deletion of the Type III restriction enzyme gene, which could not have been generated within the Mycoplasma using current state of the art method using its available native genetic systems. Transplantation produced a M. mycoides LC strain that had not previously existed in the laboratory or in nature.
  • the provided methods can be used for successful genetic engineering outside of the cell encoding the genome being engineered.
  • Methods and tools, and resulting cells, for manipulating genomes and large nucleic acids are provided, for example, to engineer and alter organisms that can produce useful compounds, such as vaccines, drugs, biologically produced proteins or chemicals, and biofuels.
  • useful compounds such as vaccines, drugs, biologically produced proteins or chemicals, and biofuels.
  • methods are needed for manipulation of genomes and chromosomes of organisms having poor genetic systems, such as intractable organisms, to generate modified genes, genomes, and organisms that produce new gene products such as those useful in energy production and medicine.
  • the present invention also provides methods to transplant them from the host cells into an environment in which gene products can be expressed, such as an appropriate recipient cell.
  • expression may be sufficient in host cells, for example, it may be desirable to transplant the donor genomes from the host cells into recipient cells that have cellular environments that more closely replicate that of the original donor cell or organism from which the natural or synthetic donor genome was derived.
  • improved transfer, cloning, modification, and transplantation methods, nucleic acids, and systems that can be used to manipulate and engineer genomes and other nucleic acids, particularly large nucleic acids, are provided herein.
  • nucleic acids cloned by conventional methods generally contain no more than a few genes.
  • donor nucleic acids can be toxic to host cells ⁇ e.g., if toxic proteins are expressed from the donor nucleic acid) and can becomes unstable during events such as host cell replication and/or modification using the host's genetic system. Such events may limit the ability to manipulate the nucleic acids within the host cells. Further, the fact that unwanted modifications to a donor genome, which may occur during the modification process, often have no negative impact on host cell viability can make donor genomes and other nucleic acids unstable in host cells.
  • Incompatibility issues also can impair efficient transplant of the donor genomes from the host cells back into a more natural environment for expression of gene products, such as into recipient cells of the same or closely related species as the donor.
  • modification methods that overcome such issues for successful propagation and modification of donor nucleic acids, including genomes, in genetically distinct host cells, such as yeast host cells for transplantations into recipient cells more distinct from the species of the original donor genome.
  • host cells such as yeast
  • do not contain restriction-modification systems they can express DNA methyltransferases that can modify the donor nucleic acids being propagated in the host cell, thereby inhibiting activation of the donor nucleic acid (e.g., genome) upon transplantation into a recipient cell.
  • the structure and confirmation of donor genomes isolated after propagation and modification in host cells can also differ from the confirmation and structure of the same genome propagated in a cell more closely related to the donor organism. Such differences can negatively impact transplantation.
  • donor genomes recovered from host cells are introduced into genetically distinct recipient cells (such as from eukaryotic hosts to prokaryotic recipients).
  • Donor nucleic acid sequences are selected and synthesized, assembled, and/or isolated (e.g., from donor cells) and cloned in host cells.
  • the methods comprise obtaining a donor genome from a donor cell or synthesizing a donor genome as one or more fragments and introducing the donor genome and a host vector into a heterologous host cell, wherein the donor genome and the host vector are optionally joined prior to introduction into the host cell, thereby generating a host cell comprising the donor genome comprising the host vector, and further whereinthe donor genome is an essentially intact cellular, viral, or organelle genome that is at least a minimal genome, and is greater than about 300 kb in length.
  • a first embodiment typically involves introduction of a host vector into a cell containing a donor genome, joining of the donor genome to the host vector, recovering the donor genome comprising the host vector and transforming a host cell such that the donor genome comprising the host vector is maintained v thin the host cell during host cell replication.
  • a host vector and a linearized donor genome are cotranformed in a host cell where the host vector and donor genome are joined by homologous recombination in the host cell.
  • overlapping DNA fragments (natural or synthetic) and a host vector are cotranformed in a host cell where the host vector and DNA fragments are joined by homologous recombination in the host cell.
  • a donor genome to be used in the present methods can be an essentially whole cellular, viral or organelle genome.
  • the donor genome and the host vector can be introduced into the host cell simultaneously or sequentially in either order.
  • the host vector is joined with the donor genome prior to introduction into the host cell by transforming the host vector into a donor cell containing the donor genome.
  • a host cell for use in the present embodiments can be a eukaryotic cell or a prokaryotic cell.
  • Host cells include, but are not limited to, a bacterial cell, a fungal cell, an insect cell, a plant cell, or an algal cell.
  • Host cells also include yeast cells.
  • a host vector is a vector useful for homologous recombination.
  • a host vector for use in the present embodiments can be a centromeric plasmid.
  • the host vector is a yeast centromeric plasmid and the host cell is a yeast cell.
  • a donor genome contemplated for use in the present embodiments can be, for example, a bacterial genome, a fungal genome, a yeast genome, an archeal genome, a cyanobacterial genome, an algal genome, a bacteriophage genome, a mitochondrial genome, a chloroplast genome, viral genome, an organelle genome, or a synthetic genome.
  • the methods further comprise modifying the donor genome within the host cell.
  • the methods further comprise recovering the donor genome comprising the host vector from the host cell.
  • the methods further comprise introducing the recovered donor genome into a recipient cell.
  • the methods further comprise degrading or removing the endogenous genome of the recipient cell.
  • a recovered donor genome can be methylated prior to introduction into the recipient cell.
  • a recipient cell's restriction endonulease function is absent, removed or inactivated.
  • a recipient cell contemplated for use in the present embodiments can he, for example, a bacterial cell, a yeast cell, a fungal cell, an insect cell, a plant cell, or an algal cell.
  • the methods further comprise introducing a second donor genome into the host cell, wherein the second donor genome is different from the first donor genome, thereby producing a host cell containing two different donor genomes.
  • Introducing the second donor genome can comprise mating the host cell containing the first donor genome with a second host cell containing the second donor genome.
  • Introducing the recovered donor genome into the recipient cell can phenotypically transform the recipient cell to a phenotype corresponding to the donor genome incorporating any modifications thereto.
  • a process for making a cell which exhibits a phenotype encoded by a donor genome comprising: (a) introducing into a host cell, the donor genome and a host vector suitable for cloning the donor genome in the host cell such that a product comprising the donor genome comprising the host vector is obtained; (b) recovering the product comprising the donor genome comprising the host vector obtained in step (a) from the host cell; (c) introducing the product of (b) into a recipient cell under conditions such that the recipient cell exhibits a phenotype encoded by the product; and (d) recovering the cell resulting from step (c); wherein the donor genome is an essentially intact cellular, viral, or organelle genome that is at least a minimal genome, and is greater than about 300 kb in length and comprises the minimal components of nucleic acid material necessary for the recipient cell to exhibit the phenotype encoded by the donor genome.
  • the method further comprises modifying the donor genome of (a) within the host cell. In another aspect, the method further comprises degrading or removing an endogenous genome of the recipient cell. In yet another aspect, the method further comprises modifying the donor genome of (a) within the host cell and degrading or removing an endogenous genome of the recipient cell. [0105] Provided herein is a cell which exhibits a desired phenotype encoded by a donor genome and not otherwise exhibited by the cell, wherein the cell is produced by the processes described herein.
  • Also provided herein is a cell which comprises a donor genome and exhibits a desired phenotype encoded by the donor genome and not otherwise exhibited by the cell, wherein the donor genome comprises greater than 300 kb of foreign genomic nucleic acid material and the minimal components of a genome necessary for the cell to exhibit the desired phenotype.
  • the modification methods and tools include aspects that minimize the risk of instability of the donor nucleic acid sequence within the host cell during modification.
  • donor nucleic acid sequences are transplanted from host cells into recipient cells, which can be of a different species and/or different branches of life than the donor cells and the host cells, or of the same species as the donor cells.
  • the transplant methods include aspects that minimize the risk of incompatibility and toxicity among the donor genome, host cells, and the recipient cells.
  • the transfer, modification, and transplantation methods can be performed separately, and can also be performed sequentially, in combination.
  • the transfer, modification, and transplantation methods can be performed separately, and can also be performed sequentially, in combination.
  • the three steps can be combined in a method by which donor genomes are transferred into a host cell, modified within the host cell, and transplanted into a recipient cell to generate a new cell, thereby generating a genome or cell not previously existing in the laboratory or in nature.
  • the recipient cells can be further grown into, or transferred into, a non-human organism not previously existing.
  • the provided methods and compositions are particularly useful for manipulating and engineering genomes from organisms that are genetically intractable.
  • the methods, nucleic acid sequences, and systems are used to clone whole bacterial genomes from Mycoplasma genitalium, Mycoplasma pneumoniae, and Mycoplasma mycoides LC as circular centromeric plasmids in yeast, to modify the donor genomes within the yeast using the yeast genetic systems with modifications to minimize incompatibility and, further, to transplant the modified bacterial genomes into recipient cells of a different species, thereby generating genomes and organisms not previously existing in the lab or in nature.
  • the provided methods, nucleic acid sequences, systems, and organisms can be used to engineer organisms that synthesize biofuels.
  • bacteria such as Escherichia coli can be genetically modified
  • many prokaryotes having the potential to produce industrially useful compounds or to function in extreme environments have very poor or non-existent genetic systems.
  • Prochlorococcus marinus is among the most abundant photosynthetic organisms on earth. While it is desirable to manipulate and engineer this and other such organisms to produce biofuels, the ability to manipulate and engineer such organisms is limited by the lack of available methods to genetically alter them.
  • the provided methods can be used to carry out such manipulations.
  • nucleic acid sequences encoding components of new metabolic pathways can be introduced into the genomes of such organisms by transfer and modification within host cells.
  • Such re-engineered genomes can be transplanted into suitable recipient cells to produce new cells, e.g., new cells that can convert sunlight and carbon dioxide into a biofuel.
  • Such engineered cells and organisms also are provided herein.
  • the provided method also can be useful for engineering archea, cloning new organelles into eukaryotes and adding chromosomes to cells and organisms.
  • eukaryotic mitochondria and chloroplasts are remnants of endosymbiotic bacteria that have become trapped in their hosts.
  • the provided methods can be used to engineer such organelle genomes in hosts, e.g., in yeast using plasmids, using homologous recombination, thereby creating new mitochondria and chloroplast genomes having improved energy production efficiency and/or metabolism, such as in yeast or algae.
  • the provided methods can be used to manipulate viruses, such as those with large genomes that are too large for manipulation in simple plasmids, to produce viruses and bacteriophages having therapeutic uses.
  • viral genomes are cloned and manipulated to improve their immunogenicity and other therapeutic advantages.
  • the provided methods can be used to manipulate fungi to produce fungii useful in the production of, for example, wine, bread, beer and medicine.
  • fungal genomes are cloned and manipulated to improve their resistance to temperature, disease-causing organisms, and other advantages.
  • the provided methods can be used to manipulate yeast to produce yeast useful in ethanol fuel, nutritional suppliements, probiotics, fermentation for production of beverages (alcoholic and non-alcoholic) or for use in baking.
  • Methods and processes of the invention are amenable to automation and to adaptation to high throughput methods, for example, allowing for the joining of multiple nucleic acid molecules and transformation into host or recipient cells simultaneously by computer-mediated and/or robotic methods that do not require human intervention.
  • the present invention is directed to systematic methods and the products thereof that permit efficient and extensive assembly, cloning, modification, and
  • nucleic acid assembly reactions can be performed on a solid surface as opposed to in a reaction tube, for example, on a chip using microfluidics.
  • a donor genome or other nucleic acid sequence is selected for transfer, modification, and/or transplantation.
  • the nucleic acid sequences that are transferred, modified, transplanted, and generated by the methods described herein can be of any natural or synthetic organism.
  • the donor genome is derived from any desired cell or any nucleic acid-containing subunit thereof, either by isolation or chemical synthesis.
  • the nucleic acid sequences include genomes (such as whole genomes, portions of whole genomes that are at least minimal genomes and/or at least minimal replicating genomes, cellular genomes, organelle genomes, and viral genomes), chromosomes, and other large nucleic acid sequences from known organisms and new organisms.
  • the nucleic acid sequences can be of any source within the organisms, including organelle genomes, such as mitochondrial and chloroplast genomes, chromosomes, portions of genomes or chromosomes of plants and animals, algal sources, and any genomic material that supports cell viability, including the whole and minimal cellular genome of bacteria and other prokaryotes, and eukaryotic organisms.
  • organelle genomes such as mitochondrial and chloroplast genomes, chromosomes, portions of genomes or chromosomes of plants and animals, algal sources, and any genomic material that supports cell viability, including the whole and minimal cellular genome of bacteria and other prokaryotes, and eukaryotic organisms.
  • the methods can be used, for example, to join portions of genomes of various organisms in the same DNA molecule to generate new genomes and organisms not present in nature or in the laboratory.
  • the donor genomes and other nucleic acids can be cloned, propagated and/or isolated from cells, such as cells or tissues (including genetically engineered organisms), or can be chemically synthesized in vitro. Methods for isolating and preparing the nucleic acids and genomes are described below. i. Donor organisms, genomes and other nucleic acids
  • Genomes and other nucleic acid sequences used and generated in the provided methods include those derived from fungi, yeast, bacteria, other prokaryotes, and algae but are not limited to such organisms. They can be of any organism, natural or synthetic, e.g., organisms of the kingdoms Protista, Archaebacteria, Eubacteria, Fungi, Plantae, andAnimalia, and viruses, including bacteriophages.
  • nucleic acid sequences are those derived from bacteria, archea, cyanobacteria (e.g., Prochlorococcus marinus, Synechocystis PCC6803, etc.), algae, viruses (e.g., Haemophilus influenzae genomes), fungi (e.g., Saccharomyces cerevisiae,
  • Exemplary Mycoplasma strains include Mycoplasma genitalium (e.g., M. genitalium strain MS5, described in Example 1, M. genitalium G37 (GenBank No. L43967)), Mycoplasma mycoides (e.g., M. mycoides subspecies mycoides Large Colony (LC) strain GM12 (Example 1), Mycoplasma capricolum subsp. capricolum (strain California KidTM) (ATCC 27343), Mycoplasma mycoides subsp.
  • Mycoplasma genitalium e.g., M. genitalium strain MS5, described in Example 1, M. genitalium G37 (GenBank No. L43967)
  • Mycoplasma mycoides e.g., M. mycoides subspecies mycoides Large Colony (LC) strain GM12 (Example 1), Mycoplasma capricolum subsp. capricolum (strain California KidTM) (ATCC 27343)
  • mycoides strain GM12 (Damassa et al, 1983), Mycoplasma capricolum subsp. capricolum (M.capricolum), such as wtMcapricolum and a Mcapricolum mutant (M. capricolum- ARE) and Mycoplasma pneumonia (e.g., M. pneumoniae strain M129-B170 (ATCC 29343); M. pneumoniae M129, GenBank Accession Number U00089.2 (GI: 26117688)), Mycoplasma gallisepticum (ATCC 15302), Mycoplasma pneumoniae Eaton (ATCC15531), and derivatives thereof.
  • strain GM12 strain GM12
  • M.capricolum Mycoplasma capricolum subsp. capricolum
  • Mcapricolum mutant M. capricolum- ARE
  • Mycoplasma pneumonia e.g., M. pneumoniae strain M129-B170 (ATCC 29343); M. pneumoniae M129, GenBank
  • Exemplary genomes and nucleic acids include full and partial genomes of a number of organisms for which genome sequences are publicly available and can be used with the disclosed methods, such as, but not limited to, Aeropyrum pernix; Agrobacterium tumefaciens; Anabaena; Anopheles gambiae; Apis mellifera; Aquifex aeolicus; Arabidopsis thaliana; Archaeoglobus fulgidus; Ashbya gossypii; Bacillus anthracis; Bacillus cereus; Bacillus halodurans; Bacillus licheniformis; Bacillus subtilis; Bacteroides fragilis;
  • Chlamydia muridarum Chlamydia trachomatis; Chlamydophila caviae; Chlamydophila pneumoniae; Chlorobium tepidum; Chromobacterium violaceum; Ciona intestinalis;
  • Clostridium acetobutylicum Clostridium perfringens; Clostridium tetani; Corynebacterium diphtheriae; Corynebacterium efficiens; Coxiella burnetii; Cryptosporidium hominis;
  • Helicobacter hepaticus Helicobacter pylori; Homo sapiens; Kluyveromyces waltii;
  • Lactobacillus johnsonii Lactobacillus plantarum; Legionella pneumophila; Leifsonia xyli; Lactococcus lactis; Leptospira interrogans; Listeria innocua; Listeria monocytogenes;
  • Mycoplasma mycoides; Mycoplasma penetrans; Mycoplasma pneumoniae; Mycoplasma pulmonis; Mycoplasma mobile; Nanoarchaeum equitans; Neisseria meningitidis;
  • Neurospora crassa Nitrosomonas europaea; Nocardia farcinica; Oceanobacillus iheyensis; Onions yellows phytoplasma; Oryza sativa; Pan troglodytes; Pasteur ella multocida;
  • Plasmodium falciparum Plasmodium yoelii yoelii; Populus trichocarpa; Porphyromonas gingivalis Prochlorococcus marinus; Propionibacterium acnes; Protochlamydia
  • Xanthomonas axonopodis Xanthomonas campestris; Xylella fastidiosa; Yarrowia lipolytica; Yersinia pseudotuberculosis; and Yersinia pestis nucleic acids.
  • algae includes cyanobacteria (Cyanophyceae), green algae
  • algae Chlorophyceae
  • yellow-green algae Xanthophyceae
  • golden algae Chrysophyceae
  • brown algae Phaeophyceae
  • red algae Rhodophyceae
  • diatoms Bacillariophyceae
  • pico-plankton Prasinophyceae and Eustigmatophyceae
  • members of the taxonomic classes Dinophyceae, Cryptophyceae, Euglenophyceae, Glaucophyceae, and Prymnesiophyceae.
  • Microalgae are unicellular or colonial algae that can be seen as single organisms only with the aid of a microscope.
  • Microalgae include both eukaryotic and prokaryotic algae (e.g., cyanobacteria).
  • Photosynthetic bacteria include cyanobacteria, green sulfur bacteria, purple sulfur bacteria, purple nonsulfur bacteria, and green nonsulfur bacteria.
  • Exemplary genomes and nucleic acids include full and partial genomes of a number of algal organisms for which genome sequences are publicly available and can be used with the disclosed methods, such as, but not limited, Achnanthes, Amphiprora, Amphora, Ankistrodesmus, Aster omonas, Boekelovia, Borodinetta, Botryococcus,
  • photosynthetic bacteria including for example, green sulfur bacteria, purple sulfur bacteria, green nonsulfur bacteria, purple nonsulfur bacteria, or cyanobacteria may be used.
  • Cyanobacterial species that can be used include, without limitation, Agmenellum, Anabaena, Anabaenopsis, Anacystis, Aphanizomenon, Arthrospira, Asterocapsa, Borzia, Calothrix, Chamaesiphon, Chlorogloeopsis, Chroococcidiopsis, Chroococcus, Crinalium, Cyanobacterium, Cyanobium, Cyanocystis, Cyanospira, Cyanothece, Cylindrospermopsis, Cylindrospermum, Dactylococcopsis, Dermocarpella, Fischerella, Fremyella, Geitleria, Geitlerinema, Gloeobacter, Gloeocapsa, Gloeothece, Halospirulina, Iyengariella,
  • Prochlorococcus Prochloron, Prochlorothrix, Pseudanabaena, Rivularia, Schizothrix, Scytonema, Spirulina, Stanieria, Starria, Stigonema, Symploca, Synechococcus,
  • Synechocystis Synechocystis, Tolypothrix, Trichodesmium, Tychonema, or Xenococcus species.
  • the genomes and other nucleic acid sequences include modified and synthetic nucleic acid sequences derived from such genomes.
  • the presently described methods are equally applicable to yet unpublished nucleic acid sequences, as they become available, including those that characterize multicellular organisms such as higher plants, such as corn and rice, mammals such as rodents (mice, rats, rabbits, etc.), pigs, cows, bulls, horses, primates, sheep, and companion animals (e.g., dogs, cats, etc.).
  • cells isolated from a human can be used to obtain a donor genome. Many of these are available at the present time.
  • the nucleic acid sequences and genomes include those that do not mimic those present in nature.
  • the genomes and other nucleic acid sequences selected for manipulation by the methods are derived from intractable organisms or other organisms with poor genetic systems or genetic systems that are less desirable than those of host organisms, including certain prokaryotes and other non-yeast organisms, such as those in which common genetic techniques are inefficient, such as double crossover homologous recombination and transposon mutagenesis. These techniques are inefficient, for example, in certain bacterial organisms, such as Mycoplasma species.
  • integration of plasmid DNA by single crossover events has been carried out for targeted addition and disruption of genes in M. mycoides LC (Janis, C, et al. 2005, Appl Environ Microbiol 71 :2888-93), this organism contains a paucity of selection markers, limiting the number of genetic modifications that can be performed in a single M. mycoides LC cell.
  • organisms such as prokaryotes, including those with poor genetic systems, with the potential to produce industrially useful compounds and/or to function in extreme environments (e.g., elevated temperatures, elevated pressure, etc.).
  • Exemplary organisms include those that can be used to produce biofuels.
  • Other exemplary organisms include those that undergo photosynthetic processes.
  • Genomes and organisms can be genetically modified using the methods described herein and known in the art to generate novel genomes and organisms for the production of biofuels. For example, genes involved in photosynthesis and other metabolic processes can be modified to generate organisms that produce oils, e.g., biofuels, in place of glucose or another carbon source.
  • genomes are those of organisms engineered with the capacity to convert sunlight and carbon dioxide into a biofuel.
  • One such exemplary organism is Prochlorococcus marinus, which is among the most abundant photosynthetic organisms on earth but has an ineffecient genetic system.
  • the methods are carried out to modify and engineer genomes, such as whole (complete) genomes (e.g., whole cellular, viral, and organelle genomes), and portions of whole genomes containing genetic material sufficient to effect and/or sustain viability of a cell (minimal cellular genome), viability, within a host cell, of an organism that depends on a host cell for viability (e.g., minimal viral genome), or organelle function within a host cell (minimal organelle genome), under at least one set of environmental conditions.
  • the genome is a minimal genome or a minimal replicative genome.
  • the genome contains additional nucleic acid sequences beyond those found in a minimal genome or in a whole genome.
  • the genome can be naturally-occurring or synthetic, such as genetically engineered genomes including modified genomes and hybrid genomes that contain nucleic acid sequences and/or portions of genomes from more than one species.
  • the genomes are cellular genomes, containing nucleic acid sequences sufficient to cause and/or sustain viability of a cell, e.g., those encoding molecules required for replication, transcription, translation, energy production, transport, production of membranes and cytoplasmic components, and cell division.
  • the genome is a viral or organelle genome.
  • the nucleic acid sequences are organelle nucleic acid sequences, e.g., organelle genomes, such as plastid, e.g., chloroplast, and mitochondrial genomes.
  • organelle genomes such as plastid, e.g., chloroplast, and mitochondrial genomes.
  • Eukaryotic organelles such as mitochondria and chloroplasts, are cytoplasmic DNA-containing, membrane-bound compartments thought to be remnants of endosymbiotic bacteria that became trapped in their hosts.
  • mitochondria are found naturally in all eukaryotic cells and chloroplasts and other plastids are found naturally in plants and algae. Plastid genomes vary in size from 35 to 217 kb, with the majority being between 115 and 165 kb.
  • Mitochondrial genomes vary greatly in size among different species, and can be from below 20 kb to over 350 kb.
  • the methods of the present application allow for engineering of organelle genomes in host cells using nucleic acids and genetic systems within the host cells.
  • organelles can be modified in yeast hosts using yeast plasmids and homologous recombination.
  • the provided methods can be used to generate novel mitochondria or chloroplast genomes, for example, to increase energy production or metabolism in eukaryotic cells, such as yeast and algae.
  • the nucleic acids are viral and bacteriophage nucleic acids, such as viral and bacteriophage genomes.
  • viral and bacterial nucleic acids and genomes can be modified and engineered by the provided methods to generate viruses with therapeutic uses.
  • viral genomes can be cloned and manipulated using the methods described herein. Viruses have been used for gene therapy, vaccines, and as Trojan horses in medical applications; however, viral genomes are too large for manipulation in simple plasmids. Similarly, bacteriophages have been used as antibiotics for decades; yet genomes of the T-phage are too large to be worked with easily.
  • nucleic acid sequences modified, engineered and generated by the provided methods are not genomes.
  • the nucleic acids include chromosomes and other nucleic acid sequences.
  • the nucleic acid sequences and genomes are large nucleic acid sequences.
  • the genome or other nucleic acid sequence is at least at or about 100 kb, 150 kb, 200 kb, 250 kb, 300 kb, 350 kb, 400 kb, 450 kb, 500 kb, 550 kb, 600 kb, 650 kb, 700 kb, 750 kb, 800 kb, 850 kb, 900 kb, 950 kb, 1 megabase (MB), 1.1 MB, 1.2 MB, 1.3 MB,
  • the provided methods are also useful in manipulating and cloning smaller nucleic acid sequences such as, for example, those smaller than about 100 kb. ii. Propagation, isolation, and synthesis of donor genomes and other nucleic acids
  • the nucleic acid sequences Prior to transfer, can be propagated in and/or isolated from cells or tissues.
  • the donor nucleic acid sequences can be isolated from donor cells or tissues (e.g.. cells and tissues from a donor organism) or can be transformed into and propagated within other cells, using well-known cloning, cell, and plasmid techniques and systems.
  • the nucleic acid sequences in the cells can be natural or synthetic, including partially synthetic. In some cases, the nucleic acid sequences are amplified, such as by PCR, after isolation from cells or tissues.
  • Donor nucleic acids can also be chemically synthesized in vitro using chemical synthesis and assembly methods and, thus, are not isolated from any particular tissue or cell prior to use in the described methods.
  • Methods for chemical synthesis of DNA and RNA and assembly of nucleic acids are known, and include oligonucleotide synthesis, assembly, and polymerase chain reaction (PCR) and other amplification methods (such as, for example, rolling circle amplification, whole genome amplification), such as those described herein and in U.S. Application No. 12/247,126, to Gibson et al, filed October 7, 2008.
  • Synthesis of DNA for example, can be from DNA (e.g., by PCR) or from RNA, e.g., by reverse transcription.
  • nucleic acids are synthetic genomes. Synthetic genomes can be produced, for example, as described herein and in U.S. Application No. 12/247,126, by Gibson et al, filed October 7, 2008). iii. Nucleic acid sequences, vector-host systems, and culture conditions
  • nucleic acid sequences can be isolated from a variety of sources, genetically engineered, amplified, and/or expressed generated recombinantly. Recombinant polypeptides generated from these nucleic acid sequences can be individually isolated or cloned and tested for a desired activity. Any recombinant expression system can be used, including bacterial, mammalian, fungal, yeast, insect or plant cell expression systems.
  • nucleic acid sequences can be synthesized in vitro, such as by well-known chemical synthesis techniques, and/or obtained from commercial sources, and optionally assembled, such as for large nucleic acids and genomes, for example, as described in U.S. Application No. 12/247,126, to Gibson et al, filed October 7, 2008.
  • nucleic acid sequences such as, e.g. , subcloning, labeling probes (e.g., random-primer labeling using Klenow polymerase, nick translation, amplification), sequencing, hybridization and the like are well described in the scientific and patent literature.
  • labeling probes e.g., random-primer labeling using Klenow polymerase, nick translation, amplification
  • sequencing hybridization and the like are well described in the scientific and patent literature.
  • Another useful means of obtaining and manipulating nucleic acid sequences used to practice the provided methods is to clone from genomic samples, and, if desired, screen and re-clone inserts isolated or amplified from, e.g., genomic clones or cDNA clones.
  • Sources of nucleic acid sequences used in the described systems and methods include genomic or cDNA libraries contained in, e.g., mammalian artificial chromosomes (MACs), see, e.g., U.S. Patent Nos. 5,721,118; 6,025,155; human artificial chromosomes, see, e.g., Rosenfeld (1997) Nat. Genet.
  • MACs mammalian artificial chromosomes
  • yeast artificial chromosomes YAC
  • bacterial artificial chromosomes BAC
  • PI artificial chromosomes see, e.g., Woon (1998) Genomics 50:306-316
  • Pl-derived vectors PACs; see, e.g., Kern (1997) Biotechniques 23:120-124); cosmids, recombinant viruses, phages or plasmids.
  • Methods for isolation of nucleic acid sequences are well- known. As will be apparent to the skilled artisan, the method of isolation depends upon the type and size of sequence(s) being isolated, the type of organism, and the type of tissue or cell from which the sequence(s) is being isolated. Methods for isolation of genetic material from a variety of organisms are well known and can be used in the present embodiments to isolate donor nucleic acid sequences (genomes). For example, conventional methods for DNA isolation are well known and can be used with the provided methods, depending upon the size, and can be performed with a number of commercially available kits for isolation of nucleic acid sequences; for example, commercially available kits can be used for isolation of genomic DNA from cells.
  • Natural and synthetic nucleic acid sequences can be reproduced, e.g., copied, by amplification. Amplification can also be used to clone or modify the provided nucleic acid sequences. Thus, provided are amplification primer sequence pairs for amplifying the provided nucleic acid sequences. One of skill in the art can design amplification primer sequence pairs for any part of or the full length of these sequences. Amplification can also quantify the amount of nucleic acid sequence in a sample, such as the amount of donor genome in a host cell.
  • Amplification methods are also well known in the art, and include, e.g., polymerase chain reaction, PCR, ligase chain reaction (LCR), transcription amplification (see, e.g., Kwoh (1989) Proc. Natl. Acad. Sci. USA 86:1173); and, self-sustained sequence replication (see, e.g., Guatelli (1990) Proc. Natl. Acad. Sci. USA 87:1874); Q Beta replicase amplification (see, e.g., Smith (1997) J. Clin. Microbiol.
  • nucleic acid sequences are cloned and propagated in bacterial cells, such as Escherichia coli, for example, E. coli DH10B [F ' -mcrA A(mrr-hsdRMS- mcrBC) ⁇
  • E. coli and other laboratory strains are well known and can be used in connection with the provided methods.
  • E. coli cells are grown in medium, e.g., Luria-Bertani (LB) broth medium or in LB agar, at 37°C.
  • medium e.g., Luria-Bertani (LB) broth medium or in LB agar, at 37°C.
  • Nucleic acid sequences can be propagated, for example in Mycoplasma cells.
  • the Mycoplasma cells can be either the donor Mycoplasma cells that naturally contain donor nucleic acids or Mycoplasma cells into which donor nucleic acids, e.g., synthetic genomes, have been transformed.
  • Exemplary Mycoplasma species include, for example, Mycoplasma capricolum subsp. capricolum (strain California KidTM) (ATCC 27343) and Mycoplasma mycoides subsp. mycoides (strain GM12) (Damassa et al, 1983), Mycoplasma capricolum subsp. capricolum (M. capricolum), such as wt M. capricolum and a M.
  • capricolum mutant obtained by inactivation of the CCATC-restriction enzyme gene in wt M. Capricolum as described below in the Examples. Methods for growth of these and other Mycoplasma cells and other cells are known. In one example, Mycoplasma donors are grown at 37°C in liquid or solid SP4 medium (Tully et al. 1977), containing 17% of fetal bovine serum (Invitrogen).
  • Cell culture and plasmid systems can be engineered in order to facilitate selection of cells containing the desired nucleic acid cloned and propagated using known methods.
  • growth of bacterial cells such as E. coli is carried out in medium supplemented with antibiotic or other selection agent, e.g., 50 ⁇ g/ml of ampicillin, 5 ⁇ g/ml of tetracycline or 125 ⁇ g/ml of puromycin, depending on resistance genes located within nucleic acids, e.g., plasmids used for cloning and propagation.
  • Mycoplasma cells that have been transformed with plasmid, donor genomes, or other nucleic acids can be grown in medium, e.g., SP4 medium, supplemented with 5 ⁇ g/ml of tetracycline or 8 ⁇ g/ml of puromycin.
  • medium e.g., SP4 medium, supplemented with 5 ⁇ g/ml of tetracycline or 8 ⁇ g/ml of puromycin.
  • Expression of desired gene products within cells can be detected using well-known methods. For example, ⁇ -galactosidase activity can be detected by plating Mycoplasma or other cell types on solid medium containing 150 ⁇ g/ml of 5-bromo-4-chloro-3-indolyl-P-D- galactopyranoside (X-gal, Promega).
  • X-gal 5-bromo-4-chloro-3-indolyl-P-D- galactopyranoside
  • X-gal 5-bromo-4-chloro-3-indolyl-P-D- galactopyranoside
  • Nucleic acid sequences can be isolated in agarose plugs to minimize damage. Large nucleic acid sequences, such as DNA ranging from a few kilobase pairs to 10 MB and larger (e.g., genomes), can be sheared by mechanical force (e.g., pipetting) during
  • genomic nucleic acid seqeunces in agarose can minimize damage.
  • cells containing the donor DNA are embedded in agarose and lysed.
  • Methods for isolating genomic nucleic acid seqeunces in agarose are also well known, and can be performed using commercially available kits, such as the CHEF Mammalian Genomic DNA Plug Kit (cat. No. 170-3591), the CHEF Bacterial Genomic DNA Plug Kit (cat. No. 170- 3592), and the CHEF Yeast Genomic DNA Plug Kit (cat. No. 170-3593), all from Bio-Rad Laboratories, Hercules, CA. Preparation of DNA using such kits can be carried out using conditions and protocols recommended by the supplier.
  • Donor nucleic acid sequences can also be isolated in agarose plugs using low melting point agarose and the Bio- Rad CHEF Mammalian Genomic DNA Plug Kit, following the protocol suggested by the manufacturer.
  • cells are suspended in medium without serum or PBS and a number of cells (e.g., 5 x 10 7 or 5 x 10 8 per mL agarose to be made) are centrifuged and resuspended in one-half the final volume of agarose to be made.
  • low-melting point agarose e.g., Bio-Rad 2 % CleanCutTM agarose
  • sterile water melted and equilibrated to 50 °C.
  • the cell suspension is equilibrated to the same temperature and mixed gently with the agarose.
  • the mixture is transferred to a plug mold and allowed to solidify.
  • a mixture containing proteinase K is added (e.g., in 5 ML Proteinase K Reaction Buffer, containing 100 mM EDTA, pH 8, 0.2 % sodium deoxycholate, 1 % sodium lauryl sarcosine, and 1 mg/mL proteinase K, per mL plug) and incubated at 50°C overnight.
  • cells in agarose are incubated for a period between overnight and two days at 50°C in 0.4 M EDTA, 0.4 % N-lauroyl sarcosine, 2 mg/mL proteinase K, followed by a buffer change and a second treatment under the same conditions for the same time.
  • agarose plugs containing donor nucleic acids can be dialyzed against 10 mM Tris pH 8.0 (optionally containing EDTA at 1 mM or 50 mM), e.g., for 1 hour. In one example, this dialysis is followed by dialysis for two times, two hours each, in 10 mM Tris, 50 mM ETDA, 0.1 mM PMSF, and re-dialysisin 10 mM Tris, 50 mM EDTA for storage.
  • samples are then dialyzed against 10 mM (6%) PEG6000 (United States Biochemical), 0.6 M NaCl for several hours, as described by Katsura et ah, Electrophoresis 21, 171 (Jan, 2000) prior to transfer.
  • plugs further are melted at 65°C for 5 min, followed by addition of two volumes of 65°C TE and gentle stirring, followed by incubation at 65 °C for 5 minutes.
  • the plugs are melted and digested prior to transformation into host cells using, for example, ⁇ -agarase.
  • plugs are electrophoresed twice by CHEF (Bio-Rad) ⁇ e.g., first on a 1 % pulse-field agarose gel, with 0.5 x TBE and 50-90 second switch time over 20 hours, and second on a 1 % low melting point gel, with 1 x TAE and 60-120 second switch time, over 24 hours), to remove broken DNA but leave genomes intact, followed by dialysis against sterile 1 x TAE and melting at 73°C, equilibration to 42°C, and digestion with ⁇ -agarase (New England Biolabs), for example for 1.5 hours.
  • CHEF Bio-Rad
  • Donor nucleic acid sequences can be an organelle genome, for example, an organelle genome isolated from a donor cell. Methods for isolating organelle genomes are known in the art. Kits for isolating organelles from cells are available from a variety of manufacturers, including Pierce (Rockford, 111.), Sigma-Aldrich (St. Louis, Mo.) and others.
  • Organelle genomes and other nucleic acid can be isolated from various cell types, such as eukaryotic and prokaryotic cells, including yeast cells, plant cells, algae, and mammalian cells. It is understood, as is known in the art, that the isolation procedure may vary depending upon the organism of the cell from which the organelle nucleic acid is isolated and the type of organelle isolated.
  • the organelle nucleic acid isolation procedure can include separation of organelle DNA from nuclear DNA in a total genomic DNA sample, for example, based on molecular weight by fractionation or gel separation.
  • organelles can be purified from total cellular fractions prior to extracting DNA.
  • Methods are known for isolation of chloroplasts (and other plastid) genomes from plants. Isolation is typically carried out by (1) separating plastids from other organelles, (2) lysing chloroplasts, and (3) purifying nucleic acid. In one example, sucrose or Percoll step gradients are used to obtain chloroplasts from cell lysates, which then are lysed and used to recover DNA.
  • plants are placed in the dark to reduce chloroplast starch levels, green leaves washed in tap water, and placed (10-100 g) into isolation buffer for homogenization in a prechilled blender, followed by filtration through cheesecloth and centrifugation.
  • Pellets are loaded onto a one-step gradient with 18 mL 52 % sucrose, overlayed with 7 mL 30 % sucrose, and optionally more sucrose gradients to enhance yield, such as at least six sucrose gradients with 200 g starting material.
  • the step gradients are centrifuged, e.g., at 25,000 rpm for 30-60 minutes at 4°C, and the chloroplast band removed from the 30-52% interface using a wide-bore pipette.
  • Chloroplasts then are lysed as described and centrifuged to remove debris. DNA then is purified, e.g., as described using a CsCl gradient. DNAsel treatment can be used to modify the sucrose gradient method and to destroy nuclear DNA. Alternatively, a high salt (e.g., NaCl 1.25 M) method can be used for isolation without step-gradient centrifugation as is known in the art. Chloroplasts can be sorted from mitochondria and nuclei using fluorescence activated cell sorter (FACS) separation.
  • FACS fluorescence activated cell sorter
  • mtDNA mitochondrial DNA
  • Highly purified mtDNA can be prepared from vertebrate or invertebrate tissues using sucrose pad gradients and cesium chloride gradients.
  • tissue e.g., brain, testes, ovary, liver, kidney, heart, skeletal muscle of vertebrates and invertebrate embryos
  • tissue is prepared from the organism, if necessary, by homogenizing the tissue, followed by repeated centrifugation at low speed to remove cellular debris and nuclei, followed by centrifugation at high speeds and lysis of the pellet.
  • a sucrose step gradient can be performed on the pellet by ultracentrifugation at 25,000 rpm for 1 hour at 4°C, in which the gradient contains a pad of 10 mL of 1.5 M sucrose in TE buffer and a 10 mL layer of 1 M sucrose.
  • the pellet is resuspended in TE buffer and layered on the gradient, followed by centrifugation for 1-2 hours at 27,000 rpm at 4°C.
  • Highly purified mitochondria which appear as a milky white band at the interface of the 1 and 1.5 M sucrose pads, is collected with a Pasteur pipette and placed in a tube for further concentration by high-speed centrifugation (e.g., 13,000 rpm at 4°C).
  • the pelleted mitochondria are then lysed, for example, by resuspending in TE buffer and adding sodium dodecyl sulfate (SDS) (e.g., 0.3 mL 10 % SDS per mL TE). After the solution appears clear, saturated CsCl is added and the mixture incubated on ice for 20 minutes, followed by centrifugation at 12,000 rpm for 10 minutes, saving the supernatant.
  • SDS sodium dodecyl sulfate
  • DNA is purified with a CsCl-propidium iodide (PI) (or ethidium bromide) gradient (e.g., 8 g CsCl and 0.6 mL 2 mg/ml PI per 8 mL supernatant, mixing and adjusting density to 1.56 g/mL).
  • PI CsCl-propidium iodide
  • ethidium bromide e.g., 8 g CsCl and 0.6 mL 2 mg/ml PI per 8 mL supernatant, mixing and adjusting density to 1.56 g/mL.
  • the upper band contains nuclear DNA and the lower band contains mtDNA, which can be collected and, optionally, followed by further CsCl gradients. Kits are commercially available for isolation of mtDNA.
  • mitochondrial DNA is prepared from mammalian tissue, such as muscle or liver tissue, using the mtDNA Extractor CT Kit (Wako, Osaka Japan, Catalog No.: 291-55301). Protocols for isolation of fungal (e.g., yeast) mitochondrial DNA, which cannot typically be released efficiently into cell homogenates as supercoiled circular DNAs, are also known. In one example, CsCl density gradient centrifugation on crude DNA preparations is carried out in the presence of a dye that binds preferentially to AT rich DNA (e.g., DAPI or 6zs-benzimide).
  • a dye that binds preferentially to AT rich DNA e.g., DAPI or 6zs-benzimide
  • mtDNA is extracted from isolated mitochondria, which are isolated by pelleting nuclei and cellular debris by centrifugation of lysates at 2000g for 20 minutes, followed by separation of supernatants on sucrose gradients (e.g., 2 mL 60 % sucrose, overlayed with 4 mL 50 % sucrose, overlayed with 4 mL 44 % sucrose, followed by centrifugation at 120,000g for 90 minutes in a swing-out rotor and collection of the mitochondria from the 44/55% interface).
  • sucrose gradients e.g., 2 mL 60 % sucrose, overlayed with 4 mL 50 % sucrose, overlayed with 4 mL 44 % sucrose, followed by centrifugation at 120,000g for 90 minutes in a swing-out rotor and collection of the mitochondria from the 44/55% interface.
  • the provided embodiments are methods and nucleic acids for introducing the donor nucleic acids, including the donor genomes, into host cells, for example, for modification within the host cells using host cell machinery.
  • the host cell is a heterologous host cell that provides desired capabilities typically not present in the cell from which the donor genome is derived, e.g., recombination machinery.
  • the donor nucleic acid can be isolated from a cell or tissue prior to transfer, chemically synthesized and/or assembled in vitro, and/or copied from such an isolated or synthesized nucleic acid by in vitro or in vivo methods.
  • transfer of the donor nucleic acid (e.g., donor genome) into the host cell is carried out by first joining the donor nucleic acid (which may be circular, linearized, or fragmented) to a host nucleic acid which, typically, is a host vector, in order to generate a nucleic acid containing the donor nucleic acid and the host vector, which can be propagated and modified within the host cell.
  • Joining of the donor nucleic acid to the host vector can be carried out in vitro or in vivo in the donor cell or in the host cell.
  • the host vector is transformed into the donor cell, where it is recombined with the donor genome, followed by isolation of the genome with the vector insertion (see, e.g., Figure 2A).
  • the donor genome and a host vector are separately cotransformed into the host cell and the donor and host nucleic acids recombine within the host cell.
  • the donor genome may be linearized (see, e.g., Figure 2B), or fragmented (see, e.g., Figure 2C) prior to cotransformation with the vector into the host cell.
  • the host cells can be any host cells, and typically are heterologous cells having genetic systems that are desirable for modification of nucleic acids in the laboratory, for example, improved genetic systems compared to the donor organisms or cells.
  • Exemplary aspects of desirable genetic systems are the ability to support homologous recombination, including double crossover homologous recombination, and transposon mutagenesis, a defined and well -characterized set of selection and other markers, and the capacity for cloning large nucleic acids. It is also desirable that the host cell has properties that make it compatible with the donor nucleic acid during cloning, propagation, and modification of the nucleic acid within the host cell.
  • particular host cells can be selected to minimize gene toxicity.
  • Host/donor combinations can be selected such that gene expression from donor nucleic acids does not occur in the host cell or is reduced in the host cell compared to in the donor cell.
  • the host and donor contain different translation and/or transcription signals and/or machinery, such as yeast and bacterial organisms.
  • one or more codon is translated as an amino acid by the donor but is treated as a stop codon by the cell machinery.
  • the donor translates the codon (e.g., UAG) as an amino acid (e.g., tryptophan) while the host cell reads the same codon as a stop codon (e.g., Mycoplasma versus eukaryotic organisms).
  • donor genomes and other nucleic acids can be maintained, replicated, and modified within host cells having desirable genetic systems without (or with minimal) expression of gene products encoded by the donor genome.
  • the host cell can include any cell compatible with the cloned donor genome or nucleic acid.
  • genomes from algae may be cloned into yeast and manipulated to provide more favorable characteristics when re-introduced into the same or different algal recipient cell.
  • these algal genes can also be manipulated and provided to plant cell cultures. Similar manipulations are feasible for vertebrate and invertebrate cells.
  • the host cell is a yeast cell.
  • Yeast hosts include the "workhorse species,” Saccharomyces cerevisiae, and other yeast species such as
  • Saccharomyces pombe which can be used to clone even larger genomes.
  • Yeast hosts are particularly suitable for manipulation of donor genomic material because of their unique set of genetic manipulation tools.
  • the natural capacities of yeast cells, and decades of research have created a rich set of tools for manipulating DNA in yeast. These advantages are well known in the art.
  • yeast with their rich genetic systems, can assemble and reassemble nucleotide sequences by homologous recombination, a capability not shared by many readily available organisms.
  • Yeast cells can be used to clone larger pieces of DNA, for example, entire cellular, organelle, and viral genomes that are not able to be cloned in other organisms.
  • one embodiment of the described methods utilizes the enormous capacity of yeast genetics to advance synthetic biology and synthetic genomics by using yeast as host cells for manipulation of genomes of intractable and other organisms and synthetic genomes.
  • yeast host cells are yeast strain VL6-48N, developed for high transformation efficiency parent strain: VL6-48 (ATCC Number MYA-3666TM)), the W303a strain, and recombination-deficient yeast strains, such as the RAD54 gene-deficient strain, VL6-48-A54G (MATa his3-A200 trpl- ⁇ ura3-52 lys2 ade2- 101 met 14 rad54-Al:: kanMX), which can decrease the occurrence of a variety of recombination events in yeast artificial chromosomes (YACs).
  • VL6-48N developed for high transformation efficiency parent strain: VL6-48 (ATCC Number MYA-3666TM)
  • W303a strain the W303a strain
  • recombination-deficient yeast strains such as the RAD54 gene-deficient strain, VL6-48-A54G (MATa his3-A200 trpl- ⁇ ura3-52 lys2 ade2- 101 met 14 rad
  • yeast can be used to introduce a number of different genetic modifications, including single nucleotide changes (e.g., insertions, deletions, mutations), modification of target nucleic acid portions and regions, and construction of entirely new chromosomes.
  • Serial modifications to a cloned copy of an otherwise intractable genome or other large nucleic acid can be performed in yeast in rapid succession.
  • the mating capacity of yeast is favorable for modifying genomes and other large nucleic acids.
  • Yeast recombination machinery when activated during yeast mating, can be used to generate libraries, e.g., combinatorial libraries containing variants of cloned genomes or nucleic acids.
  • Yeast Artificial Chromosome (YAC) libraries have been constructed for several different bacteria (Azevedo et al, PNAS USA 90, 6047 (1993); Heuer et al, Electrophoresis 19, 486 (1998); Kuspa et al, PNAS USA 86, 8917 (1989).
  • Large prokaryotic DNA segments can be cloned in yeast using the universal genetic code.
  • Toxic gene expression typically is not a barrier to cloning donor nucleic acids in yeast.
  • yeast The transcription (Kozak, Gene 234, 187 (1999)) and translation (Kornberg, Trends Cell Biol 9, M46 (1999) signals in yeast are different from those in bacteria. In fact, most prokaryotic genes likely are not expressed in yeast. There is no restriction barrier in yeast (Belfort and Roberts, Nucleic Acids Res 25, 3379 (1997). If there is a barrier, it may be a replication barrier, rather than a gene expression barrier (Stinchcomb et al., PNAS USA 77, 4559 (1980)). Gene toxicity is minimized because regulation of gene expression in a eukaryote such as yeast is different from that in
  • Mycoplasmas use the codon UGA for tryptophan rather than as a translation stop signal. Thus, most Mycoplasma genes, if expressed, would produce truncated proteins in yeast. This largely avoids the possibility of toxic gene products.
  • Donor may be obtained in their native form from donor organisms and modified with yeast vectors prior to transformation into yeast, or may be assembled from natural or synthetic fragments together with yeast vectors prior to transformation into yeast cells or simultaneously co-transformed into yeast cells. New organisms are created by transferring these genomes, which have been optionally manipulated as desired, into compatible recipient cells.
  • suitable techniques for transferring genomes to yeast host cells modifying the genomes within host cells while maintaining their stability and integrity, and transplanting the cloned and manipulated genomes from yeast host cells back into recipient cells that more closely resemble the original donors, thus creating organisms which previously did not exist and/or could not have been created through genetic manipulation of their original cells with available genetic engineering and cloning tools.
  • donor nucleic acids are transformed into and propagated within host cells using host vectors.
  • the host cell generally contains, or will support introduction of, a host vector for transfer, maintenance, and modification, of the donor nucleic acid within the host cell.
  • the host vector contains nucleic acid sequences to facilitate transfer of the donor nucleic acid to and from a donor cell, a host cell, and a recipient cell, and other cells, such as bacterial cells used for cloning and propagation (e.g., E. coli), such as the tri-shuttle vectors described in the examples herein (see, e.g., Figure 3).
  • the vector contains any nucleic acids (e.g., origin of replication) needed to promote replication of the vector within one or more desired cell type and selection and/or resistance markers for use with the different cell type(s).
  • nucleic acids e.g., origin of replication
  • Resistance markers are well known. The skilled artisan will be able to determine appropriate resistance markers for different host/donor combinations. In some cases, it can be desirable to use markers that are not clinically relevant. In other cases, the choice of resistance marker depends on properties of the donor, host, and/or recipient cells. For example, antibiotics that target the cell wall may not be useful in Mycoplasma and other organisms lacking cell walls.
  • antibiotics that target the cell wall may not be useful in Mycoplasma and other organisms lacking cell walls.
  • the resistance markers are genes encoding antibiotic resistance, such as ampicillin, kanamycin, and tetracycline resistance, such as the tetracycline resistance protein (TetM), and chloramphenicol acyltransferase (CAT), aminoglycoside resistance protein (aacA/aphD), and combinations thereof.
  • tet-resistance markers are useful in bacteria, such as Mycoplasma, in which tetracyclines have a potent effect and which exhibit low levels of spontaneous resistance.
  • Genes conferring Puromycin resistance also can be used, for example, for cloning and modifying Mycoplasma nucleic acids and using Mycoplasma cells.
  • Puromycin is an antibiotic that mimics the 3 '-terminal end of aminoacylated tRNA and attaches to the carboxyl-terminus of growing protein chains to stop protein synthesis. Because puromycin conscripts rRNA recognition elements used by all of the various tRNAs in a cell, it is unlikely that spontaneous antibiotic resistance can be acquired via a simple point mutation, which can happen with other markers in some cases. Puromycin is readily available, relatively inexpensive, is not used in the clinic and it is a potent inhibitor of translation in both prokaryotes and eukaryotes. No known rRNA based resistance exists.
  • a codon-optimized cassette has been developed to confer puromycin resistance in five different Mycoplasma species and can function in E. coli, making it functional in shuttle vectors.
  • the 597 bp puromycin N-acetlytransferase 85 gene (PAC) is synthesized using overlapping oligonucleotides as described by Smith et al. , PNAS USA
  • oligonucleotides encoding both strands of a codon optimized version (for expression in M. genitalium) are ordered from IDT (Coralville, IA).
  • the oligos are 48 bases 88 long with an overlap of 24 bases.
  • the top-strand and bottom- strand oligos are mixed, heated to 95°C, and slow cooled to allow annealing of the overlaps.
  • the reactions are ligated for 12 hours and used as a template for PCR.
  • the PCR amplicon is cloned into pGEM-3Zf(+) (Promega, Madison, Wisconsin) and sequenced to identify correct PAC clones.
  • the optimized PAC gene is then cloned under the control of the Spiroplasma citri spiralin promoter (Ps) and used to replace the tetM gene in a derivative of Mini- Tn4001tet, as well as the tet gene in pMycol (Lartigue et al. (2003), Nucleic Acids Res 31 : 6610-8).
  • the new plasmid (Mini-Tn4001PsPuro) can be used to transform M. genitalium, M. gallisepticum and M. pneumoniae, while the pMycol derivative (pMycoPuro) can be used to transform M. mycoides LC and M. capricolum.
  • the vectors further include nucleic acids that allow joining of the vectors with the donor nucleic acids.
  • the host vector contains regions of homology to portions of the donor genome or nucleic acid, such as regions of homology at the 3' and 5' termini of a linear vector that are homologous to adjacent regions within the donor nucleic acid, to facilitate joining by homologous recombination.
  • the host vector contains nucleic acid encoding a transposase and/or inverted repeats, to facilitate joining, e.g., insertion, into the donor nucleic acid, such as within a donor cell.
  • the host vectors can additionally contain restriction enzyme recognition sites and nucleic acids to support replication and segregation within host cells and other cells.
  • a yeast host vector contains an origin of replication (e.g., high copy origin from pUC19); one or more resistance markers and/or selection markers (e.g., antibiotic resistance genes and selectable host cell (e.g., yeast) markers), such as markers for selection in the host cell, in donor cells and in recipient cells.
  • an origin of replication e.g., high copy origin from pUC19
  • resistance markers and/or selection markers e.g., antibiotic resistance genes and selectable host cell (e.g., yeast) markers
  • resistance/selection markers are antibiotic resistance genes (e.g., ampicillin-resistance genes, kanamycin resistance genes and other well-known antibiotic resistance genes), and other antibiotic resistance genes; selectable yeast or other host cell markers, e.g., HIS3) and/or selection markers; nucleic acids to facilitate insertion into donor nucleic acid, e.g., transposase and inverted repeats, such as for transposition into a Mycoplasma genome; nucleic acids to support replication and segregation in the host cell, such as an autonomously replicated sequence (ARS), centromere sequence (CEN).
  • the vector contains a telomere sequence.
  • Exemplary vectors include yeast vectors, including yeast centromeric plasmids, e.g., Yeast Artificial Chromosome (YAC) vectors, such as pmycYACTn, described in Example lA(i)(a), below, and illustrated in Figure 3A; and the miniTn-Puro- JC VI- 1.7 vector constructed shown in Figure 3B.
  • YAC Yeast Artificial Chromosome
  • the pmycYACTn vector include: (i) a high copy origin from pUC19 and an ampicillin resistance marker for propagation in E. coli, (ii) the IS256 (iii) tetM and lacZ markers, both expressed from spiralin promoters (16, 17), for selection and screening in E.
  • the miniTn-Puro- JCVI-1.7 vector differs from pmycYACTn as follows: (i) it does not contain lacZ and substitutes a puromycin resistance marker for tetM and (ii) it contains a bacterial artificial chromosome (BAC) vector, for possible cloning in E. coli. iii. Cloning strategies: joining of host and donor nucleic acids and transfer of donor genomes and nucleic acids into host cells
  • the donor genome or other nucleic acid is joined to a host nucleic acid, typically a host vector, to generate a nucleic acid containing the donor nucleic acid and host nucleic acid that can be propagated and manipulated in the host cell.
  • Joining the host and donor nucleic acids can be carried out using a number of approaches, drawing on well-known cloning methods. Three general approaches are described in more detail below.
  • yeast cells are exemplary host cells.
  • Large DNA molecules have been stably cloned in yeast by the addition of a yeast centromere (CEN), which allows the molecules to be segregated along with the yeast chromosomes.
  • CEN yeast centromere
  • Such molecules have been cloned both in the linear form by the addition of telomeres to the ends, and also as circles. Because bacterial genomes are generally circular, and circles can be readily separated from linear yeast chromosomes, it can be advantageous to clone bacterial genomes as circular.
  • the donor genome is joined to the host vector within a donor cell, or other cell type that is similar to the donor, followed by isolation of the nucleic acid containing the donor genome and host vector and subsequent transfer into the host cell.
  • An example is illustrated in Figure 1. This approach can be used, for example, to transfer genomes and other large nucleic acids into host cells.
  • the host vector contains inverted repeats and/or nucleic acids encoding a transposase, to facilitate insertion into the donor genome or other nucleic acid within the cell.
  • joining the donor and host nucleic acids within a donor cell or a cell that is similar to the donor provides advantages. For example, it allows selection of vector insertions (e.g., sites of vector insertion along the length of the donor nucleic acid) that do not or are unlikely to impair or otherwise affect donor cell viability. This approach does, however, require that the donor or similar cell is amenable to introduction of foreign nucleic acid (e.g., transformation), so that the vector can be integrated into the donor nucleic acid, e.g., genome.
  • Various approaches for introduction of nucleic acids into a variety of cell types are well known.
  • yeast host vectors are transformed into bacterial cells in the presence of PEG.
  • Donor cells containing the host vector integrated into the donor nucleic acid are selected, for example, based on a resistance or other selective marker in the host vector.
  • Nucleic acids can be isolated from the donor cells, such as in agarose plugs as described above, for
  • the nucleic acid containing the donor genome and host vector is transplanted into a recipient cell to confirm that the nucleic acid can be transplanted and is compatible with a particular recipient cell.
  • transplantation of genomic DNA from one bacterial species to another can be carried out as described in Lartigue et al, Science 317, 632 (2007) and as described in Example 1 A(ii)(b), below.
  • a linear yeast host vector is joined to a circular bacterial genome within the bacterial cell.
  • the resulting circular nucleic acid is isolated, for example, in agarose plugs as described above, and transformed into yeast host cells. An example of this process is described in Example 1 A, below.
  • the donor genome and the host vector are co-transformed, together or separately, into the host cell, whereupon they join, e.g., by homologous recombination, within the host cell.
  • This approach is advantageous in its simplicity, with minimal sample handling and number of steps.
  • the vector is inserted into the donor genome or nucleic acid by homologous recombination.
  • a linear yeast host vector is co-transformed into a yeast host cell along with a circular bacterial genome, for example, a synthetic genome or one that has been isolated from a donor cell, e.g., in agarose plugs as described above.
  • the host vector contains region(s) of homology to the donor genome or nucleic acid.
  • the linear yeast vector contains, at each terminus, a region of homology to a portion of the bacterial genome.
  • the bacterial genome is cut with a restriction enzyme that cuts near the region of homology to the host vector, prior to transformation into the host cell.
  • This process generates a double-strand break near the insertion site of the vector, greatly improving efficiency of the joining of the host vector and donor genome (by insertion of the vector into the genome) within the host cell.
  • a restriction enzyme recognition site is chosen within the donor genome or other nucleic acid that is compatible with maintaining genome or nucleic acid integrity after insertion of the vector at that site.
  • An example of this process is described in Example IB, below.
  • An example of a third approach, which a modification of the second approach, is illustrated in Figure 2C. This approach is carried out by co-transforming into the host cell, along with the host vector, a plurality of overlapping nucleic acid fragments, which are fragments of the donor genome or nucleic acid.
  • each of the fragments contains homology to a region of the donor genome or nucleic acid and the regions of homology overlap along the length of the donor genome or nucleic acid.
  • the fragments and vector recombine upon transformation into the host cell, for example by homologous recombination through regions of homology.
  • overlapping fragments of a circular bacterial donor genome are co-transformed into a yeast host cell along with a linear yeast vector.
  • the yeast vector contains regions of homology at its termini to portions of the bacterial genome.
  • the fragments and vector recombine, thereby joining the donor genome and host vector.
  • Example 1C An example is described in Example 1C, below.
  • the donor nucleic acids are isolated from the donor cells or similar cells, as described above.
  • large donor nucleic acids including genomes can be isolated from donor and other cells in agarose plugs, as described above.
  • donor nucleic acids are transformed into host cells.
  • the host vector is not previously joined to the donor nucleic acid, it can be transformed into the host cell simultaneously or sequentially in any order, using the same transformation methods.
  • Transformation methods are well known in the art and will vary depending upon the host cell.
  • yeast spheroplasts are prepared from the yeast host cells, for example, as described below, and the nucleic acids are transformed by mixing with the spheroplasts.
  • the transformation is performed in the presence of PEG.
  • donor nucleic acids and/or host vectors can be incubated with the spheroplasts for 10 minutes at room temperature, followed by addition of 800 PEG 8000 and gentle mixing by inversion and another 10 minute incubation at room temperature.
  • spheroplast preparation and transformation is carried out as described by Kouprina and Larionov, Nat Protoc 3, 371 (2008).
  • the OD to which cells are grown can be modified.
  • yeast medium is inoculated with single-cell colonies of yeast hosts, and grown overnight at 30 °C with vigorous shaking to ensure good aeration until an appropriate OD660 is reached.
  • Samples can be centrifuged and resuspended in sorbitol by vortexing, centrifuged and resuspended, e.g., in SPE solution (1 M sorbitol, 0.01 M sodium phosphate, 0.01 M Na 2 EDTA, pH 7.5).
  • Yeast cell walls are removed, for example using ZymolaseTM.
  • the level of spheroplasting can be evaluated by comparison of optical densities of the cell suspension in sorbitol solution versus 2 % SDS solution (in which spheroplasts are lysed).
  • Spheroplasts can be centrifuged and resuspended in 1 M sorbitol by very gentle rocking, and washed and resuspended in STC solution (1 M sorbitol, 0.01 M Tris-HCl, 0.01 M CaCl 2 , pH 7.5). Transformation is carried out by mixing the nucleic acids with the spheroplasts, as described above, optionally in the presence of PEG.
  • a selection procedure typically is performed to select cells into which donor nucleic acids and host vectors have been successfully transformed.
  • the spheroplasts can be centrifuged and resuspended in SOS solution and incubated for 40 minutes at 30°C without shaking.
  • the spheroplasts can be placed in selection medium, such as melted SORB-TOP-His selection medium as described herein, and equilibrated at 50°C, and plated on plates containing selection medium and grown, e.g., at 30 °C until transformants are visible.
  • selection medium such as melted SORB-TOP-His selection medium as described herein
  • donor nucleic acids transformed into host cells can be isolated and analyzed, both before and after modification within the host cells by the provided modification methods. As with isolation from donor and other cells, isolation methods will vary depending on the cell type.
  • native host nucleic acids are removed or reduced from isolated nucleic acid samples, e.g., to isolate or enrich for donor nucleic acids. This process can be carried out by pre-electrophoresis to remove chromosomal host DNA, or by digestion with restriction enzymes that digest host, but not donor, nucleic acids.
  • the donor nucleic acids are isolated in agarose plugs, for example, using the protocol "Preparation of Agarose Embedded Yeast DNA" from the Bio-Rad CHEF- DR III manual, and as described in the Examples herein.
  • agarose plugs containing DNA from the host cells are pre-electrophoresed at constant voltage for several hours to remove yeast host chromosomal DNA.
  • the removal of host DNA can be carried out by first digesting DNA in the plugs with AsiSl, Fsel, and Rsrll, or other enzymes that cleave yeast chromosomes but do not have recognition sites in donor nucleic acids, such as M. genitalium or M. mycoides LC.
  • Analysis of donor nucleic acids can be carried out by any of a number of well- known methods for analyzing DNA. Typically, it is desired to carry out methods to confirm the size and/or sequence of the nucleic acids, and the correct insertion and orientation of the vector and other nucleic acids, including the confirmation of any modifications.
  • isolated DNA is subject to linearization by heating and/or restriction digestion, followed by separation by gel electrophoresis, such as field-inversion (Bio-Rad FIGE
  • Analysis can be, for example, by PCR, such as multiplex PCR, with primers designed to bind various regions along the length of the desired donor nucleic acid or modified donor nucleic acid. Typically, primers also are designed to recognize the host vector.
  • the analysis is carried out by visualization of the size of the isolated nucleic acid on a gel, or by restriction digestion and performing a Southern blot or other hybridization method, for example, as described in Gibson et al, Science 319, 1215 (2008). Specific examples of MPCR and Southern blot analysis of isolated donor genomes are described in the Examples. Modifications of the analysis methods will be apparent to the skilled artisan. Sequencing methods are well known and also can be used to analyze the donor nucleic acids transferred into and propagated within host cells. v. Generation of host cells containing a plurality of donor genomes
  • a plurality of donor nucleic acids are introduced into a single host cell.
  • a host cells containing one donor genome or other donor nucleic acid is crossed to another such host cell containing transferred nucleic acid from a different donor, generating a host cell containing both nucleic acids.
  • a diploid yeast strain containing two donor genomes from different donors such as two Mycoplasma genomes from different species, can be generated by crossing two different haploid strains, each carrying one of the donor genomes. Crossing haploid yeast strains can be carried out using well-known methods.
  • a HIS3 and TRP marker can be introduced into two different haploid cells, respectively, carrying different donor genomes, followed by selection of diploid cells on medium lacking histidine and tryptophan, as described in the Examples herein.
  • the methods are carried out by introducing one or more targeting construct into the donor nucleic acid.
  • the constructs contain portions of homology to the donor nucleic acid, resistance genes, selectable markers, nucleic acids encoding enzymes, such as restriction enzymes, restriction sites, and/or other nucleic acids used in cloning and homologous recombination.
  • the constructs are introduced into host cells containing the donor nucleic acids.
  • a target region of the donor nucleic acid ⁇ e.g., the donor genome) is selected for modification. It is not necessary that the methods modify each residue of the target region. For example, one or more target portions or target positions within the target regions can be modified. The modifications include insertions, deletions, mutations, substitutions, and/or other modifications of one or more nucleotides within the target region.
  • the donor nucleic acid is seamlessly modified.
  • the target region or a portion thereof first is replaced with a nucleic acid construct containing a marker, such as a counter-selectable marker.
  • the marker is then removed from the nucleic acid, by deleting the marker or replacing it with another nucleotide sequence.
  • the marker and surrounding portions are replaced by introducing a second nucleic acid construct having homology to portion(s) of the target region near or adjacent to the marker.
  • This second construct need not be less than 100 % homologous to the portion(s) of the target region.
  • the constructs can contain one or more mutation, deletion, or insertion, compared to the portion of the target region, whereby the target region is modified upon replacement with the construct.
  • the methods typically include one or more homologous recombination step.
  • removal of the marker is facilitated by introducing a break (e.g., double-strand break) in the nucleic acid sequence of the donor nucleic acid containing the construct with the marker.
  • the break is introduced near, e.g., adjacent to, or within the target region.
  • the break is generated by inducibly expressing an enzyme, such as an endonuclease, that recognizes and cleaves the desired nucleic acid sequence.
  • the enzyme is encoded by a nucleic acid within the target region or the construct inserted into the target region.
  • removal of the marker is facilitated by introducing a nucleic acid sequence into the donor nucleic acid, whereby insertion of the nucleic acid sequence generates tandem repeat regions flank the target region or portion thereof.
  • the nucleic acid sequence is included as part of the targeting construct.
  • the methods include both introduction of a break and introduction of a sequence generating tandem repeat regions.
  • the method is a Tandem Repeat with Endonuclease Cleavage (TREC) method, in which double-strand breaks, generated by an inducibly expressed enzyme, and tandem repeats are used to facilitate recombination events and avoid damage and unwanted mutation.
  • TRIP Tandem Repeat with Endonuclease Cleavage
  • the provided methods provide advantages compared to conventional and other available methods, particularly for modification of donor nucleic acids in hosts of different species, genera, and orders.
  • the donor nucleic acids which may come from a donor organism having a poor genetic system, are modified within a host having a rich genetic system, such as a yeast host cell, for example, by homologous recombination methods.
  • a host having a rich genetic system such as a yeast host cell
  • homologous recombination methods for example, nucleic acid fragments several hundred kb in length can be cloned and manipulated in yeast (Saccharomyces cerevisiae) host cells using well-known methods and standard genetic tools, including linear and circular forms of yeast artificial chromosomes (YAC).
  • Transplantation of modified donor nucleic acid to recipient cells can be used, for example, for functional studies of genes and gene regulation, and for production of modified gene products.
  • the provided methods can be used to successfully modify donor genomes within host cells, to engineer and modify genomes from organisms that are genetically intractable.
  • the modification methods can be used to produce genomes, organisms, and gene products produced by the organisms that are commercially useful, such as for production of vaccines, drugs, biological proteins and chemicals, biofuels, and protein therapeutics such as enzymes and antibodies.
  • donor genomes are modified to produce new immunological compositions to elicit an immune response, such as live viruses and other immunogens.
  • donor genomes are modified for the production of biofuels, for example, by introducing DNA encoding for enzymes involved in the oil synthesis pathways, for example, by replacing metabolism pathway genes with those for biofuel production.
  • the donor genome e.g., of a photosynthetic bacteria
  • the recipient cell produces biofuels in place of normal photosynthesis products, such as glucose.
  • biofuels in place of normal photosynthesis products, such as glucose.
  • Other uses are discussed hereinbelow.
  • the provided methods can be used to directly engineer or redesign a synthetic bacterial genome in vivo, for example, in yeast host cells.
  • the provided modification methods include aspects for overcoming
  • a donor nucleic acid such as a donor genome
  • the donor nucleic acid does not typically contribute to the viability of the host cell, or does not contribute to the viability of the host apart from individual selection marker(s) present in the cloning vector.
  • the donor and host are different types of organisms, such as of different orders or kingdoms, for example, when the donor is a prokaryote and the host is a eukaryote, such as a yeast.
  • the M as discussed in the study described in Example 4, below, the M.
  • genitalium genome propagated as a circular YAC (with a histidine marker) in yeast does not have functional complementation with its host, except histidine prototrophy. Any deletion and rearrangement in the bacterial genome is likely neutral for the yeast host.
  • the provided methods overcome these problems and can be used to propagate and modify donor genomes within host cells, while minimizing the risk of unwanted mutations within the donor genome.
  • the provided methods can accurately modify a donor genome, such as a bacterial genome, cloned in yeast host cells, with high efficiency
  • the provided methods further can be used in seamless modification of the donor nucleic acids within the host cells, including mutation, deletion, and/or insertion of nucleotides within a target region of the donor nucleic acid, where no unwanted additional nucleic acid sequence is added or removed. i. Counter-selectable markers
  • a first step of the methods includes introduction of counter-selectable markers into the donor nucleic acid.
  • the markers typically are inserted by homologous recombination, whereby a portion of the target region is replaced with the counter-selectable marker.
  • Counter-selectable markers are advantageous in that both the presence and the absence of the marker can be selected for. The presence of the marker is selected for with one set of growth conditions, while the absence is selected for with a different set of growth conditions.
  • An exemplary, well-known counter-selectable marker is the URA3 yeast gene. The presence of the URA3 gene in a yeast host allows its growth on medium lacking uracil.
  • the genetic marker URA3 can be integrated, e.g. , through
  • homologous recombination into a target region within the donor nucleic acid cloned in a host cell. Integration of the marker is selected for by growth on medium lacking uracil. Removal of the marker, e.g., by deletion or replacement with another nucleotide sequence, for example in a second round of homologous recombination, is selected for by counter-selection, e.g., on 5-FOA.
  • Methods employing counter-selectable markers are desirable in that they can be used for seamless modification. Further, replacement or removal of the counter-selectable marker restores auxotrophy (e.g., dependence on uracil), such that the host cells can be modified using the same method in further rounds of modification.
  • auxotrophy e.g., dependence on uracil
  • Methods are available for introducing and replacing counter-selectable markers in yeast host cells.
  • methods are known for introduction of a URA3 marker by a first round of homologous recombination and replacement of the marker with a second round of homologous recombination.
  • An example of such a method is described in Example 4A, below, in which a site-specific mutagenesis was performed to correct a single base cytidine deletion (309,388) found in the CDS139 locus of a donor synthetic M. genitalium genome maintained in yeast using this conventional method involving two sequential homologous recombination events.
  • yeast hosts containing the mutant bacterial donor genome were transformed with a cassette containing the URA3 marker and 50 bp terminal portions homologous to portions of the target region, replacing a target region containing the single-base deletion CDS 139 locus.
  • the second round of transformation introduced a construct containing the non-mutant DNA sequence back to the same locus, replacing the marker.
  • a break such as a double-strand break (DSB)
  • DSB double-strand break
  • This process typically is carried out by inducibly expressing an enzyme, such as an endonuclease, e.g., l-Scel, that recognizes and cleaves a nucleotide sequence located in proximity to or within the target region.
  • an enzyme such as an endonuclease, e.g., l-Scel, that recognizes and cleaves a nucleotide sequence located in proximity to or within the target region.
  • the enzyme is an endonuclease or other enzyme that generates double-strand breaks.
  • the targeting construct containing the selectable marker further contains a gene encoding the enzyme, under the control of an inducible promoter.
  • the construct further includes a recognition sequence of the enzyme. This construct is introduced into the donor nucleic acid within the host cell. Expression of the enzyme is induced by growth of the host cells under particular conditions that induce expression from the inducible promoter.
  • the promoter is the GAL1 promoter, expression from which can be induced by growth on medium containing galactose as the only carbon source.
  • Example 4B(i) Available methods include inducible introduction of double-strand breaks for the purpose of improving efficiency of recombination-based modification in yeast.
  • One such method, Delitto perfetto is described in Storici et al, Nat Biotechnol, 19, 773-776 (2001).
  • An example of this method is described in Example 4B(i), below. The method is based on the showing that introduction of a double-strand break (DSB) in a target nucleic acid stimulates recombination by several orders of magnitude (Storici et.al, PNAS USA, 100, 14994-99 (2003)).
  • Dilletto perfetto was used in attempt to correct the same single- base deletion in the CDS 139 locus of the M. genitalium donor genome. The process is illustrated in Figure 10A.
  • the targeting construct for introducing the marker further contains a nucleic acid sequence, the introduction of which results in tandem repeat regions flanking the target region or portion thereof containing the inserted marker.
  • the construct initially is inserted, either upstream or downstream of the target region or portion thereof, and contains a nucleic acid portion having homology to a portion downstream or upstream of the target region or portion, respectively. Insertion of this portion near the homologous portion in the target nucleic acid generates the tandem repeats.
  • insertion of the construct generates a portion within the target nucleic acid, 5' of the marker, having homology to a portion 3' of the target region. In another example, insertion of the construct generates a portion within the target nucleic acid, 3' of the marker, having homology to a portion 5' of the target region.
  • the modified donor nucleic acid e.g., modified donor genome
  • tandem repeat sequences facilitates homologous recombination between the two sequences, for example, for removal of a portion of the construct containing the counter-selectable markers.
  • Such methods are well known and based on a precise excision of a nucleic acid segment by homologous recombination (HR) between two tandem repeat sequences.
  • HR homologous recombination
  • An example, known as the "Tandem repeat pop-out" method is described in Akada, R. et al, Yeast, 23, 399-405 (2006).
  • An example of such an approach is described in Example 4B(ii), below, used to delete a region of the CDS139 locus in the M. genitalium donor genome. The process is illustrated in Figure 10B. This technique can be adapted for use in gene replacement.
  • methods using tandem-repeat induced HR can introduce and subsequently remove a cassette containing a counter-selectable marker via a single transformation event.
  • a cassette carrying the counter-selectable marker and the sequence that generates the tandem repeat can be introduced into a donor genome in a yeast host by transformation, followed by selection for spontaneous homologous recombination between homologous regions in the cassette and the genome.
  • Selection for the initial introduction of the marker can be carried out as described above, e.g., by growth in the absence of histidine in the case of URA3. Subsequent transfer of cells into counter-selection medium (e.g., 5-FOA) selects for spontaneous "pop-out" of the marker by homologous recombination between the tandem repeat sequences.
  • counter-selection medium e.g., 5-FOA
  • Such methods can be adapted for deletion, point mutation, and gene replacement, by varying the portions of the cassette sharing homology with the target nucleic acid.
  • tandem repeats and enzymatic cleavage near the target region can be used to facilitate removal of the selectable marker.
  • One such embodiment deemed the "Tandem Repeat Endonuclease Cleavage" (TREC) method, combines conventional homologous recombination replacement using a counter-selectable marker, inducible introduction of double-strand break near or at the target region by expression of an endonuclease, and introduction of tandem-repeat sequences flanking a nucleic acid sequence containing the marker.
  • the methods can be used to accurately modify bacterial donor genomes and large nucleic acids cloned in yeast host cells with high efficiency.
  • flanking tandem repeat sequences and proximal or adjacent double-strand break greatly enhances the efficiency of target-specific recombination and allows genetic engineering of bacterial genome in yeast hosts.
  • An example in which this method was used to successfully seamlessly delete the CD 139 locus in the M. genitalium genome grown in yeast is described in Example 4C, below. The method is illustrated in Figure IOC.
  • the methods described herein can be used to introduce any modification, such as point mutations (e.g., nucleic acid and codon substitution, including conservative and non- conservative substitutions), deletions, insertions, and other modifications to target regions of donor nucleic acids within host cells, such as bacterial genomes within yeast host cells.
  • point mutations e.g., nucleic acid and codon substitution, including conservative and non- conservative substitutions
  • deletions e.g., deletions, insertions, and other modifications to target regions of donor nucleic acids within host cells, such as bacterial genomes within yeast host cells.
  • the target cassette for introducing the selectable marker contains a portion of homology to a portion of the donor target region (which optionally contains one or more mutations, deletions, insertions, substitutions, or other modifications compared to the homologous portion) and a selectable marker, typically a counter-selectable marker, such as URA3.
  • the cassette contains a portion homologous to a 5' portion of the target region and a portion homologous to a 3' portion of the target region.
  • a second targeting construct is generated, to replace the selectable marker with nucleic acid having homology to the target nucleic acid.
  • This second targeting construct contains homology to the target region or portion thereof, and optionally contains one or more mutations, deletions, insertions, substitutions, or other modifications compared to the homologous portion within the target region.
  • the targeting cassette for introduction of a double-strand break near, within, or adjacent to the target region, the targeting cassette further includes a gene encoding an enzyme, such as an endonuclease, which cleaves nucleic acid such as dsDNA at a particular sequence, and further contains a nucleotide sequence recognized by the enzyme, typically at or near a terminus of the cassette.
  • an enzyme such as an endonuclease
  • the gene encoding the endonuclease is under the control of an inducible promoter, such as the GAL1 promoter, such that expression of the gene can be induced by growth of the host cells under particular environmental conditions, such as in the presence of galactose as the only carbon source.
  • the targeting cassette for generation of tandem-repeat sequences, includes a further portion of homology to the target nucleic acid, which is upstream or downstream of the target region, along the length of the target nucleic acid, such that upon integration of the cassette through homologous recombination, a tandem-repeat sequence will be present in the target nucleic acid.
  • the cassette contains a first portion of homology to a portion of the target nucleic acid that is upstream or downstream of the target region along the length of the target nucleic acid (to generate tandem repeats), a second and third portion of homology to 3' and 5' portions of the target region, respectively (for insertion of the cassette by homologous recombination), a nucleic acid encoding an enzyme (e.g., endonuclease) under the control of an inducible promoter, a nucleotide sequence recognized by the endonuclease, and the selectable marker, typically a counter- selectable marker.
  • an enzyme e.g., endonuclease
  • the second and third portions of homology flank a sequence containing the first portion of homology (which generates the tandem repeat).
  • the second and third portions of homology further flank a sequence containing the nucleic acid encoding the enzyme (e.g.,
  • endonuclease typically also flank a sequence containing the selectable marker.
  • the nucleotide sequence recognized by the enzyme is located adjacent to the second or third homologous portion and is on the opposite terminus of the construct relative to the first portion of homology (which generates the tandem repeat region).
  • one or both of the second or third portions of homology contains one or more nucleotide mutations, insertions, or deletions, compared to the homologous portion in the target nucleic acid.
  • FIG. 1 An exemplary targeting cassette, used in TREC in the Examples below, is illustrated in Figure IOC.
  • This exemplary construct contains an l-Scel recognition site, a nucleic acid encoding a l-Scel endonuclease under the control of a GAL-1 promoter, a URA3 counter-selectable marker, and a portion (labeled "Repeat") that is homologous to a portion of the target nucleic acid sequence upstream of the target region (labeled "Repeat" in the target nucleic acid, also pictured).
  • the cassette further contains a 50 bp portion of homology to the target region that is 5 Of the l-Scel site and a 50 bp portion of homology to the target region that is 3 Of the "Repeat” portion of homology.
  • the portion of the targeting cassette containing the recognition site, the endonuclease-encoding gene and inducible promoter, and the selectable marker is termed the "CORE" cassette.
  • This cassette was used, as described in Example 4C, below, to modify a M. genitalium donor genome, which had been transferred into a yeast host using the provided methods, within the host cell, by deleting a 450 base-pair portion of a target region of the genome.
  • a similar construct was used to delete the Type II Restriction Enzyme gene in a M. mycoides LC genome within a yeast host cell using the provided methods.
  • Variations of these targeting cassettes such as those described in Figs. 10A-D and described elsewhere herein, also can be used with the provided methods.
  • variations of the cassettes can be used to introduce mutations, substitutions, insertions, and other modifications such as modified nucleotides, into the target nucleic acid.
  • modifications of the provided cassettes and methods will be apparent to the skilled artisan.
  • the cassettes can be generated using any of a number of well-known nucleic acid synthesis, amplification, joining, and assembly methods, such as those described herein and commercially available methods.
  • the cassettes are by amplification and/or assembly of nucleic acid fragments making up portions of the cassettes.
  • the fragments can be generated using well-known methods, such as chemical synthesis or amplification (e.g., PCR) from a plasmid, genomic DNA or other nucleic acid containing the desired nucleotide sequence.
  • the fragments are assembled to form the cassette using fusion PCR, using a recombinant PCR technique, as described in Shevchuk, N.A. et al. , Nucleic Acids Res, 32, el 9 (2004).
  • chimeric fusion primers are used to amplify two different fragments to be joined, which then are joined in a primer-less polymerase reaction (e.g., primer-less PCR).
  • the chimeric primers each contain a portion of homology to the first fragment to be joined, and a portion of homology to the second fragment to be joined.
  • amplifying both fragments using the primers generates regions of overlapping homology among the products of the amplification, such as 40 bp homology at the termini of the products.
  • cassettes can be prepared using conventional synthetic methods as described, or can be purchased from commercial suppliers.
  • Other assembly methods can be used, such as a one-step isothermal DNA assembly method is described in Gibson et al. , Nature Methods 6, 343 - 345 (2009), and in U.S. Patent Application No. 12/371,543, filed February 19, 2009, by the concerted action of a 5' exonuclease, a DNA polymerase, and a DNA ligase.
  • DNA fragments are first recessed by the 5' exonuclease, yielding single-stranded overhangs, which then specifically anneal, followed by gap-filling and covalent joining using the polymerase and the ligase.
  • Other assembly methods are described in U.S. Patent
  • the cassettes are transformed into host cells containing the donor genomes, such as those produced according to the provided methods. Transformation methods are well known.
  • the cassettes are introduced into yeast host cells containing the donor genome using lithium acetate integrative transformation, according to a published method (Gietz, D. et al, Nucleic Acids Res, 20, 1425 (1992)), with 2-3 ⁇ g PCR product and 25 ⁇ g carrier DNA (Salmon testis DNA, Sigma, St. Louis, MO).
  • cells are grown in medium lacking uracil and individual URA + transformants are selected and optionally analyzed by PCR, using diagnostic primers that specifically bind to portions of the target donor nucleic acid flanking the region at which the cassette is inserted. Whether the cassette is correctly inserted is determined by evaluating the presence and size of amplicon using such primers, which produce different sized amplicons depending on whether the cassette is inserted. An example is described in Example 4C(ii) below. Cells containing the correct insertion are used in subsequent rounds of homologous recombination.
  • cells containing the counter-selectable marker When inducible expression of an enzyme to generate ds breaks is carried out, cells containing the counter-selectable marker then are grown under conditions that induce expression of the enzyme from the inducible promoter, such as growth in galactose- containing medium.
  • cells are grown on SG (synthetic galactose)-His medium, containing galactose as the only carbon source, for example, for 4 hours or 24 hours, to induce expression of the enzyme that introduces the ds break. Growth on glucose- containing medium can be carried out as a control.
  • a second nucleic acid containing a sequence homologous to the target nucleic acid that will replace the selectable marker is transformed into the cells.
  • this second nucleic acid contains one or more mutations, deletions, insertions, or substitutions compared to the target nucleic acid. See Examples 4 A and 4B.
  • the second homologous recombination event occurs spontaneously, through the tandem repeats generated in the target nucleic acid after insertion of the vector. With the TREC method, a combination of these methods is used for removal of the selectable marker.
  • Loss of the counter-selectable marker is selected for by growth under conditions that favor the loss.
  • URA3 when URA3 is used as the marker, before such a selection (e.g., after a second round of transformation), cells are grown in the presence of uracil, overnight at 30 °C, to deplete residual orotidine-5 '-phosphate decarboxylase (encoded by URA3 gene) in yeast cells having lost URA3 gene. Cells having lost the counter-selectable marker are then selected in an environment that favors the loss, such as in the presence of 5- FOA, such as on HIS plates containing 5-FOA, to select loss of the URA3 gene.
  • PCR analysis using the same or different diagnosis primers, flanking the site of insertion can be carried out to verify deletion of the cassette.
  • Multiplex PCR can be carried out to analyze the integrity of donor nucleic acids, such as genomes, modified using the provided modification methods.
  • MPCR Multiplex PCR
  • MPCR can be performed as described in D. G. Gibson et al, PNAS USA, 105:20404-9 (2008).
  • Isolation of total DNA from the host cells for PCR and MPCR analysis can be performed using the isolation methods described herein, depending on the type of host cell.
  • isolation of genomic DNA from yeast host cells is carried out as described in Example 3.
  • MPCR primer sets can be designed with homology at various portions along the length of the donor genome, such as around the circular bacterial genome in yeast, with varying sizes, such that presence of each amplicon can be verified. See, e.g., D. G. Gibson et al, PNAS USA, 105:20404-9 (2008)).
  • Multiplex PCR can be carried out using well-known methods, including commercially available kits, usch as Qiagen Multiplex PCR Kit. An exemplary reaction is described in Example 4A, below.
  • each amplicon indicates that the modified genome is complete and is typically carried out to assure that spontaneous unwanted recombination events have not occurred, generating unwanted modifications.
  • Other modification methods can be used in connection with the provided methods, depending upon donor, host, and recipient cell types.
  • CTQ-LOXP system can be used.
  • the Cre-/oxP system is a known efficient site-specific recombination method that has been successfully used to remove selection markers and large genomic DNA segment in a large number of different organisms.
  • a Cre-loxP mutagenesis construct with mutant loxP genes can be produced, e.g., by two rounds of PCR reactions, as described for other methods.
  • the modification method is as efficient, substantially as efficient, or more efficient than modification by the Cre-LoxP system.
  • donor nucleic acids including donor chromosomes and/or donor genomes into host cells or recipient cells.
  • the donor nucleic acids can be transplanted from host cells to recipient cells.
  • Donor nucleic acids include those modified within host cells.
  • the donor genomes are transplanted directly from donor cells into recipient cells, for example by transplantation of native genomes into recipient cells.
  • Transplantation methods are useful for efficiently transplanting donor genomes, which have been propagated and modified within host cells, back into an environment in which gene products can be expressed from the genomes.
  • the recipient cells can be cells of the same species or a closely related species compared to donor cell or organism.
  • transplantation of prokaryotic donor genomes, propagated in eukaryotic hosts, into prokaryotic recipients can be limited by nucleic acid recovery, methylation, incompatibility and toxicity issues. Methods are needed in which a sufficient amount of purified, intact, donor nucleic acid is recovered from the host cells to generate a sufficient number of recipient cells containing transplanted donor nucleic acids, such as a detectable number.
  • bacterial genomes isolated after growth in yeast hosts can be susceptible to restriction-modification system(s) of bacterial recipient cells (Holt et al. , Bioessays 29, 580 (2007).
  • transplanting bacterial genomes that have been modified and propagated in yeast cells into cells in which donor gene products can be expressed (such as donor cells and other bacterial recipient cells) carries the risk that the transplanted genomes will be incompatible with the recipient cells.
  • yeast hosts which do not contain restriction-modification systems can nonetheless express DNA methyltransferases that can modify donor nucleic acids (such as bacterial genomes) inhibiting their activation (e.g., gene product expression) upon transplantation into a recipient cell such as a bacterium.
  • donor nucleic acids such as bacterial genomes
  • activation e.g., gene product expression
  • the structure and confirmation of donor genomes isolated after propagation and modification in host cells can differ from the confirmation and structure of the same genome propagated in a cell more closely related to the donor organism. Such differences can negatively impact transplantation of the donor nucleic acids back into recipient cells.
  • the transplantation methods described herein include aspects to overcome such limitations for successful transplantation of donor genomes, modified and/or propagated in host cells, into genetically distinct recipient cells, such as from eukaryotic hosts to prokaryotic recipients. Among these aspects are in vitro methylation, treatment with enzymes to degrade host cell protein, and transplantation into recipient cells lacking restriction- modification systems, such as by mutation of these systems in recipient cells. Exemplary studies, demonstrating success of the provided transplantation methods, are described in detail in Example 3 and Example 5, below.
  • FIG. 8 schematically illustrates three aspects of the provided transplantation methods.
  • donor DNA is isolated in agarose plugs which are melted, such as with ⁇ -agarase treatment, and transplanted directly into recipient cells.
  • This first approach is typically used when nucleic acids are transplanted between similar cells and incompatibility issues are not a concern.
  • recipient cells are modified to mutate restriction enzymes prior to
  • donor nucleic acid in agarose plugs is subjected to methylation and deproteinisation reactions, prior to melting and transplantation, in order to protect the donor nucleic acid from recipient R-M systems and conformational changes.
  • methylation is performed without deproteinisation.
  • Figure 16 schematically illustrates additional aspects of the provided
  • a bacterial genome can be moved into yeast, engineered, and installed back into a bacterium by genome transplantation.
  • a yeast vector can be inserted into a bacterial genome by transformation; that bacterial genome is cloned into yeast. After cloning, the repertoire of yeast genetic methods is used to create one or more insertions, deletions, rearrangements, or any combination thereof in the bacterial genome.
  • This engineered genome can then be isolated and transplanted into a recipient cell to generate an engineered bacterium. Before transplantation, in some cases, it may be necessary to methylate the donor bacterial DNA in order to protect it from a recipient cell's restriction system(s). This cycle can be repeated in an iterative fashion starting from the newly engineered genome (dashed arrow). i. Isolation of donor nucleic acids from host cells or donor cells
  • donor nucleic acids ⁇ e.g., donor genomes
  • Methods for isolation of nucleic acids from cells are well known, including methods for isolation of genomic DNA, including whole genomes, and methods for isolating organelle genomes. Any such method, including those described herein, can be used to isolate the donor nucleic acids.
  • One will understand that the choice of method depends on the type of nucleic acid to be isolated and the type of cell from which it is isolated.
  • isolation of large nucleic acids is carried out in agarose plugs, as described below in the Examples.
  • cells containing the donor nucleic acids are grown in the presence of Chloramphenicol or similar substance prior to isolation of the donor nucleic acids. Chloramphenicol is used to obtain compact and fully replicated donor genomes and chromosomes in the isolated nucleic acid samples (such as agarose plugs).
  • chloramphenicol in Mycoplasma cultures might help to get compact and fully replicated genomes in the agarose plugs. a. Isolation from host cells
  • the donor nucleic acids can be isolated in agarose plugs using a protocol compatible with the host cells.
  • agarose plugs can be prepared, for example, with the CHEF mammalian Genomic DNA Plug Kit (Bio-Rad), following the instructions recommended by the manufacturer for yeast DNA extraction, with optional modifications.
  • cultures of yeast host cells containing bacterial donor nucleic acids are grown at 30° C in selective medium until the OD 60 o reaches approximately 1.5.
  • 6x10 9 yeast cells are used per mL of plugs (instead of 6x10 8 cells recommended by the
  • Cell walls of the yeast hosts embedded in the agarose plugs are digested. Cell wall digestion can be carried out using lyticase (Biorad), as recommended by the CHEF kit manufacturer, or using 100T ( ⁇ -l, 3-glucan laminaripentaohydrolase; USB, Cleveland, OH).
  • lyticase Biorad
  • 100T ⁇ -l, 3-glucan laminaripentaohydrolase; USB, Cleveland, OH
  • ZymolaseTM enzyme is added inside and outside of the plugs at a concentration of 5 mg/mL. The mixture is allowed to stand for 2 hours at 37°C.
  • phenylmethanesulfonylfluoride (PMSF) is added during the second wash.
  • the isolation protocol is modified in order to isolate organelle genomes, as in the organelle genome isolation methods discussed herein. b. Removal of host nucleic acids
  • yeast genomic DNA is also isolated along with the bacterial nucleic acid that is extracted from the host cell.
  • isolation includes a "clean-up" step, in which contaminant host nucleic acids are removed, such as with restriction enzymes that recognize host nucleic acids but not donor nucleic acids.
  • plugs are treated with a cocktail of restriction enzymes that specifically digests yeast genomic DNA.
  • removal of endogenous host DNA plugs is carried out by incubation of the plugs overnight at 37°C with restriction enzymes (e.g., 50 units of AsiSl, Rsrll, and Fsel enzymes (New England Biolabs, Ipswich, MA) in a 500 ⁇ , reaction volume) that specifically cut host genomic DNA but leave donor DNA intact. Plugs then are washed at room temperature for 1 hour with 1 ml of IX TE buffer and loaded on 1% TAE agarose gel (120 minutes, 120 volts), to remove digested host DNA fragments from the plugs.
  • restriction enzymes e.g., 50 units of AsiSl, Rsrll, and Fsel enzymes (New England Biolabs, Ipswich, MA) in a 500 ⁇ , reaction volume
  • Plugs then are washed at room temperature for 1 hour with 1 ml of IX TE buffer and loaded on 1% TAE agarose gel (120 minutes, 120 volts), to remove digested
  • yeast plugs are subjected to electrophoresis in a 1 % LMP gel in 1 X TAE buffer, with a contour-clamped homogenous electric field (Chu et al, Science 234, 1582 (1986)), using the CHEF DR IIII, from Bio-Rad. Typically, pulse times are ramped from 60 to 120 seconds for 24 hours at 3.5 V/cm. After electrophoresis, plugs are removed from the wells and stored in 1 X TE buffer at 4°C.
  • agarose plugs can be removed from wells for further processing.
  • the removed plugs are washed two times for one hour in 1 mL 0.1X TE buffer and equilibrated for one hour in 1 mL of IX NEB buffer 2 (New England Biolabs, Ipswich, MA) supplemented with BSA (100 ⁇ g/mL).
  • IX NEB buffer 2 New England Biolabs, Ipswich, MA
  • BSA 100 ⁇ g/mL
  • plugs can be incubated with a restriction enzyme.
  • plugs are incubated overnight at 37°C with 50 units of PspXl restriction enzyme. Following the incubation, plugs are washed for 1 hour at room temperature with 1 mL of IX TE buffer and loaded onto a pulse-field gel.
  • host DNA is not removed prior to transformation. c. Isolation from donor cells
  • donor nucleic acids e.g., donor genomes
  • isolation methods are used that are compatible with the donor cells.
  • agarose plugs containing genomic DNA are prepared using the CHEF mammalian Genomic DNA Plug Kit (Bio-Rad), with modifications.
  • cells e.g., M. mycoides LC cells containing donor genomes or yeast cells containing the donor genomes
  • cells are grown in the appropriate medium until a desired OD and then incubated with 100 ⁇ g/ ⁇ l chloramphenicol for 90 minutes before harvesting.
  • Mycoplasma donor cells is performed as described by Lartigue et al , Science 317, 632 (2007) with optional modifications, such as modifications to cell culture prior to isolation. One such example is described in Example 2B(ii). d. Quantification of isolated donor nucleic acid
  • the amount of isolated donor nucleic acid can be quantified or estimated prior to transplantation.
  • donor nucleic acids isolated from host cells are run on agarose gel and compared to donor nucleic acids isolated from known quantities of donor cells.
  • An example is described in Example 2B.
  • the amount of isolated donor nucleic acid is quantified, such as by UV spectrophotometry.
  • One such example is described in Example 2B(iv). ii. Treatment of the isolated donor genomes and/or recipient cells
  • the provided methods include steps for overcoming incompatibility barriers between host, donor, and recipient cells/nucleic acids.
  • barriers are described herein, and can limit the transplantation of large nucleic acids such as donor genomes from host cells into recipient cells in which donor gene products can be expressed. This is particularly the case when the host cells are not closely related (e.g., from a different branch of life) to the donor and recipient organisms.
  • Such barriers are relevant for transplantation of prokaryotic genomes, propagated in eukaryotic hosts, into prokaryotic recipients.
  • the barriers can be caused by a number of factors including incompatibility and toxicity.
  • restriction-modification (R-M) systems present in recipient cells (and perhaps also in donor cells), but not present in host cells, can cause incompatibility upon transplantation of donor nucleic acids that have been propagated within the host cells.
  • Restriction-modification systems are well known and are used, typically by bacterial organisms, to protect the organism from foreign DNA. Restriction modification systems generally include proteins for recognizing and cleaving particular nucleic acid sequences in foreign DNA, and enzymes for modifying (e.g., methylating), and thereby protecting, those sequences in the organism's own nucleic acids. Restriction-modification systems include Type I, Type II, and Type III systems. Type I systems generally contain a complex of three proteins that individually recognize (specificity), cleave (restriction) and modify
  • Type II systems generally contain two separate modification and restriction enzymes, which methylate and cut DNA sequences, respectively.
  • Type III systems contain restriction and modification enzymes that form heterodimer complexes for modification and cleavage. The modification enzymes also can methylate their own DNA.
  • DNA methyltransferases by host cells (including those that do not contain restriction-modification systems) can modify donor nucleic acids and inhibit their activation (e.g., gene product expression) after transplantation into recipient cells.
  • Structural and conformational changes in donor nucleic acids following propagation and modification in host cells can negatively impact transplantation of the donor nucleic acids back into recipient cells.
  • the provided transplantation methods include steps for overcoming such limitations in order to successfully transplant donor genomes, modified and/or propagated in host cells, into genetically distinct recipient cells, such as from eukaryotic hosts to prokaryotic recipients.
  • the steps include (1) treatment of the isolated donor nucleic acids and (2) modifications to the recipient cells.
  • In vitro assays can be utilized to determine whether incompatibility issues exist between host cells, donor nucleic acids, and recipient cells, for example, due to inconsistency in restriction-modification systems among the various organisms. Restriction-modification systems expressed by the donor genome or by the recipient cell may have the potential to impair successful transplantation and activation of the donor genome in the host cell.
  • restriction-modification systems To examine the issue of a possible restriction-modification problem for donor genomes propagated in host cells of a different type of organism, such as bacterial genomes propagated in yeast, the donor, host, and/or recipient cells are assessed with respect to restriction-modification systems.
  • the presence of restriction-modification systems in donors and/or recipients (and recognition site specificities of the systems) can be identified from donor genome sequences, using known methods. See also REBASE, The Restriction Enzyme Database, available at the World Wide Web address: rebase.neb.com/rebase/rebase.html.
  • the presence of a modifying enzyme can be tested in vitro.
  • the methylation status of predicted recognition sites can be probed using commercially available restriction enzymes that recognize the predicted sites.
  • commercially available restriction enzyme isoschizomers corresponding to the predicted restriction enzyme systems can be used in digestion reactions to determine whether donor and recipient genomes are methylated at appropriate restriction sites.
  • Genomic DNA that is methylated at the predicted sites can be protected from cleavage by the commercially available enzymes that recognize the sites. If recipient genomic DNA is protected from cleavage, the presence of the modifying enzyme of the R-M system is confirmed.
  • This process also can be carried out on genomic DNA isolated from donor cells in order to assess whether the donor genome is likely to be protected from the system. An example is described in Example 2D, below.
  • cell-free extracts prepared from the recipient and donor cells can be used to determine whether predicted restriction enzymes are present and active in the cells. Methods for making cell-free extracts are well known and any can be used with the provided methods, depending upon the cell type.
  • Mycoplasma cell-free extracts are prepared as described in Example 2D(ii)(b), below. DNA containing the predicted restriction sites is incubated with the cell-free extract in a restriction digest to determine the presence in the extracts of enzymes that recognize and cut DNA at the sequence. Digested samples are run on an agarose gel to determine cleavage. If desired, DNA that is or is not methylated at particular sites can be compared in the assay.
  • Cell-free extracts also can be used as a source of methyltransferase activity, by the addition of EDTA, such as 10 mM EDTA, to inhibit nucleases.
  • EDTA such as 10 mM EDTA
  • recombinant methyltransferases such as E. coli dam methyltransferase (New England Biolabs, Ipswich, MA) can be used to methylate DNA prior to digest assays.
  • Methyltransferases can also be purified An example of such a digest assay is described in Example 2D(ii)(b), below.
  • Donor nucleic acids can be methylated in vitro following isolation from the donor cells and prior to transplantation into recipient cells. Methylation of the donor genomes, such as those that have been propagated in host cells, can protect them from restriction
  • capricolum recipient cells implying that the donor genome becomes methylated before a lethal level of restriction enzyme activities is reached. This is not surprising because most endonuclease and methyltransferase gene pairs can be cloned simultaneously in E. coli. See Holt et al, Bioessays 29, 580 (2007). One would understand that donor cells, host cells and recipient cells can be assessed with respect to their restriction-modification systems to assess whether a donor genome is to be protected prior to transplantation.
  • methylation is performed using methyltransferases that are the same as, or similar to, those of the R-M system from which protection is desired.
  • Methylation can be carried out using methylases isolated from cell extracts (e.g., recipient cell extracts) or recombinantly produced and purified methylases.
  • methylases of the R-M systems of the recipient and/or donor cells are exogenously expressed and purified using recombinant methods.
  • the coding sequences for all methyltransferases identified by R-M prediction can be codon optimized for expression in a desired system, such as yeast or bacterial cells.
  • Fragments containing the coding sequences can be constructed using any of a number of well-known synthesis and/or assembly methods, such as those described herein.
  • Example 2E(i)(a) in which methyltransferase-encoding nucleic acids were generated using a one-step isothermal DNA assembly method. After construction, the fragments are cloned into expression vectors, for expression in a cell of choice.
  • Vectors can be used to transform cells for expression of gene products.
  • Exemplary expression systems include the BL21 (DE3) codon plus cells (Stratagene, La Jolla, CA). Expression of the methyltransferases can be induced in the cells, such as by incubation with IPTG. Methyltransferases can be purified from the cells, using well-known methods. In one example, cell lysates are clarified, column-purified, and fractions containing the
  • methyltransferases dialyzed against an enzyme buffer for use in subsequent methylation reaction, such as 50 mM HEPES-NaOH pH 7.2, 50 mM NaCl, 0.1 mM EDTA, 10 % glycerol). The samples can then be concentrated if needed. Exemplary expression and purification protocols are described in detail in Example 2E(i)(b), below.
  • crude extracts from cells having the R-M systems are used to methylate the donor nucleic acids isolated from the host cells.
  • This aspect can be advantageous if it is not certain that all the R-M systems of the recipient or host cell have been determined. For example, if the R-M systems of a recipient cell are unknown, methylation using a crude extract from the recipient cell will ensure that all the relevant methyltransferases are present in the methylation reaction. Any well-known method for preparing the appropriate cell extract can be used. An example is described in Example 2E(ii), below, for preparation of crude cell extracts containing methyltransferases from Mycoplasma recipient cells. Nucleases in the cell extracts can be inhibited, such as by the addition of 10 mM EDTA, to allow their use in methylation reactions.
  • Purified methyltransferases or crude cell extracts containing methyltransferases can be used to methylate donor nucleic acids isolated from host cells.
  • agarose plugs containing the donor nucleic acid can be washed and equilibrated in
  • methylation buffer ⁇ e.g., 100 mM Tris-HCL pH 7.5; 10 mM EDTA; 3 ⁇ DTT, 200 ⁇ S- adenosylmethionine (SAM)). Plugs then can be incubated in methylation reactions, including methylation buffer and either crude cell extracts or purified methyltransferases. Parallel reactions without SAM can be used as controls. In one aspect, methylation reactions can be carried out in the presence of dam methyltransferase (New England Biolabs, Ipswich, MA).
  • dam methyltransferase New England Biolabs, Ipswich, MA.
  • each yeast plug can be inclubated for 4 hours at 50 °C in 1 ml of Proteinase K Reaction Buffer supplemented with 40 ⁇ of Proteinase K. The plugs can then be washed 4 times for 45 minutes each with 1 ml of IX TE buffer and 2 times for 30 minutes each on 0. IX TE buffer with gentle agitation at room temperature. After removing the final wash buffer, the plugs can be melted.
  • An example is described in Example 3 A(iii).
  • the effectiveness of methylation reactions on protecting donor nucleic acids from recipient R-M systems can be tested in vitro prior to the transplantation studies. Adjustments can be made depending upon the results of the R-M system assessments.
  • This process can be carried out by performing methylation reactions on donor genomic DNA or plasmids containing donor nucleic acids.
  • the methylation reaction can be followed by a restriction digest reaction, using restriction enzymes of the recipient cell, to ensure protection of the donor nucleic acids by methylation.
  • An example is described in Example 2E. c. Deproteinisation
  • donor nucleic acids can be subjected to a deproteination step after methylation and prior to transplantation to remove the proteins in the crude extract.
  • the proteins can be removed using a proteinase, such as proteinase K.
  • agarose plugs that have been subject to methylation reactions can be further incubated for 4 hours at 50 °C in proteinase K reaction buffer and proteinase . The plugs can be washed before proceeding with melting of the plugs and transplantation. See Examples 2B(iv) and 3 A(iii). d. Genetic modification of recipient cell R-M systems
  • the restriction-modification system of a recipient cell may need to be incactivated prior to transplantation of a donor nucleic acid or genome; modification of a R-M system can occur in vitro or in vivo. Instead of in vitro methylation, restriction modification systems can be removed or inactivated from at least the recipient cell, and possibly the donor genome or nucleic acid.
  • restriction enzyme(s) of the recipient cell R-M system can be inactivated by mutation of the gene encoding the enzyme. This process typically can be used as an alternative to in vitro methylation of the donor nucleic acids prior to transplantation.
  • Restriction modification system inactivation of donor genomes may not be practical in some cases. For example, expression of restriction endonucleases encoded by the donor genome immediately following transplantation can help drive transplantation by degrading the resident genome of the recipient cell. Thus, removal of the donor restriction modification systems is undesirable in such cases and methylation should be used.
  • each of the donor, host and receipient cells can be assessed in this regard prior to transplantation to identify the best system for inactivation of a R-M system.
  • Any mutation process can be used to inactivate a R-M system.
  • Example 2F In which the gene encoding the single restriction enzyme in a Mycoplasma recipient cell was inactivated prior to transplantation of donor nucleic acids.
  • the gene was mutated by interruption with a puromycin resistance marker, allowing selection of cells containing the mutated gene.
  • Other methods of inactivation are contemplated herein and include a variety of resistance markers that can also be used for selection of cells containing a mutated gene; such markers are described herein and are also known in the art. Cell extracts prepared from such mutant recipient cells can be used as controls in methylation reactions as described herein.
  • a donor genome can be methylated in vivo, for example, while still in the host cell.
  • In vivo methylation inside a host cell can be carried out by expression of donor or recipient methylases that are cloned into the host vector. This aspect may be less desirable as it may lead to unwanted changes in the donor genome.
  • expression of bacterial methylases in yeast has been shown to increase yeast homologous recombination, which could lead to alteration of a donor bacterial genome housed in either a yeast artificial chromosome (YAC) or yeast centromeric plasmid (YCp).
  • the donor nucleic acids can be further transplanted into recipient cells using methods described herein or known in the art.
  • One exemplary transplantation protocol is described in Example 3, below.
  • One method used to transplant Mycoplasma genomes from donors to Mycoplasma recipients is described by Lartigue et al, Science 317, 632 (2007).
  • Such methods can be used to modify genomes or nucleic acids from intractable cells or organisms in a separate host to confer a specific property upon the donor genome.
  • the modified genome can then be transplanted back into the same donor cell or into a donor cell, thereby conferring the phenotype of the modified genome upon the recipient cell.
  • Recipient cells typically are chosen based on their ability to support gene expression from the donor nucleic acids, such as the donor genomes.
  • the transfer of a bacterial genome into a eukaryotic host is provided herein as an exemplary method and is not intended to be limiting.
  • a eukaryotic host cell having a preferred genetic manipulation system e.g., yeast
  • differences in translation and transcription and different codon usage, among other factors can prevent expression of the donor gene products within the host cell.
  • the recipient cell may be of the same species or a similar species as a donor cell or organism. It is often of the same order or kingdom as the donor. One will be able to determine an appropriate recipient cell based on the donor genome or other nucleic acid from which expression is desired.
  • host DNA can optionally be removed (e.g., by digest and/or electrophoresis), and optionally treated with methyltransferases and/or proteinase.
  • Agarose plugs can be melted, for example, by incubation with ⁇ -Agarase I (New England Biolabs) as described in Example 3 A(ii)(b) below.
  • Transplantation can be performed in the presence of polyethylene glycol (PEG), such as PEG-6000 or PEG-8000 or other PEG to facilitate transformation.
  • PEG polyethylene glycol
  • the source, amount, and size of the PEG can be varied to determine the optimal PEG.
  • the PEG is PEG-2000, PEG-4000, PEG-6000, PEG-8000, PEG- 10000, PEG-20000, or other.
  • the concentration of PEG can be varied depending upon the conditions of the transplantation; concentrations include those, for example, at or about 5 % or at or about 10 %. An example is described in Example 3 A(ii)(c), below.
  • Melted plugs can be added to the recipient cells in the presence of PEG with gentle rocking to mix. Cells are allowed to recover, centrifuged, and grown in medium containing appropriate selection medium to select for recipient cells containing the transplanted donor nucleic acid. In one aspect, cells are plated on the medium and grown under appropriate conditions for the recipient cell type until colonies appear
  • Colonies can be picked and further grown in selection medium to produce a desired quantity of recipient cells containing the transplanted genome or other donor nucleic acid.
  • a particular ratio of recipient cells to donor nucleic acid can be maintained as needed. In one example, a ratio of between at or about 10 and at or about 10 recipient cells per 2 ⁇ g genomic DNA can be maintained.
  • the provided transplantation methods can be used to achieve approximately 30 transformants for 200 ng of endogenous genomic DNA, or between 500 and 1500 transplants per reaction, or other appropriate amount that is obtained from the host or donor cell. In one non-limiting example, transplantation is carried out with ⁇ 10 recipient cells, 20 ⁇ of melted of agarose plug containing donor genome at 100 ng/ ⁇ .
  • the ratio of recipient cells to donor nucleic acid may vary depending upon the cell types and that empirical assessment can be used to optimize the ratio.
  • FIG. 8 An exemplary transplantation method is illustrated in Figure 8 ("3"): in this method, genomic DNA containing appropriate markers and elements for propagation in host and recipient cells is isolated from host cells in agarose plugs, methylated with crude extract or purified methyltransferases and deproteinized with Proteinase K. The agarose plugs are melted, DNA incubated with recipient cells, which then are plated on selection medium.
  • whole intact M. mycoides LC donor genomic DNA containing a YCp element, a tetracycline marker and a ⁇ -galactosidase gene can be isolated from yeast hosts in agarose plugs, methylated with a M.
  • an engineered cell produced by the methods can be used as a source of donor nucleic acid in subsequent rounds of transformation, modification, and transplantation, thereby generating a further modified genome and organism.
  • an engineered bacterial cell produced by the methods can be used as a source of donor nucleic acid in subsequent rounds of transformation, modification, and transplantation, thereby generating a further modified genome and organism.
  • a host vector is inserted into a donor genome by transformation. That genome is cloned into a host cell; after cloning, the repertoire of host genetic methods is used to create insertions, deletions, rearrangements, or any combination of modifications in the donor genome.
  • This engineered genome is then isolated and transplanted into a recipient cell to generate an engineered recipient cell with an altered phenotype. Before transplantation it may be necessary to methylate the donor DNA in order to protect it from the recipient cell's restriction system(s). This cycle can be repeated starting from the newly engineered genome.
  • FIG. 16 Provided in Figure 16 is a non-limiting example of moving a bacterial genome into yeast, engineering it, and installing it back into a bacterium by genome transplantation, a yeast vector is inserted into a bacterial genome by transformation. That genome is cloned into yeast; after cloning, the repertoire of yeast genetic methods is used to create insertions, deletions, rearrangements, or any combination of modifications in the bacterial genome.
  • This engineered genome is then isolated and transplanted into a recipient cell to generate an engineered bacterium. Before transplantation it may be necessary to methylate the donor DNA in order to protect it from the recipient cell's restriction system(s). This cycle can be repeated starting from the newly engineered genome (dashed arrow).
  • yeast mutants The large number of available, verified, substantiated, and reliable selectable markers for selection and counter-selection of yeast mutants enables use of the provided methods to carry out multiple, e.g., infinite iterative rounds of seamless nucleic acid alterations within yeast host cells.
  • Serial modifications to a cloned copy of an otherwise intractable genome or other large nucleic acid can be performed in yeast in rapid succession.
  • the mating capacity of yeast is favorable for modifying genomes and other large nucleic acids.
  • Yeast recombination machinery when activated during yeast mating, can be used to generate libraries, e.g., combinatorial libraries containing variants of cloned genomes or nucleic acids.
  • the provided methods and compositions can be used to solve problems related to the environment, energy production and medicine.
  • the provided methods and compositions are useful in producing, engineering and modifying genomes and organisms and other products for commercial use, such as immunogens, biological proteins and chemicals, vaccines, biofuels, and useful proteins such as enzymes.
  • the provided methods can be used to manipulate and engineer nucleic acids from any organisms, particularly those having poor genetic systems, such as those whose genomes are not easily manipulated by conventional methods.
  • the provided methods are useful in building synthetic genomes and transplanting the genomes into recipient cells to generate synthetic cells.
  • the methods can be used to produce medically useful proteins, including enzymes, protein and nucleic acid therapeutics, antibodies, immunogens, vaccines, and other cellular products.
  • a vaccine generally refers to an immunogenic preparation that improves immunity to a particular microorganism (bacterial or viral) associated with a disease.
  • a vaccine typically contains a small amount of an agent that resembles a microorganism. The agent stimulates the body's immune system to recognize the agent as foreign, destroy it, and remember it, so that the immune system can more easily recognize and destroy any of these microorganisms that it later encounters.
  • Vaccines can be prophylactic (e.g., to prevent or ameliorate the effects of a future infection by any natural or "wild" pathogen), or therapeutic (e.g., cancer vaccines).
  • Vaccines may be monovalent (also called univalent) or multivalent (also called polyvalent).
  • a monovalent vaccine can be designed to immunize against a single antigen or single microorganism.
  • a multivalent or polyvalent vaccine can be designed to immunize against two or more strains of the same microorganism, or against two or more microorganisms.
  • a monovalent vaccine may be used for rapidly developing a strong immune response.
  • Vaccines are used to try to reduce risk of illness, while retaining the ability to induce a beneficial immune response.
  • Vaccines can contain dead or inactivated organisms or purified products derived from them. Immunogenic compositions are useful for treating human and non-human populations (e.g., primates, veterinary animals, etc.).
  • immunogenic compositions such as those including live cells and viruses, including, but not limited to, modified Adenoviridae (e.g., adenovirus), Picornaviridae (e.g., coxsackievirus, hepatitis A virus, poliovirus), Herpesviridae (e.g., various types of Herpes simplex virus, Epstein-barr virus, Human cytomegalovirus), Hepadnaviridae (e.g., Hepatitis B virus), Flaviviridae (Hepatitis C virus, yellow fever virus, dengue virus, West Nile virus, etc.), Retroviridae (e.g., Human immunodeficiency virus (HIV)), Orthomyxoviridae (e.g., influenza virus), Paramyxoviridae (e.g., Measles virus, Mumps virus, Parainiflu
  • Parvovirus B19 Parvovirus B19), influenza (e.g., H1N1 influenza, Haemophilus influenzae type B, etc.), polio, vaccinia, varicella zoster, reovirus, retroviruses, poxviruses, Parvoviruses,
  • influenza e.g., H1N1 influenza, Haemophilus influenzae type B, etc.
  • polio vaccinia
  • varicella zoster reovirus
  • retroviruses retroviruses
  • poxviruses Parvoviruses
  • Picornaviruses Picornaviruses, paramyxoviruses, and BCG.
  • the provided technology is useful for production of immunological compositions to elicit an immune response from an organism, such as immunogenic compositions, such as those including live cells and bacteria, including, but not limited to, modified Bordetella (e.g., Bordetella pertussis), Borrelia (e.g., Borrelia burgdorferi), Brucella (e.g., Brucella abortus, Brucella canis, Brucella melitensis, and Brucella suis), Campylobacter (e.g., Campylobacter jejuni), Chlamydia (e.g., Chlamydia pneumonia, Chlamydia psittaci, and Chlamydia trachomatis), Clostridium (e.g., Clostridium botulinum, Clostridium difficile, Clostridium perfringens, and Clostridium tetani), Corynebacterium (e.g., Corynebacterium (e
  • Haemophilus influenza Haemophilus influenza
  • Helicobacter e.g., Helicobacter pylori
  • Legionella e.g., Legionella pneumophila
  • Leptospira e.g., Leptospira interrogans
  • Listeria e.g., Listeria
  • Mycobacterium e.g., Mycobacterium leprae and Mycobacterium
  • Mycoplasma e.g., Mycoplasma pneumonia
  • Neisseria e.g., Neisseria gonorrhoeae and Neisseria meningitides
  • Pseudomonas e.g., Pseudomonas aeruginosa
  • Rickettsia e.g., Rickettsia rickettsii
  • Salmonella e.g., Salmonella typhi and Salmonella typhimurium
  • Shigella e.g., Shigella sonne
  • Staphylococcus e.g., Staphylococcus aureus, Streptococcus pneumonia, Staphylococcus epidermidis and Staphylococcus saprophyticus
  • Streptococcus e.g., Streptococcus agalactiae, Streptococcus pneumoniae and Streptococcus pyogenes
  • Yersinia e.g., Yersinia pestis
  • Yersinia pestis e.g., Yersinia pestis
  • compositions effective to treat, prevent, or substantially reduce the biological impact of: chicken pox, shinges, influenza, polio, measles, mumps, rubella, toxic shock, cholera, bubonic plague, Hepatitis A, Hepatitis B, Hepatitis C, yellow fever, malaria, tuburculosis, tetanus, encephalitis, Acquired Immune Deficiency Syndrome (AIDS), leprocy, canine distemper, canine parvovirus, infectious canine hepatitis, adenovirus-2, leptospirosis, bordatella, canine parainfluenza virus, Dengue fever, Lyme disease and another other disease for which a vaccine is useful in treating and/or managing one or more symptoms.
  • AIDS Acquired Immune Deficiency Syndrome
  • leprocy canine distemper
  • canine parvovirus infectious canine hepatitis, adenovirus-2, leptospirosis, bordatella, canine para
  • NGU Nongonococcal urethritis
  • Urinary tract infections UMI
  • Diarrhea Diarrhea and Meningitis in infants (generally)
  • ETEC Escherichia coli
  • Meningococcal disease including meningitis and Waterhouse-
  • Pseudomonas aeruginosa Localized or systemic Pseudomonas infections.
  • Salmonella typhi Typhoid fever type salmonellosis (dysentery, colitis)
  • Toxinoses e.g., Toxic shock syndrome and Staphylococcal food poisoning
  • Streptococcal pharyngitis Streptococcal pharyngitis, Scarlet fever, Rheumatic fever,
  • Yersinia pestis Plague such as Bubonic plague and Pneumonic plague
  • the methods described herein can also be used to produce compositions effectiveo treat or prevent the disease contagious bovine pleuro pneumonia (CBPP), which is caused by the bacterium Mycoplasma mycoides Small Colony.
  • CBPP contagious bovine pleuro pneumonia
  • This disease also known as lung plague, is a major pathogen of cattle, yaks, buffalo, and zebu.
  • the disease is widespread in Africa, the Middle East, Southern Europe, as well as parts of Asia. There is a real need for an improved vaccine.
  • the disease organism is a close phylogenetic relative of the bacterium used here to demonstrate aspects of the provided methods, M. mycoides Large Colony strain GM12.
  • Antigen genes and/or the genome of M. mycoides Small Colony bacterium can be cloned and manipulated using the provided technology, to generate cells, e.g., mutants, to function as live vaccines.
  • the provided methods can be used, for example, with M. mycoides LC and closely related species as model systems for exploring the pathogenicity and biology of
  • Mycoplasmas The mycoides group of Mycoplasmas causes major diseases of ruminants and there is an urgent need for vaccines.
  • the provided methods can accelerate the construction of live vaccine strains. The methods also can be used to determine the minimal gene
  • compositions can be formulated and administered with any pharmaceutically acceptable carrier or excipient.
  • compositions comprising an immunogenic vaccine formulated in a medicament for the treatment of a disease or condition identified herein. Effect of an immunogenic vaccine composition following administration to a subject can be measured with respect to one or more aspects of an immune response.
  • Immune responses include, for example, induction of antibody responses (increased antibody titers), cytokine responses, induction of T helper (T H I and T H 2) cell differentiation and proliferation, etc. Each of such responses can be quantified. Immune responses also include reduction of overall bacterial or viral load in a patient.
  • Biocrudes are biologically produced compounds or a mix of different biologically produced compounds that are used as a feedstock for refineries in replacement of, or in complement to, crude oil or other forms of petroleum.
  • these feedstocks have been pre-processed through biological, chemical, mechanical or thermal processes in order to be in a liquid state that is adequate for introduction in a petroleum refinery.
  • Microorganisms can be modified using the methods described herein to produce a biocrude, which can be further processed to a biofuel composition.
  • the biofuel can then perform as a finished fuel or a fuel additive.
  • Finished fuel refers to as a chemical compound or a mix of chemical compounds (produced through chemical, thermochemical or biological routes) that is in an adequate chemical and physical state to be used directly as a neat fuel or fuel additive in an engine. In many cases, but not always, the suitability of a finished fuel for use in an engine application is determined by a specification which describes the necessary physical and chemical properties that need to be met.
  • engines are: internal combustion engine, gas turbine, steam turbine, external combustion engine, and steam boiler.
  • finished fuels include: diesel fuel to be used in a compression-ignited (diesel) internal combustion engine, jet fuel to be used in an aviation turbine, fuel oil to be used in a boiler to generate steam or in an external combustion engine, ethanol to be used in a flex-fuel engine.
  • diesel fuel to be used in a compression-ignited (diesel) internal combustion engine jet fuel to be used in an aviation turbine
  • fuel oil to be used in a boiler to generate steam or in an external combustion engine fuel oil to be used in a boiler to generate steam or in an external combustion engine
  • ethanol to be used in a flex-fuel engine.
  • ASTM standards mainly used ion the US
  • EN standards mainly used in Europe.
  • Fuel additive refers to a compound or composition that is used in combination with another fuel for a variety of reasons, which include but are not limited to complying with mandates on the use of biofuels, reducing the consumption of fossil fuel-derived products or enhancing the performance of a fuel or engine.
  • fuel additives can be used to alter the freezing/gelling point, cloud point, lubricity, viscosity, oxidative stability, ignition quality, octane level, and flash point.
  • Additives can further function as antioxidants, demulsifiers, oxygenates, thermal stability improvers, cetane improvers, stabilizers, cold flow improvers, combustion improvers, anti-foams, anti-haze additives, icing inhibitors, injector cleanliness additives, smoke suppressants, drag reducing additives, metal deactivators, dispersants, detergents, demulsifiers, dyes, markers, static dissipaters, biocides, and/or corrosion inhibitors.
  • Some eukaryotic algae synthesize as much as 70% of their dry weight as oils. These oils, which are the product of photosynthesis, are ideal biofuel candidates. Organisms that produce these oils can be grown in ponds in deserts so no arable croplands will be lost to biofuel production. Use of such algae is typically limited by their slow growth.
  • the provided methods can be used to manipulate the genomes of organisms, for example, to engineer new organisms, e.g., prokaryotic organisms, that express enzymes involved in the oil synthesis pathways, for example, by manipulating transcriptional promoters, translation signals, and codon optimization. The methods can be used to modify genomes of
  • photosynthetic bacteria to engineer new bacteria having chimeric genomes that produce biofuels, such as the oils produced by algae, instead of the normal products of photosynthesis (glucose).
  • Recombinant microorganisms made using the disclosed methods can contain an engineered biosynthetic pathway capable of converting glucose and other sugars derived from lignocellulosic biomass to geraniol.
  • Recombinant microorganisms e.g., strains of photosynthetic microorganisms
  • branched-chain alcohols including, for example, 2-methyl-l-butanol, 3 -methyl- 1-butanol, and isobutanol.
  • One aspect involves the production of recombinant photosynthetic microorganisms via introduction of heterologous genes that encode enzymes that enhance the production and decarboxylation of 2-keto branched-chain acids, leading to the production of the
  • microorganisms can be engineered such that branched chain alcohols are enzymatically dehydrated in vivo to produce various branched-chain alpha- olefins.
  • nucleic acid molecules can be used to transform organisms, such as photosynthetic organisms and prokaryotic organisms, for synthesizing fatty acids and fatty acid products such as fatty aldehydes, fatty alcohols, fatty esters, including wax esters, and hydrocarbons. Also included are organisms transformed using the methods provided herein.
  • Recombinant microorganisms e.g., recombinant photosynthetic microorganisms
  • a nucleic acid molecule comprising at least one recombinant expression system that produces at least one exogenous acyl- ACP thioesterase, wherein said acyl- ACP thioesterase liberates a fatty acid chain that contains 6-20
  • a thioesterase can be used to liberate a fatty acid chain that contains 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbons.
  • the fatty acids thus recovered can be further modified synthetically or used directly as components of biofuels or chemicals.
  • the appropriate plastid transit peptide encoding region to the host organism may be substituted.
  • Preferred codons may also be employed, depending on the host.
  • Genomes of microbes can be further modified to include an expression system for a heterologous gene that encodes a ⁇ -ketoacyl synthase (KAS) that preferentially produces acyl-ACPs having medium chain lengths.
  • KAS ⁇ -ketoacyl synthase
  • Such KAS enzymes would serve to increase the availability of acyl-ACP molecules of the proper length for recognition and cleavage by the heterologous medium-chain acyl-ACP TE.
  • a photosynthetic host cell containing a heterologous acyl-ACP TE gene may be further modified to include an expression system for a heterologous gene that encodes a multifunctional acetyl-CoA carboxylase or a set of heterologous genes that encode the various subunits of a multi-subunit type of acetyl-CoA carboxylase.
  • heterologous genes that encode additional enzymes or components of the fatty acid biosynthesis pathway could also be introduced and expressed in acyl-ACP TE-containing host cells.
  • the photosynthetic microorganism may also be modified such that one or more genes that encode beta-oxidation pathway enzymes have been inactivated or downregulated, or the enzymes themselves may be inhibited to prevent the degradation of fatty acids released from acyl-ACPs, thus enhancing the yield of secreted fatty acids.
  • the desired products are medium-chain fatty acids
  • the inactivation or downregulation of genes that encode acyl-CoA synthetase and/or acyl-CoA oxidase enzymes that preferentially use these chain lengths as substrates would be beneficial.
  • Photosynthetic microorganisms may also be modified such that one or more genes that encode storage carbohydrate or polyhydroxyalkanoate (PHA) biosynthesis pathway enzymes have been inactivated or down-regulated, or the enzymes themselves may be inhibited.
  • PHA polyhydroxyalkanoate
  • Examples include enzymes involved in glycogen, starch, or chrysolaminarin synthesis, including glucan synthases and branching enzymes.
  • Other examples include enzymes involved in PHA biosynthesis such as acetoacetyl-CoA synthase and PHA synthase.
  • the disclosed methods are also useful for production of industrial enzymes and industrial organisms.
  • the disclosed methods can be used to generate new organisms with chimeric genomes, e.g., a genome that is a chimera of Clostridium acetobutylicum and Clostridium cellulolyticum that has the genes from the former species that encode the enzymes needed to synthesize ethanol from glucose and genes from the latter species that encode cellulases that can efficiently degrade cellulose.
  • the provided methods and compositions can be used to produce cells and organisms that efficiently degrade cellulose to produce the ethanol.
  • This example describes the successful cloning of bacterial genomes in yeast host cells, using three different cloning approaches provided herein ( Figure 2). As described below, each approach yielded a host cell having a nucleic acid containing the donor bacterial genome joined to a yeast host vector. Transfer of donor genomes to host cells by these approaches can be used with the provided methods for propagation and modification of donor genomes in host cells, and transplantation of donor genomes from host cells into recipient cells ( Figure 1).
  • the first cloning approach shown in Figure 2A, was carried out by inserting the yeast host vector into the donor bacterial genome prior to transformation of yeast, thereby producing the joined molecule that then was transformed into yeast.
  • the second and third approaches shown in Figs. 2B and 2C, respectively, were carried out by joining the donor bacterial genome and yeast vector using homologous recombination, within the host cell.
  • the bacterial genome and yeast (host) vector were co-transformed into the yeast host cell ( Figure 2B).
  • the host vector was a linear yeast vector containing terminal regions of homology to a site in the bacterial donor genome and was thereby inserted by homologous recombination.
  • This example describes successful cloning (transfer and propagation) of three different Mycoplasma donor genomes (M. genitalium strain MS5; M. mycoides subspecies mycoides, Large Colony strain GM12; and M. pneumoniae strain M129-B170 (ATCC 29343)) in host yeast cells, using the first approach for introduction of a donor genome into a host cell.
  • the M. genitalium strain MS5 was a derivative of M. genitalium G37 (GenBank No. L43967). It was created by interruption of a gene in the G37 strain, as described in Dhandayuthapani et al., J Bacteriol 183, 5645 (2001).
  • the M. mycoides subspecies mycoides, Large Colony strain GM12 (genome sequence having Genbank Accession no.
  • NZ_AAZK01000004.1 (GI: 149364882) is described in DaMassa et al, Am J Vet Res 44, 322 (1983) and Lartigue et al., Science 317, 632 (2007).
  • the M. pneumoniae strain M129- B170 (ATCC 29343) was a derivative of M. pneumoniae Ml 29, GenBank Accession Number U00089.2 (GI: 26117688).
  • Transfer of each Mycoplasma donor genome was carried out by inserting a yeast vector into the donor genome to generate a nucleic acid molecule containing the genome and the host vector, and then introducing that molecule into the host cell via transformation.
  • a yeast vector into the donor genome to generate a nucleic acid molecule containing the genome and the host vector, and then introducing that molecule into the host cell via transformation.
  • the vector pmycYACTn ( Figure 3 A) was 10 kb in length and contained: (i) a high copy origin (ori) from pUC19 and an ampicillin resistance marker for propagation in E. coli; (ii) the IS256 transposase gene and inverted repeats for transposition into a Mycoplasma genome; (iii) tetMwA lacZ markers, both expressed from spiralin promoters, as described in Lartigue et al, J Bacteriol 164, 1094 (1985), for selection and screening in E. coli and Mycoplasmas; and (iv) an autonomously replicated sequence (ARS) and a centromere sequence (CEN), for replication and segregation in yeast; and (v) HIS3, a selectable yeast marker.
  • ARS autonomously replicated sequence
  • CEN centromere sequence
  • the vector was constructed from overlapping fragments, illustrated in Figure 3E (labeled as fragments 1, 2, and 4-7), using a published in vitro assembly method (Gibson et al, Science 319, 1215 (2008), U.S patent application serial number 12/247,126, and
  • Fragment 1 (1846 base pairs (bp)) contained the E. coli Ampicillin resistance (bla), pUC19 origin, which was included to facilitate high yield plasmid isolation.
  • Fragment 2 (1256 bp) contained the Mycoplasma IS256 transposase and promoter and an IS256 inverted repeat (labeled as "3" in Figure 3E), which was included to facilitate vector insertion by transposition.
  • Fragments 4 2294 bp) and 5 (3335 bp) each contained the Mycoplasma Spirulin promoter, and contained Mycoplasma tetracycline resistance (tetM) and LacZ genes, respectively, which were included to facilitate selection for vector insertion into the donor genome.
  • Fragment 5 additionally contained an IS256 inverted repeat, to facilitate insertion by transposition.
  • Fragment 6 (847 bp) contained the S. cerevisiae HIS3 gene and promoter, which was included to facilitate selection of transformation into host cells.
  • Fragment 7 (505 bp) contained the S. cerevisiae ARSH4 and CEN6 genes, which was included to facilitate replication and segregation. See Figure 3E.
  • the template was a 3.7 kb Pcil-Sall fragment of the pISM31.1 vector, described in Pour-El et al, Plasmid 47(2): 129-37 (2002).
  • the template was the pARS-V plasmid, described in Noskov et al., BMC Genomics 4(1): 16 (2003).
  • Table 1 Primers for PCR of fragments used in construction of
  • E. coli -Mycoplasma-Ye&st shuttle vector pmycYACTn E. coli -Mycoplasma-Ye&st shuttle vector pmycYACTn.
  • PCR was carried out in a 100 ⁇ . reaction volume, using 10 ng of the plasmid template. Primers indicated in Table 1, Phusion DNA polymerase, HF buffer (New England Biolabs, Ipswich, MA) were included in amounts according to the manufacturer's protocol, with extra MgCl 2 added for a final concentration of 2.0 or 3.0 mM. Cycling conditions were as follows: 98°C for 30 seconds, followed by 30 cycles of incubation at 98°C for 10 sec, annealing for 30 seconds, and incubation at 72°C for 90 seconds, followed by 72°C for 5 minutes. Annealing temperatures varied among the cycles and among PCRs for different fragments, as follows.
  • Annealing temperature was between 46°C and 59°C for cycles 1-5, and increased by 5°C (to between 51°C and 64°C) for cycles 6-30.
  • cycle 1-5 annealing temperatures were 56°C and 59°C for fragment 1; 46°C and 48°C for fragment 5; 46°C and 50°C for fragment 4; 46°C for fragment 2; and 48°C and 52°C for fragments 6 and 7.
  • each temperatures was 5°C higher than the temperature for cycles 1-5.
  • PCR products were pooled and amplicons gel-purified using ⁇ -agarase (New England Biolabs, Ipswich, MA).
  • one of the PCR primers contained the standard 20 base pairs of homology to the template at the desired location, but further contained 26 base pairs of homology to 2 additional copies of the IS256 inverted repeat, which were also present in other parts of the plasmid.
  • these IS256 copies were separated from the desired template portion of pISM31.1 with a double restriction enzyme digest with Pcil and Sail, followed by agarose gel-purification of the correct resulting 3.7 kb fragment, which then was used as the template in the PCR amplification of fragment 2.
  • CBA chew back assembly
  • Transformants were selected with carbenicillin. DNA from selected clones was screened for the correct sized plasmid using three separate restriction digests. The presence of the various elements of the plasmid was tested phenotypically, as follows. Propagation in E. coli ensured functionality of the pUC19 origin; selection in E. coli with carbenicillin and tetracycline and screening with X-gal verified that intact copies of the bla, tetM, and lacZ markers were present; and successful transformation of yeast with the isolated vector demonstrated that the HIS3, ARSH4, and CEN6 markers were viable. Presence of a functional transposon was confirmed by transformation into Mycoplasma, as described below. b. Construction of miniTn-Puro-JCVI-1.7 vector
  • the miniTn-Puro-JCVI-1.7 vector (Figure 3B) was 14 kb in length, and was identical to the pmycYACTn, with the following exceptions: (i) it did not contain lacZ; (ii) instead of a tetM marker, it contained a puromycin resistance marker; and (iii) it contained a bacterial artificial chromosome (BAC) vector. ii. Insertion of host vectors into donor genomes
  • the vector pmycYACTn vector was inserted into the donor genome by electroporation into the M. genitalium donor cells, as described in J. I. Glass et ah, PNAS USA 103, 425 (2006). Transformants were selected by growth in the presence of tetracycline and a single clone chosen for further analysis. Direct genomic sequencing (as described in J. I. Glass et ah, PNAS USA 103, 425 (2006)), using primers internal to the vector, was performed to determine the site of vector insertion.
  • the chosen clone contained the sequence between and including the two IS256 inverted repeats of pmycYACTn, indicating that transposition had occurred, as designed ( Figure 3C).
  • the transposase, pUC19 origin, and ampicillin resistance gene were lost during transposition.
  • the host vector inserted into the donor genome within the nonessential MG411 gene (J. I. Glass et ah, PNAS USA 103, 425 (2006); C. A. Hutchison et ah, Science 286, 2165 (1999)).
  • Mycoplasma donor cells were transformed with pmycYACTn using PEG, as described in K. W. King and K. Dybvig, Plasmid 26, 108 (1991). Transformants were selected by growth on plates supplemented with tetracycline.
  • the donor genomes containing yeast vector insertions were isolated in agarose plugs using low melting point agarose and the Bio-Rad CHEF Mammalian Genomic DNA Plug Kit (Bio-Rad Laboratories, Hercules, CA), following the protocol suggested by the manufacturer.
  • Agarose plugs containing the Mycoplasma genomes with insertions were dialyzed against 10 mM Tris pH 8.0 (in some cases with 1 mM EDTA) for 1 hour. Plugs were then dialyzed against 10 mM (6%) PEG 6000 (United States Biochemical), 0.6 M NaCl for several hours, as described in S. Katsura et al, Electrophoresis 21, 171 (2000).
  • This PEG/NaCl treatment was not carried out for isolation of M. mycoides LC genomes containing vector for transfer into W303a cells (see below), or for isolation of M. pneumoniae genomes containing vector for transfer into VL6-48N cells (see below).
  • yeast spheroplasts were transformed with DNA from the plugs, using the published method described by N. Kouprina and V. Larionov, Nat Protoc 3, 371 (2008), except that cultures were sometimes grown to less than the recommended OD.
  • mycoides LC cll.l genome with insert also was transferred into a recombination-deficient yeast strain, VL6-48-A54G, which is defective in the RAD54 gene (MATa his3-A200 trpl- ⁇ ura3-52 lys2 ade2- 101 metl4 rad54-Al::
  • VL6-48-A54G Transformation into the VL6-48-A54G strain was done to address the possibility that the genome would be unstable in other yeast, due to recombination among the multiple nearly-identical 1.5 kb IS 1296 copies.
  • Yeast strains defective in the RAD 54 gene can decrease the occurrence of a variety of recombination events in yeast artificial chromosomes (YACs) (Y. Le and M. J. Dobson, Nucleic Acids Res 25, 1248 (1997)).
  • YACs yeast artificial chromosomes
  • DNA from each of the transformed yeast host cells was analyzed to confirm transfer of complete host vector-containing donor genomes into the host cells.
  • Mycoplasma genomes cloned in yeast were screened by multiplex PCR (MPCR) to confirm completeness. MPCRs were carried out using 1 or 2 sets of 10 amplicons each, using primers from IDT with the Multiplex PCR Kit from Qiagen (Valencia, CA). Individual reactions are described in more detail below.
  • DNA was isolated in agarose plugs using the protocol "Preparation of Agarose Embedded Yeast DNA" from the Bio-Rad CHEF-DR III manual. Where indicated, plugs were pre-electrophoresed at constant voltage for several hours to remove yeast chromosomal DNA. Where indicated, to increase the efficiency of this step, plugs were first digested with AsiSI, Fsel, and RsrII, which cleave yeast chromosomes but do not have recognition sites in M. genitalium or M. mycoides LC.
  • Isolated DNA was subject to restriction digestion (with indicated enzyme(s)), followed by field-inversion (Bio- Rad FIGE Mapper) or pulsed-field (Bio-Rad CHEF-DR II or III system) electrophoresis, as indicated, or linearization by heating at 55 °C for 1 hr. Where indicated, Southern blots were performed on the gels. Southern blotting was carried out as described in D. G. Gibson et al, Science 319, 1215 (2008), except in some cases where probe labeling and detection used the Amersham AlkPhos Direct Labeling and Detection System with CDP-Star (GE Healthcare, Piscataway, NJ). a. Isolation and analysis of M. genitalium genomes from yeast host PCR
  • An M. genitalium synthetic genome (sMgTARB AC37, generated as described in (D. G. Gibson et al, Science 319, 1215 (2008)) and a native M. genitalium genome were used as positive controls.
  • PCR primers listed in Table 3 were designed and used to generate amplicons to assess completeness of the transferred M. mycoides genomes. As shown in Figure 5, an amplicon was located between most pairs of IS 1296 elements, indicated by arrowheads on the map of the genome with insert illustrated in Figure 5. An additional 230 bp amplicon was a region of the HIS3 marker of the yeast vector. Another primer set (NSF1179/18 and NSR1642/16; see Table 3) produced a 464 bp amplicon (region of S. cerevisiae rDNA), and was used as a positive control for the assay. The sets of primers and sizes of amplicons produced thereby are listed in Table 3.
  • a CHEF gel analysis of three of these five clones recovered after transformation of the VL6-48N strain with M. mycoides cll.l genomes containing yeast vectors was performed as follows: DNA from clones (07, 14, and 38) was isolated in agarose plugs as described above, without pre-electrophoresis. The isolated DNA was digested with BssHll and then separated by pulse-field gel electrophoresis (CHEF). For each clone, the parent clone and 3-4 subclones were analyzed (gel results not shown). Markers and controls used in the analysis were: (1) Low Range PFG Marker (New England Biolabs, Ipswich, MA); (2) S.
  • CHEF pulse-field gel electrophoresis
  • Genomes isolated from 20 individual clones recovered after transfer of M pneumoniae genomes containing yeast vectors to strain VL6-48N were analyzed by multiplex PCR as described above to confirm completeness. Two different multiplex PCRs were performed. Primers for set 1 and set 2 and the size of amplicons produced thereby are listed in Table 4 (gel results not shown). As shown in Figure 4 A, amplicons (indicated with black bars and numbers; inside: set 1, outside: set 2) were evenly spaced around the M. pneumoniae genome containing the vector insert. The results indicated that thirteen of the twenty transformants were complete.
  • CHEF gel analysis was performed on nine of these complete transformants.
  • DNA from these clones was isolated in agarose plugs using the protocol "Preparation of Agarose Embedded Yeast DNA" from the Bio-Rad CHEF-DR III manual.
  • plugs were pre-electrophoresed at constant voltage for several hours to remove yeast chromosomal DNA.
  • the DNA then was digested with NotI or Sbfl.
  • the fragments (numbered 1-6) of the M. pneumoniae genome with vector insert that are produced by these enzymes are indicated on the map in Figure 4B, with the fragments and their sizes listed in columns next to the map.
  • Table 5 summarizes results from Example 1(A), in which three Mycoplasma genomes containing integrated yeast vectors were transferred into yeast. Clones were screened for completeness by multiplex PCR with 1 or 2 sets of 10 amplicons each. Clones were tested for size by restriction digestion and gel electrophoresis, followed in some cases by southern blot.
  • the linearized, whole M. genitalium genome was transferred into yeast cells using the method depicted in Figure 2B, by homologous recombination of the genome with a yeast vector within the host cells.
  • the M. genitalium genome contains 3 single-cut restriction sites, 2 of which lie within its rRNA operon. The third lies at the 3' end of a tRNA coding sequence. Because the cloning could be designed to preserve the integrity of the tRNA, a vector was inserted by homologous recombination adjacent to this Ascl site.
  • the yeast cloning vector pARS-VN (described in V. N. Noskov et al.
  • PCR of the vector with these primers amplified the entire vector except for 9 bp between the unique Clal and Xhol restriction sites, producing a 6.5 kb product.
  • M. genitalium DNA was isolated in agarose plugs, as follows, to minimize breakage.
  • M. genitalium genomic DNA was isolated in two batches in low melting point agarose plugs from strain MS5 grown in SP-4 medium. The culture for batch 2 was supplemented with gentamicin to 200 ⁇ g/ml. Adherent cells were rinsed twice with PBS and then scraped into a buffer containing 8.0 mM HEPES, pH 7.4, 272 mM sucrose, and 10% glycerol.
  • Each plug contained DNA from about 6 (batch 1) or 10 (batch 2) cm 2 of confluent cells.
  • cells in agarose were incubated for a period between overnight to 2 days at 50°C in 0.4 M EDTA, 0.4% N-lauroyl sarcosine, and 2 mgs/ml proteinase K, followed by a buffer change and a second treatment under the same conditions for the same time range.
  • Plugs were dialyzed thoroughly against 10 mM Tris, 50 mM EDTA, then dialyzed 2 times for 2 hours each in 10 mM Tris, 50 mM EDTA
  • the plugs were melted and digested with agarase, as follows.
  • the first batch of plugs was electrophoresed twice by CHEF (Bio-Rad), which removed broken DNA, while leaving circular genomes intact (to increase the frequency of intact circular genomes in the plugs).
  • the first electrophoresis was carried out on a 1% pulsed-field agarose gel, with 0.5X TBE and a 50-90 second switch time, over 20 hrs.
  • the second electrophoresis was carried out on a 1% low melting point gel, with IX TAE and a 60-120 sec switch time, over 24 hrs. Both gels were run at 120°, 6 V/cm, at 14°C.
  • Three plugs from each of the two batches were dialyzed thoroughly against sterile IX TAE, melted for several minutes at 73°C, equilibrated to 42°, then digested with ⁇ -Agarase I (New
  • yeast spheroplasts were transformed with DNA from the plugs, using the published method described by Kouprina and Larionov, Nat Protoc 3, 371 (2008), except that cultures were sometimes grown to less than the recommended OD.
  • yeast cells were suspended in 1M sorbitol and treated with ZymolyaseTM ( ⁇ -l, 3-glucan laminaripentaohydrolase) before transformation to remove cell walls. DNA recovered from agarose plugs was incubated with the spheroplasts. After recovery in growth medium, cells were plated in selective medium.
  • Clones were picked and evaluated by multiplex PCR and gel electrophoresis as described in Example 1 A(v)(a), above, except that two sets of 10 amplicons, instead of one set, were used.
  • the primers used to generate the first set of amplicons were those listed in Table 2, above.
  • the primers used to generate the second set of amplicons, and the sizes of amplicons produced thereby, are listed in Table 6, below.
  • Transformation with Ascl-digested DNA yielded forty-five transformants. All of these were examined by multiplex PCR, with the primers to produce the twenty amplicons as discussed above (Table 2, Table 6). Twenty-one appeared to be complete. These twenty-one transformants were examined by Southern Blot of a CHEF gel and fifteen appeared to be the correct size. Transformation with undigested M. genitalium genomes yielded fifty
  • This example describes successful transfer to and propagation of M. genitalium donor genomes in yeast host cells, using the method illustrated in Figure 2C.
  • genomes are assembled in the host cell (yeast) by homologous recombination of multiple overlapping fragments of the genome.
  • This strategy has been performed using pieces derived from E. coli BAC clones ⁇ See Gibson et al, Science 319, 1215 (2008), including supplemental online materials;
  • This example describes production of a diploid yeast host strain, carrying two donor Mycoplasma genomes that were transferred in using the method depicted in Figure 2A (see Example 1 A, above). For this process, two haploid strains were crossed as described in D. C. Amberg et al. , Methods in Yeast Genetics: A Cold Spring Harbor Laboratory Course Manual. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, ed. 2005, 2005), pp. 230, each of which carried one of the genomes.
  • the W303a strain (mating type a) containing the M. genitalium cll6-2 was mated with the VL6-48 strain (mating type a) containing M. mycoides LC cll.l (see Example 1 A and Table 1, above).
  • the HIS3 marker in the M. genitalium genome was replaced with a TRP marker, as follows, to allow selection of diploids carrying both genomes on medium lacking histidine and tryptophan.
  • a 1059 bp TRP1 fragment was amplified by PCR from the plasmid pRS304 (described by Sikorski and Hieter, Genetics 122, 19 (1989), Genbank Accession No. U03436.1, gi number 416305).
  • the fragment which had homology to M. genitalium MS 5 cll6-2, was amplified from the plasmid using primers with the following sequences:
  • AGCAGATTGTACTGAGAGTGCACC (SEQ ID NO: 131) and CTACATAAGAACACCTTTGGTGGAGGGAACATCGTTGGTACCATTGGGCGC
  • GCATCTGTGCGGTATTTCACACCGC SEQ ID NO: 1312.
  • each primer sequence the portion homologous to the M. genitalium cll6-2 sequence is in bold.
  • the 1059 bp fragment was transformed into yeast using lithium acetate, using techniques described by Gietz et al, Nucleic Acids Res 20, 1425 (1992).
  • AGTCAAGTCCAGACTCCTGT (SEQ ID NO: 134)
  • yeast vector in the synthetic M, genitalium genome generated as described by Gibson et al, Science 319, 1215 (2008) and in Example 1C, above was transferred from its original site within the RNaseP gene to a new site in MG411 so as not to interrupt an essential gene.
  • the yeast clone containing the synthetic M. genitalium as described in Example 1C 5 above was co-transformed with two (2) fragments.
  • the first fragment which was 1842 bp in length, inserted yeast vector sequence containing URA3, the GAL1 promoter, and a centromere into MG411. This fragment was generated by PCR using primers with the sequences:
  • the template for this PCR was a construct having the sequence provided in SEQ ID NO: 137 (Table 8).
  • the second fragment was 302 bp in length, having the following sequence:
  • TRP1 was inserted into MG411, as follows: an 1177 bp TRP1 gene fragment with homology to MG411 was amplified from the plasmid pRS304 (Genbank Accession No. U03436.1, gi number 416305), using two primers with the following sequences:
  • TRP1 gene insertion was confirmed by PCR using a set of primers (sequences GCCATTGTTTCACTAATTGC (SEQ ID NO: 143) and TAATCCTATCTTTGGAGCTT (SEQ ID NO: 144)) that amplified 1739 bp (if insertion occurred) and 680 bp if it did not. Cotransformants, which were His- Trp- Ura+, were selected. Restoration of RNase? was confirmed by PCR amplification of a 513 bp product using primers with sequences CTCCATCATGCGCAGTAATA (SEQ ID NO:145) and CTTTAAA
  • CAGGCAGGAATTTGATTCCC (SEQ ID NO: 148).
  • the vector inserted in the new site did not contain an ARS.
  • the results of these studies confirmed that the vector containing the M. genitalium donor genome did not require the presence of the ARS sequence for maintenance in yeast host cells.
  • the M. genitalium is AT-rich and thus is likely to contain sequences that can function as ARS in yeast. ARS-like sequences are frequent in eukaryotic AT-rich DNA (See Montiel et al, Nucleic Acids Res 12, 1049 (1984); Stinchcomb et al., PNAS USA 77, 4559 (1980)).
  • molecules as large as about 2 MB were recovered and detected by Southern blotting. These molecules likely represented clones of concatamers, and reveal that the provided methods can be used to clone and transfer larger genomes and nucleic acid molecules into yeast host cells. Such methods can be used to generate host cells containing donor genomes, which then can be propagated and modified in the host cells and transplanted into recipient cells using the provided methods.
  • FIG. 18 A provides a schematic of the YCpMmycl .1 genome and the position of the integrated YCp is shown. The nine (9) individual primer pairs used in the PCR amplifications are shown at their approximate locations in the genome and are numbered corresponding to the amplicons in Figure 18B.
  • the stability of the M. mycoides genome during propagation in yeast was tested by two methods. In the first, a yeast culture of a clone containing the genome was plated on solid synthetic media lacking histidine for two days and then indivisual colonies were patched onto a new plate.
  • the methods include steps for overcoming potential incompatibilities among the different cell types, such as methods for successfully transplanting into recipient cells donor genomes that have been propagated in host cells.
  • Example 3 describes the successful transplantation of a whole donor genome ( mycoides LC (Genbank accession NZ_AAZK00000000.1 (GI: 149364883), which had been propagated in yeast host cells, into recipient cells of a different species (M capricolum), using the provided methods.
  • This Example describes analysis of differences among these three different organisms (donor, host, and recipient), and the development of various processes that can be used to overcome these differences using the provided methods.
  • Example 2B demonstrates that sufficient amounts of purified, intact donor
  • Mycoplasma genomic DNA can be recovered from yeast host cells for transplanting into the recipient cells.
  • Example 2C demonstrates a provided transplantation method for transplanting native bacterial donor genomes into recipient cells with high efficiency.
  • Example 2D describes evaluation of restriction-modification systems (not present in yeast) in the host and recipient cells, and of methyltransferase (which are expressed in yeast) expression and effects of methyltransferases on the donor genomes and activation.
  • Example 2E describes treatments used in the provided methods to overcome host-donor-recipient incompatibility issues associated with restriction-modification (R-M) systems.
  • Example 2F describes mutation of the R-M system of recipient cells, and
  • Example 2G demonstrates successful transplantation of donor genomes (transferred from Mycoplasma to yeast host cells) from the yeast host cells to recipient bacteria of different species.
  • Escherichia coli DH10B [T-mcrA A(mrr-hsdRMS-mcrBC) ⁇
  • E. coli cells were grown in Luria- Bertani (LB) broth medium or in LB agar at 37° C. Depending upon the selection markers present in a given plasmid, E.
  • Mycoplasma capricolum subsp. capricolum strain California KidTM
  • Mycoplasma mycoides subsp. mycoides strain GM12
  • the Mycoplasma cells were grown at 37°C in liquid or solid SP4 medium (Tully et al.
  • Mycoplasma transformed with plasmid or whole-genome were grown at 37°C in SP4 medium supplemented with 5 ⁇ g/ml of tetracycline or 8 ⁇ g/ml of puromycin.
  • Beta- galactosidase activity was detecting by plating Mycoplasma on solid medium containing 15( ⁇ g/ml of 5-bromo-4-chloro-3-indolyl- -D-galactopyranoside (X-gal, Promega, Madison, WI).
  • M. capricolum Two strains of Mycoplasma capricolum subsp. capricolum (M. capricolum) were used as recipient cells in the following Examples: wild-type (wt) M. capricolum and a restriction-free M. capricolum mutant (M caprico um- RE) that was obtained by inactivation of the CCATC -restriction enzyme gene in wt M. Capricolum (described in Example 2F, below). Donor genomic DNA for transplantation was from a Mycoplasma mycoides subsp.
  • mycoides LC M mycoides LC
  • cl 1.1 mycoides LC (M mycoides LC) clone (cl 1.1), described in Example 1, above, the genome of which contained the tetracycline resistance marker, the lacZ gene, and the yeast centromere plasmid integrated between ORF04334 (lppA) and ORF04335 (transposase B of IS1296).
  • Mycoplasma genomic DNA was prepared in agarose plugs either directly from M. mycoides LC cells clone 1.1 (native genomic DNA) or from yeast host cells carrying M. mycoides clone 1.1 genome, generated as described in Example 1, above.
  • yeast W303a cells that had been transformed with the Mycoplasma mycoides Large Colony ( mycoides LC) GM12, clone 1.1, genome, as described in Examples lA(ii)b and lA(iv), above, were embedded in agarose plugs as described below.
  • This genome carried the tetracycline resistance gene (tetM) and ⁇ -galactosidase gene (lacZ). DNA from the plugs was isolated and evaluated. Genomic DNA from native yeast not carrying donor genomes and native donor Mycoplasma cells isolated were evaluated in a similar manner for comparison. i. Yeast agarose plugs containing donor genomes
  • Yeast cultures were grown at 30°C in selective medium until the OD 0 o reached approximately 1.5.
  • Yeast cells were embedded in agarose plugs and DNA isolated from the plugs, using the CHEF mammalian Genomic DNA Plug Kit from Bio-Rad Laboratories (Valencia, CA), following the manufacturer's suggested protocol with the following details/modifications.
  • 6 x 10 9 yeast cells instead of 6 x 10 8 cells were used per mL of plugs to be made, to yield 6 x 10 cells per plug.
  • mycoides LC genomic DNA and those containing control yeast DNA were prepared for analysis using the CHEF mammalian Genomic DNA Plug Kit from Bio-Rad (Valencia, CA).
  • a series of 3 agarose plugs (A, B, and C) was made for the yeast containing the donor genomes (A2, B2, and C2) and for the native yeast (Al, Bl, and CI).
  • the plugs were washed at room temperature, two times for one hour in 1 mL of IX TE buffer, and equilibrated for one hour at room temperature in 1 mL of IX NEB buffer 2 supplemented with BSA (100 ⁇ g/mL).
  • plugs B and C were incubated overnight at 37 °C with 50 units of AsiSl, Rsrll, and Fsel restriction enzymes (New England Biolabs) in a 500 ⁇ reaction volume, which specifically cut yeast genomic DNA and left the donor DNA intact.
  • Plug A was incubated without these enzymes under the same conditions. All 3 plugs then were washed at room temperature for 1 hour with 1 ml of IX TE buffer and loaded on 1% TAE agarose gel (120 minutes, 120 volts), to remove digested yeast genomic DNA fragments from the plugs.
  • agarose plugs were removed from the wells and washed two times for one hour in 1 mL 0.1X TE buffer and equilibrated for one hour in 1 mL of IX NEB buffer 2 (New England Biolabs, Ipswich, MA) supplemented with BSA (100 ⁇ g/mL).
  • IX NEB buffer 2 New England Biolabs, Ipswich, MA
  • BSA 100 ⁇ g/mL
  • M. mycoides LC ⁇ tetM, lacZ, YCp All intact genomic DNA isolation from M. mycoides LC was performed as described by Lartigue et al, Science 317, 632 (2007), with some modifications, in particular in the way the cells were cultured prior to isolation. Five hundred (500) mL M. mycoides LC ⁇ tetM, lacZ, YCp) cells were grown in SP4 medium, supplemented with 10 ⁇ / ⁇ 1 tetracycline and 10 ⁇
  • MLC gDNA M mycoides LC genomic DNA
  • mycoides LC cells per plug For comparison to the genomic DNA from yeast, the plugs containing native DNA and the plugs containing M. mycoides LC genomic DNA isolated from yeast (see above) were digested with PspXl to linearize MLC genome as described above for analysis on a pulse-field gel. iii. Comparison of recovered genomic DNA on pulse field gel
  • the amount of M. mycoides LC genomic DNA in yeast cells was estimated by comparing the amount of isolated genomic DNA from yeast cells in agarose plugs to the various amounts of native genomic DNA isolated from 2-fold serial dilutions of M. mycoides LC cells in agarose plugs. For this process, the yeast agarose plugs (Example 2B(i), plugs Al, Bl, and CI and A2, B2, and C2) and the eight M.
  • mycoides LC agarose plugs (Example 2B(ii) were subjected to electrophoresis in a 1% certified pulse-field agarose gel (Bio-Rad, Valencia, CA) in TAE IX, with contour-clamped homogeneous electric field (Chu et al, Science 234, 1582 (1986)) (CHEF DR III; Bio-Rad). Pulse times were ramped from 60 to 120 s over 27h at 4.5 V/cm. After migration, the gel was stained with SYBR® GOLD nucleic acid stain (Invitrogen, Carlsbad, CA) (1/10,000 dilution) and PFGE patterns were scanned with a GE Typhoon 9410 imager.
  • SYBR® GOLD nucleic acid stain Invitrogen, Carlsbad, CA
  • PFGE patterns were scanned with a GE Typhoon 9410 imager.
  • the S. cerevisiae CHEF DNA size marker was used to evaluate DNA
  • mycoides donor genomes selective digestion of yeast genomic DNA with the enzyme cocktail (AsiSl, Rsrll Fsel restriction enzymes), followed by electrophoresis (samples B2 and C2; see Example 2B(i), above) improved recovery of M. mycoides donor genomic DNA (data not shown).
  • linearization of the M. mycoides LC genome (C2) greatly improved recovery of the 1.2 Mb M. mycoides (C2).
  • No 1.2 Mb band was detected in the parallel samples from native (wild-type) yeast cells, which contained no M. mycoides genome (Bl, CI), confirming that the band at 1.2 Mb indeed represented the presence of M. mycoides genome in the yeast cells. The same band appeared in lanes containing native M. mycoides genomic DNA.
  • the amount of M. mycoides LC genomic DNA recovered from yeast cells was compared to that recovered from native M. mycoides LC genomic DNA standards, which had also been treated with PspXl as described above.
  • the amount of M. mycoides LC genomic DNA obtained from 6 x 10 8 yeast cells was similar to the amount of genomic DNA recovered from native M. mycoides LC plugs from series 7 (approximately 1.5 x 10 s native M. mycoides LC cells per plug).
  • a UV spectrophotometer was used to determine the amount of native M, mycoides LC genomic DNA present in melted plugs prepared from native M. mycoides cells as described in Example 2B(ii) above. Results are listed in Table 9, below. As shown in this table, plugs from series 7 contained approximately 12 ng/ ⁇ of genomic M. mycoides LC DNA (1.2 ⁇ g per 100 ⁇ , plug). As noted above, the pulse-field gel revealed comparable 1.2 Mb band intensity in the lane with this sample (7) and the lane containing DNA recovered from yeast sample (C2) (see Example 2B(iii) above)). Thus, it was determined that that quantity of M. mycoides LC genomic DNA per 100 ⁇ plug in recovered from the host cells containing donor genomes was equal roughly to 1 ⁇ g. Table 9: Quantification and transplantation of native M. mycoides LC genomic DNA
  • the recipient cells were centrifuged at 4575g for 15 minutes at 10° C, washed once in S/T buffer (10 mM Tris-HCl, pH 6.5; and 250 mM NaCl), resuspended in 200 ⁇ of CaCl 2 (0.1 M) and incubated on ice for 30 minutes.
  • S/T buffer 10 mM Tris-HCl, pH 6.5; and 250 mM NaCl
  • CaCl 2 0.1 M
  • the agarose plugs (series 1-7) containing M. mycoides LC genomic DNA were washed 2 times, for 30 minutes each, in 0.1X TE buffer [2 mM TRIS- HC1, pH 8.0 - 5 mM EDTA] under gentle agitation at room temperature. The buffer was completely removed and the agarose plugs were melted with 1/10 volume of 10X ⁇ -Agarase Buffer [10 mM Bis Tris-HCl pH 6.5; 1 mM Na EDTA] at 65° C for 10 minutes.
  • transplantation method described above can also be used to transform M. capricolum recipient cells with plasmid DNA (not in agarose plugs) by substituting 10 ⁇ g of plasmid DNA in solution for the 20 iL of melted agarose plugs.
  • Example 2D
  • the M. mycoides LC genome which has been sequenced (Genbank Accession No.: NZ_AAZK00000000; GI: 149364883), was predicted to contain six different restriction- modification systems (five Type II systems and a single Type III system).
  • the M. capricolum genome sequence showed the presence of a single Type II system.
  • the recognition site specificities of the Type II enzymes were predicted from the gene sequences (Dr. R. Roberts, personal communication), as described in Roberts RJ et al, Nucleic Acids Res. 35(Database issue) :D269-70 (2007).
  • genomic DNA from M. mycoides LC and M. capricolum was purified using the Wizard® Genomic DNA Purification Kit (Promega, Madison, WI), following the manufacturer's directions. Approximately 1 ⁇ g each of M. mycoides LC and M capricolum genomic DNA was incubated with Bed, Hinfl, HpyAV, Mbol and SfaNl, respectively, as described by the manufacturer (New England Biolabs, Ipswich, MA). The DNA was then analyzed by agarose gel electrophoresis (data not shown).
  • M. mycoides LC For M. mycoides LC, a 1 liter culture of the cells in SP4 medium was grown at 37°C until a pH of 6.2-6.3 was reached. The culture was harvested by separating the cells into five 200 mL fractions and centrifuging at 5,000xg in a SLA-1500 Sorvall rotor at 4°C for 15 minutes. Each M. mycoides LC pellet was then washed with 200 mL of 8mM Hepes, pH 7.4, and 272 mM sucrose and centrifuged at 5,000xg in a SLA-1500 Sorvall rotor at 4°C for 15 minutes.
  • Each resulting pellet was resuspended in 1 ml Extract buffer (20 mM Tris-HCl, pH 7.5, 0.1 mM EDTA, 150 mM NaCl, 1 mM DTT, and 10% glycerol) and subsequently sonicated on ice 5 times with 10-12 seconds bursts, using a microtip, at an output control of three.
  • Extract buffer (20 mM Tris-HCl, pH 7.5, 0.1 mM EDTA, 150 mM NaCl, 1 mM DTT, and 10% glycerol
  • Each solution was clarified by microcentrifugation at 18,000xg for 30 minutes at 4°C.
  • Each resulting soluble fraction was combined, tested for protein concentration, aliquoted into 200 ⁇ fractions, and stored at -80°C. Protein concentration of extracts prepared in this manner typically ranged from 15 to 25 mg/ml.
  • M. mycoides LC extract Restriction enzyme activity of the M. mycoides LC extract was tested as follows. Two (2) ⁇ g of genomic DNA, isolated using a WIZARD® Genomic DNA purification kit (Promega Corporation, Madison, WI) from M. capricolum, M. mycoides LC and M.
  • the aqueous phase was placed into a fresh tube with 4 ⁇ of 10X Blue Juice (Invitrogen, Carlsbad, CA) and 18 ⁇ of each solution was loaded onto a 0.8% IX TAE agarose gel and run under FIGE conditions of 120V, 0.1-0.6 linear and 80V, 0.1-0.6 linear for 16 hrs.
  • the agarose gel was then stained with SYBR® GOLD nucleic acid stain (Invitrogen, Carlsbad, CA) (1/10,000 dil.) for 30 minutes and scanned with a GE Typhoon 9410 imager.
  • capricolum extract was used in each reaction, and aliquots were removed and processed at 0, 15, 30 and 45 minutes intervals.
  • M. capricolum and M. mycoides LC contain active restriction-modification systems, which have the potential to affect activation of unmethylated M. mycoides LC donor genomes isolated from yeast.
  • Lambda DNA was digested by wild-type M. capricolum extract. Incubation of the DNA with the extract made from the M. capricolum RE(-) strain did not result in the appearance of bands indicating the absence of the restriction activity from this strain. e. Genome sequencing and genome comparison of M. mycoides donor and transplant clones.
  • transplants were entirely M. mycoides and not chimeras containing either yeast sequences or M. capricolum recipient cell sequences, and to determine whether the bacterial genomes were stable when cloned in yeast, the two sequences were compared. All of the assembled Atypelllres genome matched M. mycoides, except for those regions that matched the YCp vector. Additionally, except for differences deliberately built into the genome, the transplant M. mycoides genome sequence with the Type III restriction enzyme gene deletion was identical to the previously sequenced M. mycoides genome except at 95 sites. It should be noted that these 95 sequence differences are not differences between the donor M. mycoides strain used herein and the engineered genome. They are differences between the sequence of the M.
  • This example describes two methylation methods that were employed to protect Mycoplasma ⁇ e.g., M. mycoides LC) donor genome from cleavage by restriction-modification systems. Methylation assays were performed for each method, to confirm efficacy. i. Methylation by constructed and purified methyltransferases
  • CCATC-M The coding sequences of the five identified methyltransferases (CCATC-M, CCTTC-M, Typelll-M, GCATC-M and GANTC-M) from potential restriction-modification systems in M. mycoides LC were codon optimized for expression in yeast. These sequences were then constructed from multiple overlapping 60 bp oligonucleotides using a one-step isothermal DNA assembly method, described in Gibson et al., Nature Methods 6, 343 - 345 (2009) and in U.S. Patent Application No. 12/371,543, filed February 19, 2009, by the concerted action of a 5' exonuclease, a DNA polymerase, and a DNA ligase.
  • DNA fragments are first recessed by the 5' exonuclease, yielding single-stranded overhangs, which then specifically anneal, followed by gap-filling and covalent joining using the polymerase and the ligase.
  • the CCATC-M, CCTTC-M and Typelll-M sequences were cloned into the pTYBl expression vector (New England Biolabs, Ipswich, MA; SEQ ID NO: 156).
  • the GCATC-M and GANTC-M sequences were cloned into an N- terminal His tag expression vector with GATEWAY® recombination cloning technology (Invitrogen, Carlsbad, CA).
  • the CCATC-M and CCTTC-M expression plasmids were transformed into BL21(DE3) codon plus cells (Stratagene, La Jolla, CA) and transformants were used to separately inoculate 250 ml of ZYM-505 medium (Studier, FW, Protein Expr Puriflc 41 :207- 34 (2005)) containing 100 mg/ml carbenicillin and 34 mg/ml chloramphenicol and grown at 37° C with vigorous shaking (315 rpm). After approximately 4 hours, the cultures were transferred to 16° C and expression was induced by the addition of 0.3 mM IPTG, overnight.
  • the cells were pelleted, suspended in 50 ml Intein lysis buffer (25 mM HEPES-NaOH pH 7.2, 500 mMNaCl, 1 mM EDTA, 10% glycerol, plus protease inhibitors (Complete protease inhibitor cocktail, Roche Applied Sciences, Indianapolis, IN) ' ) and lysed by two passages through a high-pressure homogenizer.
  • Intein lysis buffer 25 mM HEPES-NaOH pH 7.2, 500 mMNaCl, 1 mM EDTA, 10% glycerol, plus protease inhibitors (Complete protease inhibitor cocktail, Roche Applied Sciences, Indianapolis, IN) ' ) and lysed by two passages through a high-pressure homogenizer.
  • the lysates were clarified by centrifuging at 20,000xg for 20 minutes at 4° C.
  • the clarified lysates were purified on a 1.5 ml column of chitin beads as suggested by the manufacturer (New England Biolabs, Ipswich, MA).
  • Fractions containing the appropriate methyltransferases were pooled and dialyzed against Enzyme buffer (50 mM HEPES-NaOH pH 7.2, 50 mM NaCl, 0.1 mM EDTA, 10% glycerol). Following dialysis, the
  • methyltransferases were concentrated using an Amicon Ultra Centrifugal Filter Unit
  • the TypeIII-M protein was purified using the same protocol, with the exception that after purification on the chitin column, the protein was further purified using a HiTrap MonoQ column (GE Heathcare).
  • the protein was loaded in Buffer A (50 mM HEPES-NaOH pH 7.2, 50 mM NaCl, 1 mM EDTA, 10% glycerol) and eluted with a linear gradient from 0- 100% Buffer B (50 mM HEPES-NaOH pH 7.2, lM NaCl, 1 mM EDTA, 10% glycerol).
  • Buffer A 50 mM HEPES-NaOH pH 7.2, 50 mM NaCl, 1 mM EDTA, 10% glycerol
  • Buffer B 50 mM HEPES-NaOH pH 7.2, lM NaCl, 1 mM EDTA, 10% glycerol.
  • the fractions containing TypeIII-M were pooled,
  • the M.GCATC and M.GANTC expression plasmids were transformed into BL21(DE3) codon plus cells and transformants were used to separately inoculate 2 ml of ZYM-505 medium containing 100 mg/ml carbenicillin and grown at 37 °C with vigorous shaking overnight. One milliliter of the overnight culture was used to inoculate 250 ml of ZYM-5052 media (Studier, FW, Protein Expr Purific 41 :207-34 (2005)). Cells then were grown for 20 hrs at 27° C with vigorous shaking.
  • the cells were pelleted, suspended in 50 ml Nickel lysis buffer (50 mM HEPES-NaOH pH 7.2, 500 mM NaCl, 30 mM imidizole, 10% glycerol, plus protease inhibitors (Complete protease inhibitor cocktail, Roche Applied Sciences, Indianapolis, IN)) and lysed by two passages through a high-pressure homogenizer.
  • the lysates were clarified by centrifuging at 20,000xg for 20 minutes at 4 °C.
  • the clarified lysates were purified using a 5 ml HisTrap column with Nickel lysis buffer as the running buffer and Nickel lysis buffer with 300 mM imidizole as the elution buffer.
  • the M.GCATC protein was pooled, dialyzed into Enzyme buffer containing 100 mM NaCl and concentrated as above.
  • the M.GANTC protein was further purified using a 1 ml HiTrap MonoQ heparin column utilizing Buffers A and B as for the M.Typelll.
  • the M.GANTC containing fractions were pooled, dialyzed into Enzyme buffer containing 100 mM NaCl and concentrated as described above.
  • the purified methyltransferases were used in methylation assays to determine whether they could methylate a plasmid containing Mycoplasma DNA.
  • the methylation assays were performed using buffer conditions described by Wilson and Hoffman, Anal Biochem 191, 370 (Dec, 1990). Reactions were performed in 100 ⁇ volumes, at 37° C.
  • Reaction mixtures contained 100 mM Tris-HCl, pH 7.5, 10 mM EDTA, 3 ⁇ DTT, 200 ⁇ S-adenosylmethionine (SAM), 3 ⁇ g pSmart-pMYCOl plasmid DNA (yeast-E. coli- Mycoplasma tri-shuttle vector).
  • M. mycoides LC For M. mycoides LC, a 1 liter culture of the cells in SP4 medium was grown at 37° C until a pH of 6.2-6.3 was reached. The culture was harvested by separating the cells into 5 x 200 ml fractions and centrifuging at 5,000xg in a SLA- 1500 Sorvall rotor at 4° C for 15 minutes. Each M. mycoides LC pellet was then washed with 200 ml of 8mM Hepes, pH 7.4, and 272 mM sucrose and centrifuged at 5,000xg in a SLA-1500 Sorvall rotor at 4° C for 15 minutes.
  • Each resulting pellet was resuspended in 1 ml Extract buffer (20 mM Tris-HCl, pH 7.5, 0.1 mM EDTA, 150 mM NaCl, 1 mM DTT, and 10% glycerol) and subsequently sonicated on ice 5 times with 10-12 seconds bursts using a microtip at an output control of 3.
  • Extract buffer 20 mM Tris-HCl, pH 7.5, 0.1 mM EDTA, 150 mM NaCl, 1 mM DTT, and 10% glycerol
  • Each solution was clarified by microcentrifugation at 18,000xg for 30 minutes at 4° C.
  • Each resulting soluble fraction was combined, tested for protein concentration, aliquoted into 200 ⁇ fractions and stored at -80° C. Protein concentration of extracts prepared in this manner typically ranged from 15 to 25 mg/ml.
  • Methylation assays were used to demonstrate the ability of the crude extracts to methylate Mycoplasma plasmid DNA grown in various host cells, as follows. The
  • reaction mixtures contained 100 mM Tris-HCl, pH 7.5, 10 mM EDTA, 3 ⁇ DTT, 200 ⁇ S-adenosylmethionine (SAM) (absent in control samples, as indicated) 3 ⁇ ag of pMYCOl plasmid DNA (SEQ ID NO: 149)), isolated from E. coli and 20 ⁇ g of M. mycoides LC extracts.
  • SAM S-adenosylmethionine
  • methyltransferases 4 ⁇ of each sample was cleaved using restriction enzyme isoschizomers, purchased from New England Biolabs, appropriate for the particular sequence being evaluated, according to the manufacturer's instructions (Bed (recognition site, CCATC), Hmfl (GANTC), HpyAV (CCTTC), SfdNI (GCATC). Samples were run on 1% 48-well agarose E-gels (Invitrogen) at 70V for 25 minutes. Gels were scanned on a GE Typhoon 9410 imager. d. Evaluating GATC site methylation by M. mycoides LC extracts
  • M. mycoides LC extracts were as follows: M. mycoides LC extract was able to completely methylate Mycoplasma plasmid DNA in the case of 4 restriction-modification systems, and only partially in the case of 1 system (the GATC restriction-modification system) (gel results not shown). Because the endogenous M.
  • mycoides LC GATC methyltransferase showed low activity in the M. mycoides LC extract, the commercially available E. coli dam methyltransferase (New England Biolabs, Ipswich, MA) was used to successfully supplement its activity.
  • crude extracts protected donor plasmid DNA from host restriction- modification system and increased efficiency of transformation, crude extracts also completely inhibited genome transplantation experiments involving native M. mycoides LC genomic DNA isolated in agarose plugs and M. capricolum cells (See Example 3, below).
  • M. mycoides LC genomic DNA isolated in agarose plugs (prepared as described in Example 2B(ii), above), in the absence or presence of M. capricolum or M. mycoides LC Mycoplasma crude extracts (prepared as in Example 2E(ii)(a) above), was analyzed by fluorescence microscopy.
  • M. mycoides LC genomic DNA agarose plugs were melted with ⁇ -agarase I, as described above (Example 2C(ii)).
  • Figure 7A illustrates initial treatment of the agarose plugs and the result of untreated agarose plugs. Results of methylation and deproteinisation steps are presented in Figs. 7B-7C, respectively, in which samples and treatment are indicated above pictures.
  • proteinase K treatment restores the transplantation efficiency of native genomic DNA that had been treated with crude extracts.
  • removal of proteins in crude cell extracts e.g., by proteinase K treatment
  • M. mycoides LC donor genomic DNA isolated from yeast host cells could be methylated using crude extracts, and still be successfully transplanted into recipient cells.
  • the heterologous pSD4 Spiroplasma citri oriC plasmid (SEQ ID NO: 150) has been shown to transform M. capricolum (Lartigue, C. et al. Nucleic Acids Res. 31 :6610-8 (2003)). This plasmid is efficient for integration of foreign sequences and for targeted mutagenesis in the M. capricolum genome.
  • MCAP0050 potential restriction enzyme gene
  • the fragment was then cleaved by Xbal and cloned into an Xbal cleaved pSD4 plasmid (SEQ ID NO: 150), yielding pSD4-AMcap0050.1 (SEQ ID NO: 159).
  • Ten micrograms of this plasmid was used to transform wild type M.
  • capricolum cells using the 5% PEG mediated protocol, described in Example 2C, above.
  • Transformants were selected on solid SP4 plates containing 4 ⁇ g/mL tetracycline. After 7 days of incubation at 37° C, several colonies were picked in liquid medium containing 10 ⁇ g/ml tetracycline and cells were propagated for 15 passages (15P). Clones then were analyzed by Southern blot to verify the presence of the plasmid and its possible integration at the target gene by homologous recombination via a single crossing- over.
  • M. capricolum ARE clone 10 15P, was selected because (as evidenced by Southern blot) its hybridization pattern demonstrated the interruption of MCAP0050 gene by the plasmid. No free plasmid was detected. Centrifuge-clarified lysate extracts also were prepared as described in 2D(ii)(b), and used in the restriction enzyme assay described in Example 2D(ii). The results revealed a complete absence of restriction enzyme activity in clone 10 15P clone compared to wild type M. capricolum (data not shown).
  • M. capricolum ARE clone 10 was used as a source to make crude M. capricolum cellular extracts for in vitro
  • the M. capricolum ARE clone 10 was not, however, a good candidate recipient cell for transplantation of M. mycoides LC because it contained the same resistance marker gene (tetM) as the M. mycoides LC donor genomic DNA present in yeast. Accordingly, using the cloning strategy described above, another M. capricolum ARE mutant was constructed, which carried the YCp and puromycin resistance gene instead of the tetM gene. Centrifuge-clarified lysate extracts were prepared from the M. capricolum ARE clone 17.5 15P, as described in 2D(ii)(b), and used in the restriction enzyme assay described in Example 2D(ii). The results revealed a complete absence of restriction enzyme activity in clone 17.5 15P clone compared to wild type M. capricolum. Accordingly, this clone was chosen for transplantation.
  • tM resistance marker gene
  • Example 1 describes the successful cloning of complete bacterial genomes (including these Mycoplasma) in yeast host cells.
  • This example describes the successful transplantation of whole Mycoplasma donor genome (M mycoides LC (Genbank Accession No.: NZ AAZK00000000; GI: 149364883)) DNA, which had been propagated in yeast host cells, into Mycoplasma recipient cells of a different species ( . capricolum).
  • the donor and recipient were chosen based on their rapid growth. The method could also be performed, however, using other Mycoplasma donors and recipients, such as M. genitalium
  • the methods described in this Example can be used in conjunction with steps for modifying the donor genomes in vivo while in the host cell (e.g., using the repertoire of yeast tools during propagation of the bacterial donor genome) prior to transfer.
  • the methods can be used to genetically engineer donor genomes, such as genomes from bacteria with relatively poorly developed genetic systems.
  • the methods can be used to generate synthetic cells that contain synthetically engineered genomes.
  • a complete synthetic genome of non-pathogenic strain of Mycoplasma genitalium has been synthesized (Gibson et al, Science 319:1215-20 (2008); Gibson et al, PNAS USA 105:20404-9 (2008)).
  • the genome was assembled in the final step as a centromeric plasmid in yeast.
  • Such synthetically produced and natural genomes ⁇ e.g., Mycoplasma or other bacterial genomes) propagated in host cells ⁇ e.g., yeast host cells) can be transplanted into recipient cytoplasm to generate synthetic cells using the provided methods, such as those described in this Example.
  • host cells e.g., yeast host cells
  • the methods can be used to transplant it from yeast into a bacterial ⁇ e.g., Mycoplasma) recipient.
  • mycoides LC donor genomic DNA isolated from yeast, would be unmethylated and susceptible to the M. capricolum recipient cell restriction- modification system upon transfer into that cell. It was also possible that one or more M mycoides LC restriction enzymes could cleave the donor genome, once expressed following transplantation.
  • Example 2 Such aspects were selected and used in the following study to achieve successful transplant of donor Mycoplasma genomes, propagated in yeast, into Mycoplasma recipient cells of a different species.
  • FIG. 8 shows three alternative transplantation approaches that can be used to transplant whole genomic DNA. Variations on these three approaches were used in the examples, described below.
  • the first approach (denoted with the number "1 "labeling the arrows in Figure 8), includes digestion of agarose plugs containing the genomic DNA ⁇ e.g., with ⁇ -agarase (melting step)), followed by transplantation directly into recipient cells.
  • This method is typically used, as shown in Figure 8, for transplantation of donor genomes or nucleic acids from one cell ⁇ e.g., donor Mycoplasma cell) into a similar cell, such as from a Mycoplasma donor cell into a Mycoplasma recipient cell.
  • the second approach (denoted with the numbers "2") is identical to the first method, except that recipient cells had been modified to mutate the restriction enzyme genes (ARE; generated as described in Example 2F, above).
  • ARE restriction enzyme genes
  • genomic Mycoplasma DNA grown in yeast host cells and isolated in yeast plugs can successfully be transplanted into Mycoplasma recipient cells.
  • the third approach (denoted with the numbers "3" in Figure 8), samples were methylated and subjected to a deproteinisation step (treatment with proteinase K), prior to the melting step ( ⁇ -agarase digestion) and transplantation into recipient cells.
  • methylation and deproteinisation steps also facilitate efficient transplant of donor Mycoplasma genomes, isolated after propagation in yeast, into Mycoplasma recipient cells.
  • control studies included conditions similar to the third approach, where samples were treated in parallel, under the same conditions, without the presence of methylases ("mock
  • M. mycoides LC genomic DNA was isolated in agarose plugs from yeast strain VL6-48N (described by Larionov et al, PNAS USA 94:7384-7 (1997)), as follows.
  • Yeast cells containing the M. mycoides LC genomic DNA were grown at 30° C in selective medium until the OD reached
  • Agarose plugs were prepared from each cell type, using the CHEF mammalian Genomic DNA Plug Kit (Bio-Rad Laboratories, Valencia, CA), following the instructions recommended by the manufacturer for yeast (eukaryotic) DNA extraction, with the following modifications. 6x10 9 yeast cells were used per plug, (instead of 6x10 8 cells, as recommended by the kit), to yield 6xl0 8 cells per plug, in order to increase the amount of M. mycoides LC genomic DNA available per plug.
  • ZymolyaseTM 100T enzyme (USB Corporation, Cleveland, OH) was used instead of Lyticase (Bio-Rad Laboratories, Valencia, CA). The enzyme was added inside and outside of the plugs at a concentration of 5 mg/ml and let stand for 2 hours at 37 °C.
  • phenylmethanesulfonylfluoride was added during the second wash.
  • yeast genomic DNA isolated along with the M. mycoides LC donor genomic DNA extracted from yeast, would affect or abrogate transplantation reactions. Accordingly, with each approach, two sets of donor genomic DNA were prepared, one of which was submitted to the optional "clean-up” step after digestion of cell walls and proteinase K treatment. The "clean-up” step was designed to remove yeast genomic DNA from the samples.
  • the "clean-up" samples were either treated with a cocktail of restriction enzymes that specifically digests yeast genomic DNA and then cleared of small yeast DNA fragments by electrophoresis or directly loaded on Pulse Field agarose gel to separate circular genomes that are being caught into the well, and linear yeast chromosomes that are electrophoresed out of the well (Lartigue et al, Science 317, 632 (2007).
  • yeast plugs were washed two times, 1 hour each, in 1ml of 0.1 X TE buffer (2mM Tris-HCl pH 8.0 - 5mM EDTA), equilibrated lh in 1ml of IX NEB buffer 2 (New England Biolabs, Ipswich, MA ) supplemented with BSA, and the genomic DNA present in the plug was digested over-night with 50 units of restriction enzymes cocktail (AsiSI, RsrII and Fsel) in a 500 ⁇ 1 reaction volume.
  • restriction enzymes cocktail AsiSI, RsrII and Fsel
  • the yeast plugs were washed at room temperature for 1 hour with 1 mL of IX TE buffer (20mM Tris-HCl pH 8.0 - 50mM EDTA) and loaded on 1% TAE agarose gel (120 minutes, 120 volts). Agarose plugs were removed from the well and stored at 4 °C.
  • transplantation proceeded as follows, with the exception that with the third approach, samples were subject to methylation and proteinase K treatment prior to melting with ⁇ -agarase, as described in the Example 3 A(iii), below. a. Recipient cells
  • M. capricolum recipient cells wild-type or ARE (as produced in Example 2F) were grown in SOB(+) medium (Bacto SOB medium (Becton Dickinson, Franklin Lakes, NJ) supplemented with fetal bovine serum (17%), glucose (10 g/1), 2 ml of phenol Red (1%) and 100 ⁇ of Penicillin G (5 mg/ml)) until the pH of the culture reached pH 5.7 to 5.85 (approximately 5x 10 7 cells / ml).
  • SOB(+) medium Boacto SOB medium (Becton Dickinson, Franklin Lakes, NJ) supplemented with fetal bovine serum (17%), glucose (10 g/1), 2 ml of phenol Red (1%) and 100 ⁇ of Penicillin G (5 mg/ml)
  • the recipient cells were centrifuged at 4575xg for 15 minutes at 10° C, washed once in 6 mL S/T buffer (Tris-HCl lOmM pH 6.5; NaCl 250mM), resuspended in 400 ⁇ of CaCl 2 (0.1 M) and incubated on ice for 30 minutes.
  • S/T buffer Tris-HCl lOmM pH 6.5; NaCl 250mM
  • mycoides LC genomic DNA from yeast agarose plugs was methylated, followed by a deproteination step.
  • the plugs were washed two times 30 minutes in 200 mM Tris-HCl pH 7.5; 50 mM EDTA and equilibrated two times 30 minutes in methylation buffer (100 mM Tris-HCl pH 7.5; 10 mM EDTA, 3 ⁇ DTT, 200 ⁇ S-adenosylmethionine) with soft agitation.
  • methylation buffer 100 mM Tris-HCl pH 7.5; 10 mM EDTA, 3 ⁇ DTT, 200 ⁇ S-adenosylmethionine
  • Example 2E(ii) Each cell extract was prepared as described in Example 2E(ii), above and purified methyltransferases were prepared as described in Example 2E(i), above.
  • the methylation reactions containing M. mycoides LC cellular extracts or purified methyltransferases were also supplemented with 5 ⁇ of dam methyltransferase (New England Biolabs, Ipswich, MA).
  • each yeast plug was incubated for 4 hours at 50° C in 1 mL Proteinase K Reaction Buffer [100 mM EDTA; 0.2 % Sodium Deoxycholate; 1 % Sodium Lauryl Sarcosine; pH8.0] supplemented with 40 ⁇ of Proteinase K.
  • the plugs were then washed 4 times 45 minutes with 1 ml of IX TE buffer (20 mM Tris-HCl pH 8; 50 mM EDTA) and 2 times 30 minutes in 0.1X TE buffer with soft agitation at room temperature. After removing the final wash buffer, the plugs were melted with ⁇ -Agarase I as described in section (b), above, followed by transplantation.
  • IX TE buffer 20 mM Tris-HCl pH 8; 50 mM EDTA
  • Results were scored by selecting for growth of blue colonies on SP4 medium containing tetracycline at 37° C and are presented in Table 12A, below. As indicated in that table, for methylation with M. mycoides extracts and transplanted into wild-type cells (sample 2), some samples were cleaned-up to remove yeast DNA, as described above, either by digestion with yeast-specific enzymes (b) or by electrophoresis (c).
  • b Yeast plugs have been cleared of yeast genomic DNA using AsiSl, Rsrll, Fsel cocktail restriction enzymes protocol.
  • mycoides LC extracts or purified methylases transplanted into either wild-type or ARE recipient cells (samples 2, 3, 7, and 8), colonies were obtained that were phenotypically
  • colonies were obtained in both recipient cell types with donor genomes treated with the M. capricolum extract (samples 4 and 9) (32 and 9 colonies, Table 12A).
  • yeast DNA was present in samples containing genomic DNA. Purifying the donor genomic DNA away from yeast genomic DNA (denoted by “b” and “c” in Table 12 A, above) did not substantially alter transplantation results, demonstrating that the recipient M. capricolum cells were able to tolerate the presence of non-specific or carrier DNA (Table 12). In addition, positive transplantation results were obtained with donor genomic DNA isolated from two different yeast strains (VL6-48N and W303a), indicating that the genotype and/or phenotype of the host yeast strain may not be important for transplantation experiments.
  • YCpMmycl.l as well as the engineered YCp genomes (YCpMmycl.l-AtypeIIIres::URA3 and YCpMmycl.l-Atypelllres), were also isolated from yeast strain W303a. Transplantation of all three YCp genomes into M. capricolum recipient cells resulted in similar numbers of tetracyclineresistant blue colonies (Table 12B). The large deletion clone (YCpMmycl.l-A500kb) discussed above served as an appropriate control because it lacks many presumed essential genes yet retains the YCp element and tetM. As expected, no colonies were recovered when this genome was transplanted into M. capricolum recipient cells.
  • mycoides was done by Southern blot analysis using an M. mycoides—specific IS 1296 element as probe (data not shown). It was shown that the Type III restriction gene was deleted in the engineered bacterium by PCR by Southern blot analysis using the Type III restriction gene sequence as probe (data not shown), and by sequencing the locus ( Figure 17).
  • Wild-type M. capricolum and M. capricolum RE(-) transplantation was performed using methods described in Figure 8.
  • yeast plugs were digested with ⁇ -agarase (melting step) and transplanted into both recipient cells.
  • the treated samples were methylated and treated with proteinase K before the melting step.
  • the mock-methylated sample was treated the same as the methylated samples, except that no extract or purified methyltransferases were added.
  • VL6-48N yeast agarose plugs used in this experiment carried YCpMmycl .1.W303a yeast agarose plugs carried YCpMmycl.l, YCpMmycl.l that was engineered in yeast
  • the number of transplants is the average of at least three experiments.
  • the error reported is the absolute mean deviation.
  • This example describes the use of methods for introducing a seamless modification into a Mycoplasma donor genome that has been cloned into a yeast host cell.
  • yeast Saccharomyces cerevisiae has been developed as a host capable of cloning large DNA fragments, as both linear and circular yeast artificial chromosomes (YACs). Once cloned in yeast, these YACs can be manipulated using standard genetic tools. Transfer of this modified DNA to host cells suitable for expression allows the functional study of genes and their regulation. Cloning of whole bacterial genomes in yeast, and subsequent transplantation of such genomes back into their original cellular environments, has extended this application from the gene to the genome level.
  • YACs linear and circular yeast artificial chromosomes
  • Two-step recombination protocols utilizing counter-selectable markers can be used to modify YACs in yeast (Tucker and Burke (1996) Nucleic Acids Res, 24, 3467-3468).
  • a counter-selectable marker is first recombined into the YAC and selected for.
  • a new DNA fragment containing the desired alteration is recombined in place of the marker, which is selected against.
  • the most frequently used marker in these procedures is the URA3 gene, which restores Uracil autotrophy in deficient strains.
  • Counter-selection for the replacement of the URA3 gene is performed by treatment with 5-fluoroorotic acid (5- FOA) (Boeke et al, (1984) Mol Gen Genet, 197, 345-346).
  • 5- FOA 5-fluoroorotic acid
  • the method restores uracil auxotrophy, which can then be used again for a further round of modification.
  • This basic method for seamless modification has been improved in a number of ways.
  • the delitto perfetto method introduces a double-strand break (DSB) near the target locus by utilizing the endonuclease ⁇ -SceI (Storici et al. (2001) Nat Biotechnol, 19, 773-776).
  • tandem repeat pop-out creates tandem repeat sequences flanking the target site (Akada et al. (2006) Yeast, 23, 399-405). This method requires only one transformation followed by 5-FOA counter-selection, whereas other methods require two transformations. These methods can be adapted for deletions, point mutations, or gene replacement.
  • target locus containing a single-based cytidine deletion (309,388) in the CDS139 locus of the synthetic
  • sMgTARBAC37 M genitalium genome that had been introduced into and maintained in yeast host cells, as described by Gibson et al, Science 319, 1215 (2008) and U.S. Publication No. 20090275086, by Gibson et al, and as described in Example 1C, above.
  • Saccharomyces cerevisiae strains VL6-48N (MATa his3-A200 trpl - ⁇ 1 ura3- 52 lys2 ade2-101 metl4) and W303a (MATa ade2-l ura3-l his3-l l,15 trpl-1 leu2-3,112 canl-100 RAD5), carried the synthetic genome as described by Gibson et al, Science Id. and provided in Example 1, above.
  • Yeast cells were grown in standard rich medium (YEPD) and synthetic dextrose (SD) or synthetic galactose (SG) minimal medium (Amberg et al.
  • PCR constructs were introduced into the yeast strain containing the M. genitalium genome using lithium acetate integrative transformation according to a published method (Gietz et al. , Nucleic Acids Res, 20, 1425 (1992)), using 2-3 ⁇ g PCR product and 25 ⁇ g carrier DNA (Salmon testis DNA, Sigma).

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Mycology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne des compositions et des procédés destinés à cloner un génome donneur synthétique ou semi-synthétique dans une cellule hôte hétérologue. Dans un mode de réalisation, le génome donneur peut encore être modifié à l'intérieur d'une cellule hôte. Les génomes modifiés ou non peuvent encore être isolés à partir de la cellule hôte et transférés à une cellule réceptrice. Les procédés décrits ici peuvent être utilisés pour altérer les génomes donneurs à partir de cellules donatrices réfractaires dans plusieurs cellules hôtes modifiables.
EP10726723A 2010-03-05 2010-05-19 Procédés de clonage et de manipulation de génomes Withdrawn EP2542676A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12/718,911 US9273310B2 (en) 2007-10-08 2010-03-05 Methods for cloning and manipulating genomes
US32226910P 2010-04-08 2010-04-08
PCT/US2010/035490 WO2011109031A1 (fr) 2010-03-05 2010-05-19 Procédés de clonage et de manipulation de génomes

Publications (1)

Publication Number Publication Date
EP2542676A1 true EP2542676A1 (fr) 2013-01-09

Family

ID=42697547

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10726723A Withdrawn EP2542676A1 (fr) 2010-03-05 2010-05-19 Procédés de clonage et de manipulation de génomes

Country Status (4)

Country Link
EP (1) EP2542676A1 (fr)
JP (1) JP2013520989A (fr)
CN (1) CN102939380A (fr)
WO (1) WO2011109031A1 (fr)

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
EP3964285A1 (fr) 2011-09-26 2022-03-09 Thermo Fisher Scientific Geneart GmbH Synthèse d'acide nucléique de petit volume et de haute efficacité
WO2014153188A2 (fr) 2013-03-14 2014-09-25 Life Technologies Corporation Synthèse hautement efficace de petits volumes d'acides nucléiques
US9206435B2 (en) * 2012-08-31 2015-12-08 Synthetic Genomics, Inc. Crowding agent-induced nucleic acid transfer into a recipient host cell
SG10201800111SA (en) * 2013-07-09 2018-02-27 Harvard College Multiplex rna-guided genome engineering
KR102207770B1 (ko) 2013-08-05 2021-01-26 트위스트 바이오사이언스 코포레이션 드 노보 합성된 유전자 라이브러리
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
AU2014346559B2 (en) 2013-11-07 2020-07-09 Editas Medicine,Inc. CRISPR-related methods and compositions with governing gRNAs
US20150166984A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting alpha-antitrypsin point mutations
EP4079847A1 (fr) 2014-07-30 2022-10-26 President And Fellows Of Harvard College Protéines cas9 comprenant des intéines dépendant de ligands
LT3557262T (lt) 2014-12-09 2022-11-10 Life Technologies Corporation Didelio efektyvumo nukleorūgščių sintezė mažame tūryje
CA2975852A1 (fr) 2015-02-04 2016-08-11 Twist Bioscience Corporation Procedes et dispositifs pour assemblage de novo d'acide oligonucleique
WO2016172377A1 (fr) 2015-04-21 2016-10-27 Twist Bioscience Corporation Dispositifs et procédés pour la synthèse de banques d'acides oligonucléiques
KR20180050411A (ko) 2015-09-18 2018-05-14 트위스트 바이오사이언스 코포레이션 올리고핵산 변이체 라이브러리 및 그의 합성
CN108698012A (zh) 2015-09-22 2018-10-23 特韦斯特生物科学公司 用于核酸合成的柔性基底
IL294014B2 (en) 2015-10-23 2024-07-01 Harvard College Nucleobase editors and their uses
CN108603307A (zh) 2015-12-01 2018-09-28 特韦斯特生物科学公司 功能化表面及其制备
AU2017306676B2 (en) 2016-08-03 2024-02-22 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
EP3497214B1 (fr) 2016-08-09 2023-06-28 President and Fellows of Harvard College Protéines de fusion cas9-recombinase programmables et utilisations associées
KR102212257B1 (ko) 2016-08-22 2021-02-04 트위스트 바이오사이언스 코포레이션 드 노보 합성된 핵산 라이브러리
WO2018039438A1 (fr) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation d'acides aminés non naturels dans des protéines au moyen de l'édition de bases
KR102217487B1 (ko) 2016-09-21 2021-02-23 트위스트 바이오사이언스 코포레이션 핵산 기반 데이터 저장
JP2019530464A (ja) 2016-10-14 2019-10-24 プレジデント アンド フェローズ オブ ハーバード カレッジ 核酸塩基エディターのaav送達
GB2573069A (en) 2016-12-16 2019-10-23 Twist Bioscience Corp Variant libraries of the immunological synapse and synthesis thereof
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
KR20190119107A (ko) 2017-02-22 2019-10-21 트위스트 바이오사이언스 코포레이션 핵산 기반 데이터 저장
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
EP3595674A4 (fr) 2017-03-15 2020-12-16 Twist Bioscience Corporation Banques de variants de la synapse immunologique et leur synthèse
BR112019019300B1 (pt) * 2017-03-17 2023-04-18 Syngenta Participations Ag Método para preservar uma amostra de tecido vegetal contendo ácidos nucleicos para uso em análise de dna, reagente para o processamento de uma amostra biológica contendo ácidos nucleicos, kit para armazenamento de amostras de tecido contendo ácidos nucleicos e métodos para extrair ácidos nucleicos e para a realização de genotipagem (ou qpcr) em ácidos nucleicos preservados
WO2018176009A1 (fr) 2017-03-23 2018-09-27 President And Fellows Of Harvard College Éditeurs de nucléobase comprenant des protéines de liaison à l'adn programmable par acides nucléiques
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
AU2018284227B2 (en) 2017-06-12 2024-05-02 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
WO2018231864A1 (fr) 2017-06-12 2018-12-20 Twist Bioscience Corporation Méthodes d'assemblage d'acides nucléiques continus
EP3658573A1 (fr) 2017-07-28 2020-06-03 President and Fellows of Harvard College Procédés et compositions pour l'évolution d'éditeurs de bases à l'aide d'une évolution continue assistée par phage (pace)
WO2019139645A2 (fr) 2017-08-30 2019-07-18 President And Fellows Of Harvard College Éditeurs de bases à haut rendement comprenant une gam
SG11202002194UA (en) 2017-09-11 2020-04-29 Twist Bioscience Corp Gpcr binding proteins and synthesis thereof
AU2018352592A1 (en) 2017-10-16 2020-06-04 Beam Therapeutics, Inc. Uses of adenosine base editors
CN107784199A (zh) * 2017-10-18 2018-03-09 中国科学院昆明植物研究所 一种基于总dna测序结果的细胞器基因组筛选方法
EP3697529B1 (fr) 2017-10-20 2023-05-24 Twist Bioscience Corporation Nano-puits chauffés pour la synthèse de polynucléotides
SG11202006460SA (en) 2018-01-04 2020-08-28 Twist Bioscience Corp Dna-based digital information storage
CA3100739A1 (fr) 2018-05-18 2019-11-21 Twist Bioscience Corporation Polynucleotides, reactifs, et procedes d'hybridation d'acides nucleiques
SG11202109283UA (en) 2019-02-26 2021-09-29 Twist Bioscience Corp Variant nucleic acid libraries for antibody optimization
KR20210143766A (ko) 2019-02-26 2021-11-29 트위스트 바이오사이언스 코포레이션 Glp1 수용체에 대한 변이체 핵산 라이브러리
EP3942040A1 (fr) 2019-03-19 2022-01-26 The Broad Institute, Inc. Procédés et compositions pour l'édition de séquences nucléotidiques
CN114729342A (zh) 2019-06-21 2022-07-08 特韦斯特生物科学公司 基于条形码的核酸序列装配
MX2022013513A (es) 2020-04-27 2023-01-16 Twist Bioscience Corp Bibliotecas de ácidos nucleicos variantes para coronavirus.
MX2022014008A (es) 2020-05-08 2023-02-09 Broad Inst Inc Métodos y composiciones para la edición simultánea de ambas cadenas de una secuencia de nucleótidos de doble cadena objetivo.
US11970697B2 (en) 2020-10-19 2024-04-30 Twist Bioscience Corporation Methods of synthesizing oligonucleotides using tethered nucleotides
CN116042688B (zh) * 2023-03-06 2023-06-20 中山大学 柑橘溃疡病菌高饱和转座子突变体库及构建方法
CN116769797B (zh) * 2023-08-14 2023-10-24 浙江大学海南研究院 一种茉莉酸甲酯及PpyMYC2基因在萌芽中的应用

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5721118A (en) 1995-10-31 1998-02-24 The Regents Of The University Of California, San Diego Mammalian artificial chromosomes and methods of using same
US5866404A (en) 1995-12-06 1999-02-02 Yale University Yeast-bacteria shuttle vector
US6025155A (en) 1996-04-10 2000-02-15 Chromos Molecular Systems, Inc. Artificial chromosomes, uses thereof and methods for preparing artificial chromosomes
WO2000006715A1 (fr) 1998-07-27 2000-02-10 Genotypes Inc. Vecteur de chromosome artificiel automatique d'eucariote
US7083971B1 (en) 1999-06-07 2006-08-01 Cell Genesys, Inc. Hybrid yeast-bacteria cloning system and uses thereof
US6692954B1 (en) 2000-11-03 2004-02-17 The Scripps Research Institute Generation of human cytomegalovirus yeast artificial chromosome recombinants
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
EP1915446B1 (fr) 2005-08-11 2017-06-14 Synthetic Genomics, Inc. Methode de recombinaison in vitro
US10041060B2 (en) * 2005-12-06 2018-08-07 Synthetic Genomics, Inc. Method of nucleic acid cassette assembly
WO2008016380A2 (fr) * 2005-12-23 2008-02-07 J. Craig Venter Institute Installation de génomes ou de génomes partiels dans des cellules ou des systèmes de type cellules
AU2008254291B2 (en) * 2007-05-01 2015-09-24 Synthetic Genomics, Inc. Methods of genome installation in a recipient host cell
JP5618413B2 (ja) 2007-10-08 2014-11-05 シンセティック ゲノミクス、インク. 大型核酸のアッセンブリー
WO2009134814A2 (fr) * 2008-04-28 2009-11-05 Synthetic Genomics, Inc. Identification de séquences centromères et leurs utilisations
CA2754212C (fr) * 2009-03-06 2016-08-02 Synthetic Genomics, Inc. Procedes de clonage et de manipulation de genomes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2011109031A1 *

Also Published As

Publication number Publication date
WO2011109031A8 (fr) 2012-09-20
WO2011109031A1 (fr) 2011-09-09
JP2013520989A (ja) 2013-06-10
CN102939380A (zh) 2013-02-20

Similar Documents

Publication Publication Date Title
JP6637140B2 (ja) ゲノムのクローニングおよび操作のための方法
EP2542676A1 (fr) Procédés de clonage et de manipulation de génomes
US10975378B2 (en) Methods for cloning and manipulating genomes
JP5618413B2 (ja) 大型核酸のアッセンブリー
US20210254046A1 (en) Generation of synthetic genomes
EP2890787B1 (fr) Transfert d'acide nucléique induit par un agent de rassemblement dans une cellule hôte receveuse
Herbst Scalable approaches for gene tagging and genome walking sequencing

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120926

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20151218

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20160629