WO2003091413A2 - Expression of polypeptides in chloroplasts, and compositions and methods for expressing same - Google Patents
Expression of polypeptides in chloroplasts, and compositions and methods for expressing same Download PDFInfo
- Publication number
- WO2003091413A2 WO2003091413A2 PCT/US2003/012997 US0312997W WO03091413A2 WO 2003091413 A2 WO2003091413 A2 WO 2003091413A2 US 0312997 W US0312997 W US 0312997W WO 03091413 A2 WO03091413 A2 WO 03091413A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- polynucleotide
- polypeptide
- rbs
- chloroplast
- nucleotide sequence
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/82—Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/82—Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
- C12N15/8201—Methods for introducing genetic material into plant cells, e.g. DNA, RNA, stable or transient incorporation, tissue culture methods adapted for transformation
- C12N15/8214—Plastid transformation
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/82—Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
- C12N15/8201—Methods for introducing genetic material into plant cells, e.g. DNA, RNA, stable or transient incorporation, tissue culture methods adapted for transformation
- C12N15/8209—Selection, visualisation of transformants, reporter constructs, e.g. antibiotic resistance markers
- C12N15/821—Non-antibiotic resistance markers, e.g. morphogenetic, metabolic markers
- C12N15/8212—Colour markers, e.g. beta-glucoronidase [GUS], green fluorescent protein [GFP], carotenoid
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/82—Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
- C12N15/8241—Phenotypically and genetically modified plants via recombinant DNA technology
- C12N15/8242—Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits
- C12N15/8257—Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits for the production of primary gene products, e.g. pharmaceutical products, interferon
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/82—Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
- C12N15/8241—Phenotypically and genetically modified plants via recombinant DNA technology
- C12N15/8242—Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits
- C12N15/8257—Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits for the production of primary gene products, e.g. pharmaceutical products, interferon
- C12N15/8258—Phenotypically and genetically modified plants via recombinant DNA technology with non-agronomic quality (output) traits, e.g. for industrial processing; Value added, non-agronomic traits for the production of primary gene products, e.g. pharmaceutical products, interferon for the production of oral vaccines (antigens) or immunoglobulins
Definitions
- the present invention relates generally to compositions and methods for expressing polypeptides in plant cell chloroplasts, and more specifically to chloroplast codon biased polynucleotides encoding heterologous polypeptides, to expression vectors that allow robust expression of heterologous polypeptides in bacteria and in chloroplasts, including, for example, of protein complexes such as antibodies and antibody chimeras that are formed by a specific association of polypeptide subunits.
- a primary advantage of using genetic engineering techniques for producing therapeutic biological agents is that the methods allow for the generation of large amounts of a desired protein.
- the only other way to obtain sufficient quantities of the biological material, for example, for use as a therapeutic agent is by purifying the naturally occurring biological material from cells of an organism that produce the agent.
- growth hormone could only be obtained by isolating it from the pituitary gland of animals such as cattle.
- Insulin is another example of a biological agent that, prior to genetic engineering, was available in a sufficient amount and in a biologically active form only by isolating it from the pancreas of animals such as pigs.
- Recombinant proteins also can be produced in eukaryotic cells, including, for example, insect cells and mammalian cells, which may provide the necessary environment and accessory factors required to process an expressed protein into a biologically active agent.
- eukaryotic cells including, for example, insect cells and mammalian cells, which may provide the necessary environment and accessory factors required to process an expressed protein into a biologically active agent.
- antibodies contain a heavy chain and a light chain that form a dimer with each other, and further associates with a second heavy chain and light chain dimer to form an active antibody.
- Such a process can occur in eukaryotic cells such as mammalian cells.
- eukaryotic cells also can modify a protein, for example, by glycosylating the protein such that it contains sugar groups at specific positions.
- glycosyl groups can be strongly antigenic and, upon administration to an individual, can result in the stimulation of an immune response that can inactivate the recombinant protein and, in some cases, can produce deleterious effects that cause more harm to the individual than the condition for which the recombinant protein originally was administered.
- a polynucleotide encoding a polypeptide that is to be produced using recombinant DNA methods is contained in a vector, which is a nucleic acid molecule that facilitates manipulation of the polynucleotide.
- Vectors can be used for introducing a polynucleotide of interest in prokaryotic cells such as bacteria or into eukaryotic cells such as mammalian cells.
- the vector also contains regulatory elements that allow, for example, amplification of the vector in the host cell.
- vectors have been designed that allow passage in both prokaryotic and eukaryotic cells.
- Such shuttle vectors can be useful because they allow, for example, generation of large amounts of the vector (and polynucleotide contained therein) in bacteria, then the vectors can be transferred to mammalian cells such that the encoded polypeptide can be produced under conditions that allow for proper assembly of a biologically active protein.
- shuttle vectors provide advantages over vectors that are specific for one or a few specific cell types, they do not obviate the potential problems that may be caused by post-translational modifications such as glycosylation, which can occur in eukaryotic cells.
- post-translational modifications such as glycosylation
- glycosylation which can occur in eukaryotic cells.
- the present invention satisfies this need and provides additional advantages.
- the present invention is based, in part, on a determination that heterologous polypeptides can be expressed robustly in plants by modifying the nucleotide sequence encoding the polypeptide such that it reflects chloroplast codon usage. Accordingly, the present invention relates to a synthetic polynucleotide, which includes at least a first nucleotide sequence encoding at least a first polypeptide, wherein at least one codon in the first nucleotide sequence is biased to reflect chloroplast codon usage. In one embodiment, each codon in the first nucleotide sequence is biased to reflect chloroplast codon usage.
- the synthetic polynucleotide can contain a single nucleotide sequence encoding a single polypeptide, or can further include at least a second nucleotide sequence encoding a second polypeptide, wherein one or more of the codons of the second nucleotide sequence also can be biased to reflect chloroplast codon usage.
- the synthetic polynucleotide encodes two or more polypeptides
- the encoding nucleotide sequences can be operatively linked such that a single polynucleotide is transcribed therefrom, and the encoded polypeptides can be expressed separately or can be further operatively linked such that a fusion protein comprising the first polypeptide and the second polypeptide can be expressed.
- a first and second nucleotide sequence are operatively linked via a third nucleotide sequence, which, for example, can encode a linker peptide.
- a fusion protein comprising the first polypeptide linked via the linker peptide to the second polypeptide can be expressed from the synthetic polynucleotide.
- the polypeptide(s) encoded by a synthetic polynucleotide of the invention can be any polypeptide of interest, and generally is a polypeptide that is not normally expressed in a plastid, particularly a chloroplast.
- the encoded polypeptide(s) can be an one or more chains of an immunoglobulin (Ig) family member, e.g., Ig variable region, an Ig constant region, an Ig heavy chain, an Ig light chain, or a combination thereof; or a T cell receptor (TCR) ⁇ chain, TCR ⁇ chain, or combination thereof; or any soluble receptor such as soluble forms of a T cell receptor or fusions of such receptors with, for example, an IG heavy chain.
- Ig immunoglobulin family member
- TCR T cell receptor
- the synthetic polynucleotide encodes an Ig family member fusion protein, for example, a single chain antibody comprising a complete heavy chain operatively linked to a light chain variable region.
- a fusion protein is exemplified herein by a single chain anti-herpes simplex virus (HSV) antibody having an amino acid sequence as set forth in SEQ ID NO: 16, which can be encoded by the synthetic polynucleotide having a nucleotide sequence as forth in SEQ ID NO: 15, which is biased to reflect chloroplast codon usage.
- HSV single chain anti-herpes simplex virus
- a fusion protein encoded by a synthetic polynucleotide that is biased to reflect chloroplast codon usage is exemplified by the single chain anti-HSV Fv fragment having an amino acid sequence as set forth in SEQ ID NO:43, which is encoded by SEQ ID NO:42.
- an fusion protein encoded by a synthetic polynucleotide that is biased to reflect chloroplast codon usage is exemplified by the HSV8-lsc (large single chain) antibody having an amino acid sequence as set forth in SEQ ID NO:48, which is encoded by SEQ ID NO :48.
- a polypeptide encoded by a synthetic polynucleotide of the invention also can be a reporter polypeptide, for example, a luciferase polypeptide.
- a reporter polypeptide for example, a luciferase polypeptide.
- a luciferase reporter polypeptide is exemplified herein by the luciferase fusion protein comprising the bacterial luciferase A subunit operatively linked via a linker peptide to the bacterial luciferase B subunit, the fusion protein having an amino acid sequence as set forth in SEQ ID NO:46, which can be encoded by the synthetic polynucleotide having a nucleotide sequence as set forth in SEQ ID NO:45, which is biased to reflect chloroplast codon usage.
- a luciferase fusion polypeptide having an amino acid sequence as set forth in SEQ ID NO:46 is provided.
- a synthetic chloroplast codon biased polynucleotide encoding a reporter polypeptide such as the exemplified polynucleotide (SEQ ID NO:45) encoding a fusion bacterial luxAB polypeptide (SEQ ID NO:46) can be useful, for example, as a tool to identify chloroplast promoters, 5' untranslated regions (5' UTRs), 3' UTR, protease deficient strains, and the like, thus providing a means to obtain further improved expression of a heterologous polypeptide in a chloroplast.
- the present invention also relates to a method of producing a heterologous polypeptide in a plastid by introducing a synthetic polynucleotide that includes at least a first nucleotide sequence encoding at least a first polypeptide, wherein at least one codon in the first nucleotide sequence is biased to reflect chloroplast codon usage, into the plastid under conditions that allow expression of the at least first polypeptide in the plastid.
- the synthetic polynucleotide can be operatively linked to a nucleic acid sequence encoding at least one ribosome binding sequence (RBS), particularly an RBS that can direct translation of the polypeptide in a plastid.
- RBS ribosome binding sequence
- the synthetic polynucleotide used according to a method of the invention can be any synthetic polynucleotide comprising at least a first nucleotide sequence containing at least one codon that is biased to reflect chloroplast codon usage.
- the synthetic polynucleotide can further include at least a second nucleotide sequence encoding a second polypeptide, wherein the first nucleotide sequence can, but need not, be operatively linked to the second nucleotide sequence, and wherein the second polypeptide can, but need not, be heterologous to the chloroplast.
- the synthetic polynucleotide encodes two (or more) polypeptides
- the encoded polypeptides can be expressed as separate and distinct polypeptides, or as a fusion protein comprising the first and second (or more) polypeptides.
- a fusion protein expressed from a synthetic polynucleotide according to a method of the invention comprises a first polypeptide linked via a linker peptide to a second polypeptide.
- a method is exemplified herein by expressing a single chain antibody comprising an IgA heavy chain linked to a light chain variable region, the fusion protein having an amino acid sequence as set forth in SEQ ID NO: 16, and encoded by a nucleotide sequence as set forth in SEQ ID NO: 15, which is biased with respect to chloroplast codon usage, wherein the expressed single chain antibody maintains antigen binding specificity (see, also, single chain anti-HSV Fv fragment having an amino acid sequence as set forth in SEQ ID NO:43 (encoded by SEQ ID NO:42), and HSV8-lsc antibody having an amino acid sequence as set forth in SEQ ID NO:48 (encoded by SEQ ID NO.48).
- a method of the invention is further exemplified herein by expressing a reporter polypeptide, particularly a luciferase fusion protein comprising the luciferase A subunit operatively linked to the luciferase B subunit, the fusion protein having an amino acid sequence as set forth in SEQ ID NO:46, and encoded by a nucleotide sequence as set forth in SEQ ID NO:45, wherein expression of the heterologous luciferase in chloroplasts is detectable in vivo or in vitro.
- a reporter polypeptide particularly a luciferase fusion protein comprising the luciferase A subunit operatively linked to the luciferase B subunit, the fusion protein having an amino acid sequence as set forth in SEQ ID NO:46, and encoded by a nucleotide sequence as set forth in SEQ ID NO:45, wherein expression of the heterologous luciferase in chloroplasts is detectable in viv
- a method of the invention can be practiced in any plastid, including in plant chloroplasts.
- the plant containing the chloroplasts can be any plant that naturally contains chloroplasts, including alga (microalga or macroalga) and higher plants.
- the method can further include a step of isolating the expressed heterologous polypeptide from plant cells (or isolated chloroplasts) containing the polypeptide. Accordingly, the invention provides a heterologous polypeptide produced by the method of the invention.
- the present invention further relates to a method of detecting a plant cell that contains a plastid.
- a method can be performed, for example, by introducing a synthetic polynucleotide of the invention, wherein the polynucleotide encodes a reporter polypeptide, into a plastid, e.g., a chloroplast, of the plant cell under conditions that allow expression of the reporter polypeptide in the chloroplast, and detecting expression of the reporter polypeptide.
- the reporter polypeptide can be any polypeptide as desired, and is exemplified herein by expressing a luciferase fusion protein having an amino acid sequence as set forth in SEQ ID NO:46.
- the present invention also relates to a method of producing a polypeptide in a plastid.
- a method can be performed, for example, by introducing at least a first recombinant nucleic acid molecule into the plastid, wherein the first recombinant nucleic acid molecule includes a first nucleotide sequence encoding at least one ribosome binding sequence (RBS) operatively linked to at least one heterologous polynucleotide encoding at least one polypeptide, and wherein the RBS can direct translation of the polypeptide in a plastid, under conditions that allow expression of the at least one polypeptide, thereby producing the polypeptide in the plastid.
- the plastic can be any plastid, including, for example, a chloroplast.
- one or more codons of the first polynucleotide can be biased to reflect chloroplast codon usage.
- the encoded polypeptide is an antibody, or a subunit of an antibody.
- the first polynucleotide encodes a first polypeptide and a second polypeptide, for example, a first polypeptide comprising an Ig heavy chain or a variable region thereof, and a second polypeptide comprises an Ig light chain or a variable region thereof.
- Such an antibody expressed according to a method of the invention is exemplified by an anti-tetanus toxin antibody having an amino acid sequence as set forth in SEQ ID NO: 14, which is encoded by the nucleotide sequences as set forth in SEQ ID NO: 13.
- the first polynucleotide is biased for chloroplast codon usage.
- Such antibodies expressed according to a method of the invention are exemplified by an anti-HSV antibody having an amino acid sequence as set forth in each of SEQ ID NO:16, SEQ ID NO:43, and SEQ ID NO:48, such antibodies being encoded, for example, by the nucleotide sequences as set forth in SEQ ID NO:15, SEQ ID NO:42, SEQ ID NO:47, respectively.
- the first polynucleotide encodes a first polypeptide and at least a second polypeptide, wherein the first and second (or more) polypeptides can, but need not, be subunits of a protein complex, for example, a heterodimer, heterotrimer, etc.
- the method can further include introducing at least a second recombinant nucleic acid molecule into the plastid.
- Such a second recombinant nucleic acid molecule can include a first nucleotide sequence encoding at least a first RBS operatively linked to at least a second heterologous polypeptide encoding at least a second polypeptide, wherein the first RBS can direct translation of the polypeptide in a plastid, particularly a chloroplast.
- the first recombinant nucleic acid molecule and the second recombinant nucleic acid molecule are co-expressed in the plastid.
- the first recombinant nucleic acid molecule can be contained in a vector.
- the vector is a chloroplast vector, which comprises a nucleotide sequence of chloroplast genomic DNA that can undergo homologous recombination with chloroplast genomic DNA, and the vector containing the first recombinant nucleic acid molecule is introduced into a chloroplast.
- a vector can further contain a prokaryote origin of replication.
- a method of the invention can further include isolating the polypeptide from the plastid. Accordingly, the invention provides an isolated polypeptide obtained by such a method, for example, an isolated antibody that is expressed in and heterologous with respect to a chloroplast.
- the present invention further relates to method of producing one or more polypeptides in a plant chloroplast, including methods of producing polypeptides that specifically associate to form a protein complex.
- a method of the invention provides a means to produce functional protein complexes, for example, a bivalent antibody comprising a first heavy and light chain associated with a second heavy and light chain.
- a method of the invention can be performed, for example, by introducing a first recombinant nucleic acid molecule into a chloroplast, which includes a first polynucleotide encoding at least one polypeptide; operatively linked to a second polynucleotide, which comprises a nucleotide sequence encoding a first ribosome binding sequence (RBS) operatively linked to a nucleotide sequence encoding a second RBS, wherein the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast, under conditions that allow expression of the at least one polypeptide, thereby producing the polypeptide in the chloroplast.
- the methods of the invention can be performed using any plant (or plant cell) that contains chloroplasts, including unicellular plants and algae and multicellular plants and algae.
- the first polynucleotide used in a method of the invention encodes a first polypeptide and at least a second polypeptide, for example, a first polypeptide and a second polypeptide; or a first polypeptide, a second polypeptide, and a third polypeptide; etc., any or all of which can be the same or different.
- one or more codons of the first polynucleotide are biased to reflect chloroplast codon usage.
- polypeptides expressed in plant chloroplasts such as chloroplasts of the microalga Chlamydomonas reinhardtii assemble properly, and can associate with one or more other expressed polypeptides in the chloroplast to form a functional protein complex.
- a first polynucleotide useful in a method of the invention can encode one or more polypeptide subunits that can associate to form a functional protein complex.
- the protein complex can be a dimer, trimer, tetramer, or the like, and the subunits can be the same or different or a combination thereof.
- the protein complex is a dimer, it can be a homodimer or a heterodimer.
- the protein complex is a trimer, it can be a homotrimer, a heterotrimer, or a trimer consisting of two identical polypeptides and one different polypeptide.
- a method of the invention is particularly useful for producing functional protein complexes such as antibodies, which generally occur naturally as a complex containing two heavy chains and two light chains, cell surface receptors such as T cell receptors, growth factor receptors, hormone receptors, G-protein coupled receptors, which can associate with a G-protein, and the like.
- An advantage of using a method of the invention to produce proteins such as antibodies in a chloroplast is that the polypeptides are not glycosylated following expression in chloroplasts and, therefore, have a greatly reduced antigenicity as compared to antibodies raised in an animal or expressed in the cytoplasm of a eukaryotic cell.
- a method of producing a functional protein complex in a chloroplast can be performed using a first recombinant nucleic acid molecule, as defined, wherein the first polynucleotide encodes the two or more subunits of the complex; or using a first recombinant nucleic acid molecule, as defined, which encodes one polypeptide subunit of the complex, and a second recombinant nucleic acid molecule, which has the same defined characteristics as the first recombinant nucleic acid molecule, and which encodes an additional polypeptide subunit of the protein complex.
- a method of the invention can be practiced using a first recombinant nucleic acid molecule, wherein the first polynucleotide encodes a first polypeptide, which is an immunoglobulin heavy chain (H) or a variable region thereof, and a second polypeptide, which is an immunoglobulin light chain (L) or a variable region thereof.
- a nucleotide sequence encoding an internal ribosome entry site can be positioned between the nucleotide sequences encoding the H and L chains such that expression of the second (downstream) encoded polypeptide is facilitated.
- a H chain Upon translation of the encoded H and L chains in the chloroplast, a H chain can associate with a L chain to form a monovalent antibody (i.e., an H:L complex), and two H:L complexes can further associate to produce a bivalent antibody.
- a monovalent antibody i.e., an H:L complex
- a method of the invention also can be practiced by introducing into a plant chloroplast a first recombinant nucleic acid molecule, wherein the first polynucleotide encodes, for example, a H chain or a variable region thereof, and further introducing into the chloroplast a second recombinant nucleic acid molecule, which comprises a first polynucleotide encoding a L chain or a variable region thereof, operatively linked to a second polynucleotide that includes a nucleotide sequence encoding a first RBS operatively linked to a nucleotide sequence encoding a second RBS, wherein the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast, under conditions such that the encoded polypeptides are substantially co-expressed in the chloroplasts, wherein the heavy chains (H) and light chains (L) can associate
- the first recombinant nucleic acid molecule can be contained in a vector. Furthermore, where the method is performed using a second (or more) other recombinant nucleic acid molecules, the second recombinant nucleic acid molecule also can be contained in a vector, which can, but need not, be the same vector as that containing the first recombinant nucleic acid molecule.
- a plant cell can be genetically modified such that chloroplasts in the plant contain a stably integrated recombinant nucleic acid molecule encoding a subunit of a protein complex
- the method of the invention can comprise introducing, for example, a vector comprising a second recombinant nucleic acid molecule, which encodes one or more other subunits of the protein complex, into chloroplasts of the plant such that, upon expression, a functional protein complex is produced.
- a vector useful in a method of the invention can be any vector useful for introducing a polynucleotide into a chloroplast.
- the vector can include a nucleotide sequence of chloroplast genomic DNA sufficient to undergo homologous recombination with chloroplast genomic DNA.
- Such a chloroplast vector can contain any additional nucleotide sequence that facilitates use or manipulation of the vector, for example, one or more transcriptional regulatory elements, or selectable markers, or cloning sites, or the like, including combinations thereof.
- the vector which can be a chloroplast vector, includes a transcriptional promoter and a 5'-untranslated region (5'UTR) of a plant chloroplast gene, which further contains, or can be modified to contain, a first RBS operatively linked to a second RBS, as defined herein.
- the vector which can be a chloroplast vector, includes a prokaryote origin of replication (ori), for example, an E. coli ori, thus providing a shuttle vector that can be passaged and manipulated in a prokaryote host cell as well as in a chloroplast.
- ori prokaryote origin of replication
- a shuttle vector of the invention can contain any polynucleotide of interest, including a synthetic chloroplast codon biased polynucleotide, for example, a synthetic polynucleotide such as SEQ ID NO:45, which encodes a bacterial luxAB fusion protein (SEQ ID NO:46).
- a shuttle vector expressing SEQ ID NO:46 provides the advantage that regulatory elements or other sequences of interest can be examined for expression in bacteria, then vectors containing those elements have desirable expression characteristics can be shuttled, with the same or other synthetic or other polynucleotide operatively linked thereto, to chloroplasts, wherein improved expression of an encoded heterologous polypeptide can be obtained.
- a method of the invention can further include a step of isolating an expressed polypeptide or protein complex from the chloroplast.
- the present invention also provides an isolated polypeptide or protein complex produced by a method as disclosed herein.
- the present invention provides isolated antibodies, which are expressed in and obtained from a plant chloroplast.
- An advantage of an isolated antibody of the invention is that the polypeptide components of the antibody are not glycosylated and, therefore, the antibody has reduced antigenicity when administered to a individual.
- such an antibody of the invention can have reduced effector activities characteristic of a naturally occurring antibody, for example, complement fixation activity, thus providing antibodies that can be useful for diagnostic purposes in an individual.
- the present invention also relates to an isolated ribonucleotide sequence that includes a first ribosome binding sequence (RBS) operatively linked to a second RBS, wherein the first RBS and second RBS are spaced apart by about 5 to 25 nucleotides, and wherein, when the ribonucleotide sequence is operatively linked to a polynucleotide encoding a polypeptide, the first RBS directs translation of the polypeptide in a prokaryote and the second RBS directs translation of the polypeptide in a chloroplast.
- RBS ribosome binding sequence
- An isolated ribonucleotide sequence of the invention which generally is about 11 to 50 ribonucleotides in length, and can be about 15 to 40 ribonucleotides in length, or about 20 to 30 ribonucleotides, can be a discrete unit, or can be operatively linked to a heterologous RNA molecule.
- the first RBS and second RBS which are operatively linked in a ribonucleotide sequence of the invention, generally are spaced apart by about 5 to 25 nucleotides, and usually by about 10 to 20 nucleotides, for example, by about 15 nucleotides.
- Each of the first RBS and the second RBS independently can consist of about 3 to 9 nucleotides, usually about 4 to 7 nucleotides, and can have any sequence characteristic of a Shine- Delgarno (SD) sequence, for example, a sequence comprising 5'-GGAG-3', which is complementary to a portion of a 16S rRNA anti-SD sequence.
- SD Shine- Delgarno
- the second RBS which directs translation in a chloroplast, can be contained within a 5' UTR of a chloroplast gene, which can be a chloroplast gene encoding a soluble chloroplast protein or a membrane bound chloroplast protein, wherein the 5' UTR can further include transcriptional regulatory elements, including a promoter.
- a ribonucleotide sequence of the invention can further include an initiator AUG codon operatively linked to the first and second RBS.
- Such an initiator AUG codon can further include adjacent nucleotides of a Kozak sequence, for example, ACCAUGG, which can facilitate translation of a polypeptide in a cell.
- a ribonucleotide sequence of the invention also can be operatively linked to a polyribonucleotide encoding a polypeptide, which can contain an endogenous initiator AUG codon or can be modified to contain an initiator AUG codon, or can lack an initiator AUG codon, which can be a component of the ribonucleotide sequence of the invention.
- An isolated ribonucleotide sequence of the invention can be chemically synthesized, or can be generated using an enzymatic method, for example, from a deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) template using a DNA dependent RNA polymerase or an RNA dependent RNA polymerase, respectively.
- DNA deoxyribonucleic acid
- RNA ribonucleic acid
- Such a DNA template can be chemically synthesized, or can be isolated from a naturally occurring DNA molecule, or can be based on naturally occurring DNA sequence that is modified to have the required characteristics, for example, a DNA sequence of a prokaryote gene that has nucleotide sequence encoding an RBS positioned about 5 to 15 nucleotides upstream an initiator ATG codon, and that is further modified to contain a second RBS, which is upstream of and spaced apart from the first RBS such that the second RBS can direct translation in a chloroplast.
- the present invention also relates to a polynucleotide encoding a first RBS operatively linked to a second RBS, as defined herein.
- the polynucleotide can be DNA or RNA, and can be single stranded or double stranded.
- a polynucleotide of the invention can include an initiator ATG codon operatively linked to the nucleotide sequence encoding the first RBS and second RBS.
- a polynucleotide of the invention can include a cloning site that is positioned to allow operative linkage of an expressible polynucleotide, which can encode a polypeptide, to the first RBS and second RBS, such that the polypeptide can be expressed in a chloroplast or in a prokaryote host cell.
- the cloning site can be any nucleotide sequence that facilitates insertion or linkage of the expressible polynucleotide to the first and second RBS such that translation of an encoded polypeptide can be initiated from the first RBS and the second RBS under suitable conditions, for example, one or more restriction endonuclease recognition sites or recombinase recognition sites or a combination thereof.
- a polynucleotide encoding a first and second RBS, as defined herein, can be operatively linked to an expressible polynucleotide, which can encode at least one polypeptide, including a peptide or peptide portion of a polypeptide.
- the expressible polynucleotide can encode a first polypeptide and one or more additional polypeptides, which can be the same or different.
- the expressible polynucleotide can encode a first polypeptide and a second polypeptide, which are different from each other.
- first and second polypeptide can be expressed as a fusion protein, or can be expressed as separate polypeptides, in which case a nucleotide sequence encoding an internal ribosome entry site can, but need not, be operatively linked between the coding sequence of the first polypeptide and the coding sequence of the second polypeptide, thus facilitating translation of the second polypeptide.
- a polynucleotide of the invention also can be flanked by a first cloning site and a second cloning site, thus providing a cassette that readily can be inserted into or linked to a second polynucleotide.
- Such flanking first and second cloning sites can be the same or different, and one or both independently can be one of a plurality of cloning sites, i.e., a multiple cloning site.
- a polynucleotide of the invention contains, in operative linkage and in a 5' to 3' orientation, a nucleotide sequence encoding the second RBS, a nucleotide sequence encoding the first RBS, and an initiator ATG; and/or a nucleotide sequence complementary to such a polynucleotide.
- a polynucleotide of the invention contains, in operative linkage and in a 5' to 3' orientation, a nucleotide sequence encoding the second RBS, a nucleotide sequence encoding the first RBS, an initiator ATG, and at least one cloning site; and/or a nucleotide sequence complementary to such a polynucleotide.
- a polynucleotide of invention contains, in operative linkage and in a 5' to 3' orientation, a nucleotide sequence encoding the second RBS, a nucleotide sequence encoding the first RBS, and at least one cloning site positioned about 3 to 10 nucleotides 3' of the nucleotide sequence encoding first RBS; and/or a nucleotide sequence complementary to such a polynucleotide.
- the present invention also relates to a vector, which includes a polynucleotide encoding an operatively linked first RBS and second RBS as disclosed herein, and a nucleotide sequence of chloroplast genomic deoxyribonucleic acid (DNA), which can undergo homologous recombination with chloroplast genomic DNA.
- a nucleotide sequence of chloroplast genomic DNA generally, though not necessarily, is a silent nucleotide sequence, which does not encode a chloroplast gene, and is of a sufficient length such that the vector can undergo homologous recombination with a corresponding nucleotide sequence in the chloroplast genome.
- a vector of the invention also can contain one or more additional nucleotide sequences that confer desirable characteristics on the vector, including, for example, sequences that facilitate manipulation of the vector.
- the vector can contain, for example, one or more cloning sites, for example, a cloning site, which can be a multiple cloning site, positioned such that a heterologous polynucleotide can be inserted into the vector and operatively linked to the first RBS and second RBS.
- the vector also can contain a prokaryote origin of replication (ori), for example, an E.
- a chloroplast/prokaryote shuttle vector includes 1) a nucleotide sequence of chloroplast genomic DNA, which can undergo homologous recombination with chloroplast genomic DNA; 2) a prokaryotic origin; 3) a first RBS operatively linked to a second RBS, wherein the first (or second) RBS can direct translation of an operatively linked expressible polynucleotide in a chloroplast, and the second (or first) RBS can direct translation of the operatively linked expressible polynucleotide in a prokaryote; and 4) an operatively linked expressible polynucleotide, or a cloning site positioned such that a heterologous polynucleotide can be inserted
- a vector of the invention can be a circularized vector, or can be a linear vector, which has a first end and a second end.
- a linear vector of the invention can have one or more cloning sites at one or both ends, thus providing a means to circularize the vector or to link the vector to a second polynucleotide, which can be a second vector that is the same as or different from the vector of the invention.
- the cloning site can include a restriction endonuclease recognition site (or a cleavage product thereof), a recombinase site, or a combination of such sites.
- the vector can further contain one or more expression control elements, for example, transcriptional regulatory elements, additional translational elements, and the like.
- the vector contains an initiator ATG codon operatively linked to the sequence encoding the first RBS and second RBS, such that a polynucleotide encoding a polypeptide can be operatively linked adjacent to ATG codon and, upon transcription, can comprise an RNA that can be translated in a prokaryote and in a chloroplast.
- the vector also can contain a cloning site that is positioned to allow operative linkage of at least one heterologous polynucleotide to such an ATG codon.
- a vector of the invention also can contain a nucleotide sequence encoding a first polypeptide operatively linked to the first RBS and second RBS, wherein the encoding nucleotide sequence is modified to contain one or more cloning sites, including, for example, upstream of and near the ATG codon, downstream of and near the ATG codon, and/or at or near the C-terminus of the encoded polypeptide.
- Such a vector provides a convenient means to insert a nucleotide sequence encoding a second polypeptide therein, either by substitution of the nucleotide sequence encoding the first polypeptide, or in operative linkage near the N-terminus or C-terminus of the encoded polypeptide such that a fusion protein comprising the first and second polypeptide can be expressed.
- the present invention also relates to a cell, which contains a polynucleotide of the invention or a vector of the invention.
- the cell which can be a host cell for a vector of the invention, can be a prokaryotic or eukaryotic cell, including, for example, a bacterial cell such as an E. coli cell; a plant cell such as an algae or a monocot or dicot; an insect cell; or a vertebrate cell such as a mammalian cell.
- the polynucleotide, or vector can be contained in a plastid of the plant cell, particularly in a chloroplast, and can, but need not, be integrated into the plastid genome.
- the polynucleotide of the invention which can be contained in a vector, is operatively linked to an expressible polynucleotide, whereby the cell containing the polynucleotide provides an expression system, which allows the translation of one or more polypeptides encoded by the expressible polynucleotide.
- the expressible polynucleotide which can be biased for codon usage by the plastid, particularly chloroplast codon usage, encodes at least a first polypeptide, for example, a first polypeptide and a second polypeptide.
- the expressible polynucleotide encodes an antibody.
- the expressible polynucleotide is biased for chloroplast codon usage, for example, an expressible polynucleotide having a nucleotide sequence as set forth in SEQ ID NO:l, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO:42, SEQ ID NO:45, or SEQ ID NO:47.
- the present invention further relates to a transgenic plant, which comprises plant cells containing a polynucleotide of the invention integrated in chloroplast genomic DNA.
- a transgenic plant which comprises plant cells containing a polynucleotide of the invention integrated in chloroplast genomic DNA.
- the present invention provides a plant cell organelle or a cell or tissue obtained from such a transgenic plant, for example, a chloroplast isolated from the transgenic plant, or leaves or flowers isolated from the transgenic plant, a fruit or rhizome isolated from the transgenic plant, or a cutting of the transgenic plant, or a seed produced by the transgenic plant.
- the invention provides cDNA or chloroplast genomic DNA library prepared from the transgenic plant of the invention, or from a plant cell or plant tissue obtained from the transgenic plant.
- a transgenic plant of the invention can be any type of plant, including, for example, an algae, which can be microalgae or a macroalgae; a monocot; or a dicot such as an angiosperm (e.g., a cereal plant, a leguminous plant, an oilseed plant, or a hardwood tree), including an ornamental plant.
- an algae which can be microalgae or a macroalgae
- a monocot e.g., a cereal plant, a leguminous plant, an oilseed plant, or a hardwood tree
- an ornamental plant e.g., a cereal plant, a leguminous plant, an oilseed plant, or a hardwood tree
- the present invention further relates to a composition, which includes plant material obtained from a transgenic plant of the invention or from a plant cell genetically modified to contain a polynucleotide of the invention integrated in chloroplast genomic DNA of the plant.
- a composition which includes plant material obtained from a transgenic plant of the invention or from a plant cell genetically modified to contain a polynucleotide of the invention integrated in chloroplast genomic DNA of the plant.
- the polynucleotide encoding the operatively linked first RBS and second RBS in the transgenic plant or genetically modified plant cell is operatively linked to an expressible polynucleotide, which can, but need not, be biased for chloroplast codon usage.
- the plant material which can be cell organelles, cells, or one or more tissues obtained from a transgenic plant, for example, chloroplasts, or leaves or flowers, a fruit or rhizome, or a seed produced by a transgenic plant, provides a source of the polypeptide or polypeptides encoded by the expressible polynucleotide.
- a transgenic plant for example, chloroplasts, or leaves or flowers, a fruit or rhizome, or a seed produced by a transgenic plant
- the expressible polynucleotide encodes an antibody, or an antigen binding fragment thereof
- the plant material and, therefore, the composition provides a source of the antibody.
- a composition of the invention can be formulated such that it is in a form suitable for administration to a living subject, for example, a vertebrate or other mammal, which can be a domesticated animal or a pet, or can be a human.
- a composition comprising a plant material as disclosed herein can be useful as a nutritional supplement, a therapeutic agent, and the like.
- the expressible polynucleotide encodes an antibody, or antigen binding fragment thereof
- the composition can be useful for passive immunization of a subject such as an individual exposed to a herpesvirus, or an individual exposed to tetanus toxin.
- the present invention provides a medicament useful for ameliorating a pathologic condition such as a herpesvirus infection.
- the present invention also relates to an isolated polynucleotide encoding a fluorescent protein or a mutant or variant thereof, wherein codons of the polynucleotide are biased to reflect chloroplast codon usage.
- the polynucleotide can be a DNA sequence or an RNA sequence, and can be single stranded or double stranded, and can be a linear polynucleotide containing a cloning site at one or both ends.
- the polynucleotide also can be operatively linked to a polynucleotide encoding a first RBS and a second RBS that are spaced apart by about 5 to 25 nucleotides, such that the fluorescent protein conveniently can be translated in a prokaryote and in a chloroplast.
- One or more codons encoding a fluorescent protein of the invention can be biased, for example, to contain an adenine or a thymine at position three, thus facilitating translation of the fluorescent protein in a chloroplast.
- the fluorescent protein can be a green fluorescent protein (GFP) such as that produced by a species of Aequorea jellyfish.
- GFP green fluorescent protein
- Such polynucleotides of the invention are exemplified by polynucleotides that encodes the polypeptide set forth in SEQ ID NO:2, for example, the polynucleotide set forth in SEQ ID NO:l.
- the present invention also provides a fluorescent protein encoded by and expressed from such a polynucleotide, for example, a fluorescent protein having an amino acid sequence as set forth in SEQ ID NO:2.
- the present invention further relates to a recombinant nucleic acid molecule, which includes a first polynucleotide, which encodes at least one polypeptide and contains one or more codons biased to reflect chloroplast codon usage; and a second polynucleotide, which comprises a nucleotide sequence encoding a first RBS operatively linked to a nucleotide sequence encoding a second RBS, wherein the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast.
- the first polynucleotide can encode a single polypeptide, or can encode two or more polypeptides, which can be expressed as separate polypeptides or as a fusion protein. Where the first polynucleotide encodes two or more polypeptides, the nucleotide sequence between the coding sequences can, but need not, encode an internal ribosome entry site, which is positioned so as to facilitate translation of the second (or other) polypeptide.
- a recombinant nucleic acid molecule of the invention can further include a third polynucleotide, which can be operatively linked to the first and second polynucleotides and can, but need not, encode one or more polypeptides.
- the present invention also relates to a method of making a chloroplast/prokaryote shuttle expression vector.
- a method of making a chloroplast/prokaryote shuttle expression vector can be performed, for example, by introducing into a nucleotide sequence of chloroplast genomic DNA sufficient to undergo homologous recombination with chloroplast genomic DNA, a nucleotide sequence comprising a prokaryote origin of replication; a nucleotide sequence encoding a first RBS; and a nucleotide sequence encoding a second RBS, wherein the first RBS and second RBS are spaced apart by about 5 to 25 nucleotides; and a cloning site, wherein the cloning site is positioned to allow operative linkage of a polynucleotide encoding a polypeptide to the first RBS and second RBS such that the first RBS can direct translation of the polypeptide in a prokaryote and the second
- a method of making a chloroplast/prokaryote shuttle expression vector also can be performed by genetically modifying a nucleotide sequence of chloroplast genomic deoxyribonucleic acid (DNA), which is sufficient to undergo homologous recombination with chloroplast genomic DNA, to contain a prokaryote origin of replication, a nucleotide sequence encoding a first RBS spaced apart from a second RBS by about 5 to 25 nucleotides, and a cloning site positioned to allow operative linkage of a polynucleotide encoding a polypeptide to the first RBS and second RBS such that the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast.
- the present invention also provides a chloroplast/prokaryote shuttle vector produced by a method as disclosed herein.
- the present invention further relates to a recombinant polynucleotide, which includes a first nucleotide sequence encoding a chloroplast RBS operatively linked to at least a second nucleotide sequence encoding a polypeptide, wherein the first nucleotide sequence is heterologous with respect to the second nucleotide sequence.
- a recombinant polynucleotide can further include an operatively linked third (or more) nucleotide sequence encoding a second (or other) polypeptide, thus providing a recombinant polynucleotide encoding a dicistronic (or polycistronic) polyribonucleotide sequence.
- a nucleotide encoding an operatively linked RBS generally is positioned about 20 to 40 nucleotides 5' (upstream) to an initiator ATG codon, which, in turn, is operatively linked to the nucleotide sequence encoding the polypeptide.
- the first nucleotide sequence comprises an ATG codon positioned about 20 to 40 nucleotides 3' of sequence encoding the RBS.
- an internal ribosome binding sequence is operatively linked between two or more nucleotide sequences encoding polypeptides, which can be the same or different.
- the present invention also relates to a vector, which includes a nucleotide sequence encoding an RBS positioned about 20 to 40 nucleotides 5' to a cloning site.
- the cloning site can be any nucleotide sequence that facilitates insertion or linkage of a nucleotide sequence to the vector, for example, one or more restriction endonuclease recognition sites, one or more recombinase recognition sites, or a combination of such sites.
- the cloning site is a multiple cloning site, which includes a plurality of restriction endonuclease recognition sites or recombinase recognition sites, or a combination of at least one restriction endonuclease recognition site and at least one recombinase recognition site.
- the vector can further contain an initiator ATG codon or a portion thereof adjacent and 5' to the cloning site, thus providing a translation start site for a coding sequence that otherwise lacks an initiator ATG codon.
- the vector can contain a chloroplast gene 3' untranslated region positioned 3' to the cloning site.
- Figure 1 provides a comparison of the GFPct (SEQ ID NO: 1) and GFPncb (SEQ ID NO:3) coding regions.
- the amino acid sequence of GFPct (SEQ ID NO:2) is shown below the nucleotide sequence. Changed codons are boxed, and those that show a significant improvement in usage are shaded.
- the optimized codons were defined as codons used more than 10 times per 1000 codons in the C. reinhardtii chloroplast genome (Nakamura et al., Nucl. Acids Res. 27:292, 1999).
- the asterisk (*) indicates the two amino acid changes between GFPct and GFPncb, at positions 2 and 65.
- Figure 2 provides a characterization of pET expressed GFPct and GFPncb.
- GFPct and GFPncb proteins expressed from pET19b plasmid in E. coli were purified via Ni agarose affinity chromatography (Example 1).
- Crude E. coli lysates containing either GFPct or GFPncb proteins (20 ⁇ l) were prepared by subjecting samples to 12% SDS-PAGE without boiling, and disassembling the gel apparatus, but leaving the gel encased within the glass plates. Fluorescent gels were visualized with the indicated excitation (ex) and emission (em) filters.
- affinity purified GFPct or GFPncb proteins Five ⁇ g of affinity purified GFPct or GFPncb proteins were separated on a 12% SDS-PAGE and stained with Coomassie. 100 ng of affinity purified GFPct or GFPncb protein was subjected to 12% SDS PAGE followed by western blotting and detection with anti-GFP primary antibody. Excitation spectra were generated for affinity purified GFPct (4 ⁇ g), and GFPncb (20 ⁇ g) proteins. Relative fluorescence was recorded at excitation from 350 to 550 nm with emission fixed at 510 nm. The GFPncb (stippled line) and GFPct (solid line) excitation spectra are shown; dashed line represents the 510 nm emission peak seen in both samples.
- Figure 3 provides maps of the GFPct and GFPncb reporter gene used for expression in C. reinhardtii chloroplasts.
- Figure 3A shows relevant restriction sites delimiting the rbcL 5' UTR (Bam HI/Nde I; see, also, SEQ ID NO:5) from either GFPct (SEQ ID NO:l) or GFPncb (SEQ ID NO:3) coding regions (Ndel Xba I) and the rbcL 3 TR (Xba I/Bam HI; see, also, SEQ ID NO: 10). Size of each fragment in base pairs (bp) is indicated.
- Figure 3 B shows the site of integration into the C. reinhardtii chloroplast genome of the GFPct and GFPncb genes under control of the rbcL 5' and 3' UTRs.
- C. reinhardtii chloroplast DNA is depicted as the Eco Rl to Xho I fragment of 5.7 kb. Double headed arrows indicate regions corresponding to the probes used in the Southern blot analysis.
- Figure 4 shows the linear sequence of the mutant psbA 5'UTR's (SEQ ID NOS:35 to 41) corresponding to positions +3 to -36 relative to the initiation codon of the wild type 5'UTR (SEQ ID NO:34).
- the 5'UTR's were placed upstream of the DI cDNA, which is an intron-less copy of the wild type psbA gene. Changes to the primary sequence are underlined and the initiation codons are boxed.
- the * denotes the 5' terminus of the mRNA in vivo resulting from a processing event that cleaves the 5'UTR (see Bruick and Mayfield, Trends Plant Sci. 4:190-195, 1998, which is incorporated herein by reference).
- FIGS 5 A to 5 C provide restriction maps of HSV8-lsc genes for expression in C. reinhardtii chloroplasts.
- HSV8-lsc nucleotide (SEQ ID NO:47) and amino acid (SEQ ID NO:48) sequences are provided in the Sequence Listing.
- Figures 5A and 5B show relevant restriction sites delineating the rbcL 5'UTR (Bam HI/Nde I), the HSV8 coding region and flag tag (Ndel/Xba I), and the rbcL 3'UTR (Xba I/Bam HI; Figure 5 A), as well as relevant restriction sites of the atpA 5'UTR (Bam HI/Nde I), the HSV8 coding region and flag tag (Ndel Xba I), and the rbcL 3'UTR (Xba I/Bam HI; Figure 5B).
- Figure 5C provides a restriction map showing the site of integration of the HSV8-lsc genes into plasmid p322 for integration into the C. reinhardtii chloroplast genome.
- p322 DNA includes the 5.7 kb region from Eco Rl to Xho I in the C. reinhardtii chloroplast genome corresponding to position 44,877 to 50,577 (see world wide web at URL "biology.duke.edu chlamy_genome/chloro.html").
- Double headed arrows indicate regions corresponding to the probes used in the Southern blot analysis. Black boxes indicate (from left to right) psbA exon 5, and the 5S and a small portion of the 23 S ribosomal RNA genes, respectively.
- Figure 6 provides a characterization of HSV8-lsc binding to HS V8 viral protein obtained by ELISA.
- Affinity purified HSV8-lsc from the transgenic C. reinhardtii strains (10-6-3 and 16-3) were screened in an ELISA assay against HSV proteins prepared from virus infected cells. 100, 80, 70, 60, 30, 20, 10 or 5 ⁇ l of Flag purified HSV8-lsc were incubated in microtiter plates coated with a constant amount of viral protein. Protein concentrations in these affinity purified extracts was 13 ng/ ⁇ l, of which approximately 10% was HSV8-lsc as judged by Coomassie staining. Equal volumes of wt C. reinhardtii proteins were used as a negative control (concentration 1 ⁇ g/ ⁇ l).
- Figure 7 provides a comparison of the luxAB (SEQ ID NO:44) and luxCt (amino acid residues 2 to 695 of SEQ ID NO:46) coding regions.
- the amino acid sequence is shown with the modified codons indicated by boxed and shaded amino acids.
- the optimized codons were defined as codons used more than 10 times per 1000 codons in the C. reinhardtii chloroplast genome (Nakamura et al. 1999).
- the amino acid differences between the two proteins are indicated by boxed and unshaded amino acids, and the two amino acids changed that resulted in active luciferase are indicated by the ** above the changed amino acids.
- Figures 8 A and 8B provide maps of luxCt gene for expression in C. reinhardtii chloroplasts.
- Figure 8A illustrates relevant restriction sites delineating the atpA 5' UTR (Bam HI/Nde I), the luxCt coding region (Ndel/Xba I) and the rbcL 3' UTR (Xba I/Bam HI).
- Figure 8B provides a map showing the homologous region between plasmid p322 and the C. reinhardtii chloroplast genome into which the chimeric luxCt gene was inserted.
- C. reinhardtii chloroplast DNA depicted is the Eco Rl to Xho I fragment of 5.7 kb located in the inverted repeat region of the chloroplast region.
- Double headed arrows indicate regions corresponding to the probes used in the Southern and.Northern blot analysis. Black boxes indicate, from 1 to r, psbA exon 5, 5s rRNA and 23s RNA genes, respectively.
- a method of the invention is exemplified by expressing functional antibodies, including single chain antibodies that properly assemble and function to specifically bind antigen, as well as antibodies and antigen binding fragments thereof that are expressed as single chains and that specifically associate to form homodimers that specifically bind antigen.
- the polynucleotides encoding the antibodies are operatively linked to a 5 '-untranslated region (5'UTR) comprising a ribosome binding sequence (RBS) that directs translation of the antibodies in chloroplasts.
- 5'UTR 5 '-untranslated region
- RBS ribosome binding sequence
- the polynucleotides encoding the antibodies are operatively linked to a first RBS, which directs translation in a prokaryotic cell, and a second RBS, which directs translation in a chloroplast.
- the polynucleotide encoding an antibody is biased for chloroplast codon usage.
- a synthetic polynucleotide which includes at least a first nucleotide sequence encoding at least a first polypeptide, wherein at least one codon in the first nucleotide sequence is biased to reflect chloroplast codon usage, is introduced into a cell, wherein the encoded polypeptide is expressed.
- each codon in the first nucleotide sequence is biased to reflect chloroplast codon usage
- the synthetic polynucleotide contains at least a second nucleotide sequence, which can, but need not, be operatively linked to the first nucleotide sequence, and encodes at least a second polypeptide, wherein expression of the polynucleotide can, but need not, generate a fusion protein comprising the first and second (or more) polypeptides.
- a synthetic polynucleotide which includes at least a first nucleotide sequence encoding at least a first polypeptide, wherein at least one codon in the first nucleotide sequence is biased to reflect chloroplast codon usage.
- synthetic polynucleotide means a nucleic acid molecule that has been modified by changing a codon in the polypeptide that is not biased for chloroplast codon usage to a codon that is biased for chloroplast codon usage (see Table 1, below).
- polypeptides encoded by such synthetic polynucleotides are robustly expressed in chloroplasts.
- compositions for practicing a method of the invention are provided.
- Advantages provided by the present invention include the ability to obtain robust expression of functional polypeptides in plant chloroplasts, wherein the polypeptides are not glycosylated and, therefore, have reduced antigenicity upon administration to a subject, as well as the ability to produce large amounts of functional polypeptides without a requirement for a fermentation facility and the expense associated therewith.
- a method of the invention provides a means to express one or more polypeptides in a plant chloroplast, whereby the polypeptides can assemble to produce functional protein complexes.
- polypeptides expressed in chloroplasts not only assemble properly, but also, where the polypeptides comprise subunits of a protein complex, the polypeptides can specifically associate to produce a functional protein complex.
- protein complex refers to a composition that is formed by the specific association of at least two polypeptides, which can be the same or different.
- Polypeptides that specifically associate to function as protein complexes are well known and include enzymes, growth factors, growth factor and hormone receptors, and the like.
- the term “specifically associate” or “specifically interact” or “specifically bind” refers to two or more polypeptides that form a complex that is relatively stable under physiologic conditions.
- the terms are used herein in reference to various interactions, including, for example, the interaction of a first polypeptide subunit and a second polypeptide subunit that interact to form a functional protein complex, as well as to the interaction of an antibody and its antigen.
- a specific interaction can be characterized by a dissociation constant of at least about 1 x 10 "6 M, generally at least about 1 x 10 "7 M, usually at least about 1 x 10 "8 M, and particularly at least about 1 x 10 "9 M or 1 x 10 "10 M or greater.
- a specific interaction generally is stable under physiological conditions, including, for example, conditions that occur in a cell or subcellular compartment of a living subject, including a plant or an animal, which can be a vertebrate or invertebrate, as well as conditions that occur in a cell culture such as used for maintaining cells or tissues of an organism.
- Methods for determining whether two molecules interact specifically are well known and include, for example, equilibrium dialysis, surface plasmon resonance, gel shift analyses, and the like.
- an antibody is used broadly herein to refer to a polypeptide or a protein complex that can specifically bind an epitope of an antigen.
- an antibody contains at least one antigen binding domain that is formed by an association of a heavy chain variable region domain and a light chain variable region domain, particularly the hypervariable regions.
- An antibody generated according to a method of the invention can be based on naturally occurring antibodies, for example, bivalent antibodies, which contain two antigen binding domains formed by first heavy and light chain variable regions and second heavy and light chain variable regions (e.g., an IgG or IgA isotype) or by a first heavy chain variable region and a second heavy chain variable region (V HH antibodies; see, for example, U.S. Pat. No. 6,005,079), or on non-naturally occurring antibodies, including, for example, single chain antibodies, chimeric antibodies, bifunctional antibodies, and humanized antibodies, as well as antigen-binding fragments of an antibody, for example, an Fab fragment, an Fd fragment, an Fv fragment, and the like.
- first heavy and light chain variable regions and second heavy and light chain variable regions e.g., an IgG or IgA isotype
- V HH antibodies first heavy chain variable region and a second heavy chain variable region
- non-naturally occurring antibodies including, for example, single chain antibodies, chimeric antibodies
- a method of the invention is exemplified using a polynucleotide encoding a single chain antibody comprising a heavy chain operatively linked to a light chain, wherein the antibody specifically binds tetanus toxin (see SEQ ID NOS:13 and 14).
- the method is exemplified using a polynucleotide encoding a single chain antibody comprising a heavy chain operatively linked to a light chain, wherein the antibody specifically binds herpes simplex virus types 1 and 2, and wherein the polynucleotide encoding the antibody is biased for chloroplast codon usage (see SEQ ID NOS: 15 and 16; SEQ ID NOS:42 and 43; and SEQ ID NOS:47 and 48; see, also, Example 3).
- Polynucleotides useful for practicing a method of the invention can be isolated from cells producing the antibodies of interest, for example, B cells from an immunized subject or from an individual exposed to a particular antigen, can be synthesized de novo using well l ⁇ iown methods of polynucleotide synthesis, can be produced recombinantly or can be obtained, for example, by screening combinatorial libraries of polynucleotides that encode variable heavy chains and variable light chains (see Huse et al., Science 246:1275-1281 (1989), which is incorporated herein by reference), and can be biased for chloroplast codon usage, if desired (see Example 1, and Table 1).
- Polynucleotides encoding humanized monoclonal antibodies can be obtained by transferring nucleotide sequences encoding mouse complementarity determining regions from heavy and light variable chains of the mouse immunoglobulin gene into a human variable domain gene, and then substituting human residues in the framework regions of the murine counterparts.
- General techniques for cloning murine immunoglobulin variable domains are known (see, for example, Orlandi et al., Proc. Natl. Acad.
- the methods of the invention also can be practiced using polynucleotides encoding human antibody fragments isolated from a combinatorial immunoglobulin library (see, for example, Barbas et al, Methods: A Companion to Methods in Immunology 2:119, 1991; Winter et &l, Ann. Rev. Immunol. 12:433, 1994; each of which is incorporated herein by reference).
- Cloning and expression vectors that are useful for producing a human immunoglobulin phage library can be obtained, for example, from Stratagene Cloning Systems (La Jolla, CA).
- a polynucleotide encoding a human monoclonal antibody also can be obtained, for example, from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge.
- elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy and light chain loci.
- the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas, from which polynucleotides useful for practicing a method of the invention can be obtained.
- Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., Nature Genet.
- transgenic mice 7:13, 1994; Lonberg et al, Nature 36 ⁇ :856, 1994; and Taylor et al., Intl. Immunol 6:579, 1994; each of which is incorporated herein by reference, and such transgenic mice are commercially available (Abgenix, Inc.; Fremont CA).
- the polynucleotide also can be one encoding an antigen binding fragment of an antibody.
- Antigen binding antibody fragments which include, for example, Fv, Fab, Fab', Fd, and F(ab')2 fragments, are well known in the art, and were originally identified by proteolytic hydrolysis of antibodies.
- antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
- Antibody fragments produced by enzymatic cleavage of antibodies with pepsin generate a 5S fragment denoted F(ab')2- This fragment can be further cleaved using a thiol reducing agent and, optionally, a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments.
- a thiol reducing agent optionally, a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages
- an enzymatic cleavage using pepsin produces two monovalent Fab' fragments and an Fc fragment directly (see, for example, Goldenberg, U.S. Pat. No. 4,036,945 and U.S. Pat. No.
- Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR).
- CDR peptides can be obtained by constructing a polynucleotide encoding the CDR of an antibody of interest, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al, Methods: A Companion to Methods in Enzymology 2:106, 1991, which is incorporated herein by reference).
- Polynucleotides encoding such antibody fragments, including subunits of such fragments and peptide linkers joining, for example, a heavy chain variable region and light chain variable region can be prepared by chemical synthesis methods or using routine recombinant DNA methods, beginning with polynucleotides encoding full length heavy chains and light chains, which can be obtained as described above.
- the present methods are based, in part, on the determination that proper positioning of a ribosome binding sequence (RBS) with respect to a coding sequence results in robust translation in plant chloroplasts (see below; see, also, Example 2), and that polypeptides that are known to specifically associate to form protein complexes when produced naturally in an organism (e.g., antibodies) also can associate properly in chloroplasts (see Example 3).
- RBS ribosome binding sequence
- polypeptides that are known to specifically associate to form protein complexes when produced naturally in an organism e.g., antibodies
- An advantage of expressing such polypeptides in chloroplasts is that the polypeptides do not proceed through cellular compartments typically traversed by polypeptides expressed from a nuclear gene and, therefore, are not subject to certain post-translational modifications such as glycosylation.
- polypeptides and protein complexes produced by a method of the invention can be expected to be less antigenic than the same polypeptides would be if expressed from a polynucleotide introduced into the nucleus of a eukaryote.
- a method of the invention provides a means to produce functional polypeptides such as single chain antibodies, and protein complexes such as a bivalent antibody, which include, for example, a first heavy and light chain associated with a second heavy and light chain.
- a method of the invention can be performed, for example, by introducing a recombinant nucleic acid molecule into a chloroplast, wherein the recombinant nucleic acid molecule includes a first polynucleotide, which encodes at least one polypeptide (i.e., 1, 2, 3, 4, or more), operatively linked to a second polynucleotide, which includes a nucleotide sequence encoding a first RBS operatively linked to a nucleotide sequence encoding a second RBS, under conditions that allow expression of the at least one polypeptide.
- Such conditions include those that allow or facilitate entry of the recombinant nucleic acid molecule into the chloroplast and, preferably, incorporation of the recombinant nucleic acid molecule into the chloroplast genome.
- Such methods include those exemplified herein, as well as other methods known and routine in the art.
- operatively linked means that two or more molecules are positioned with respect to each other such that they act as a single unit and effect a function attributable to one or both molecules or a combination thereof.
- a polynucleotide encoding a polypeptide can be operatively linked to a transcriptional or translational regulatory element, in which case the element confers its regulatory effect on the polynucleotide similarly to the way in which the regulatory element would effect a polynucleotide sequence with which it normally is associated with in a cell.
- a first polynucleotide coding sequence also can be operatively linked to a second (or more) coding sequence such that a chimeric polypeptide can be expressed from the operatively linked coding sequences (see, for example, SEQ ID NO: 30, showing site where polynucleotide, which encodes a GFP and was biased for chloroplast codon usage (i.e., SEQ ID NO:l) was inserted into the PsbD gene, such that a fluorescent fusion protein comprising the PsbD gene product fused to GFP was generated).
- the chimeric polypeptide can be a fusion polypeptide, in which the two (or more) encoded peptides are translated into a single polypeptide, i.e., are covalently bound through a peptide bond, for example, a single chain antibody comprising a heavy chain variable region operatively linked (through a linker peptide, if desired) to a light chain variable region; or can be translated as two discrete peptides that, upon translation, can specifically associate with each other to form a stable protein complex, for example, an antibody heavy chain and an antibody light chain, which form a quaternary structure resulting in a functional monovalent antibody, and which can further associate to produce a functional bivalent antibody.
- Examples of synthetic polynucleotides encoding such fusion proteins include SEQ ID NO:45, which encodes a bacterial luciferase fusion protein, and SEQ ID NOS: 15, 42, and 47, which encode single chain anti-HSV antibodies.
- polynucleotide or “nucleotide sequence” or “nucleic acid molecule” is used broadly herein to mean a sequence of two or more deoxyribonucleotides or ribonucleotides that are linked together by a phosphodiester bond.
- the terms include RNA and DNA, which can be a gene or a portion thereof, a cDNA, a synthetic polydeoxyribonucleic acid sequence, or the like, and can be single stranded or double stranded, as well as a DNA/RNA hybrid.
- nucleic acid molecules which can be isolated from a cell
- synthetic polynucleotides which can be prepared, for example, by methods of chemical synthesis or by enzymatic methods such as by the polymerase chain reaction (PCR).
- PCR polymerase chain reaction
- the nucleotides comprising a polynucleotide are naturally occurring deoxyribonucleotides, such as adenine, cytosine, guanine or thymine linked to 2'-deoxyribose, or ribonucleotides such as adenine, cytosine, guanine or uracil linked to ribose.
- a polynucleotide also can contain nucleotide analogs, including non-naturally occurring synthetic nucleotides or modified naturally occurring nucleotides.
- Nucleotide analogs are well known in the art and commercially available (e.g., Ambion, Inc.; Austin TX), as are polynucleotides containing such nucleotide analogs (Lin et al., Nucl. Acids Res. 22:5220-5234, 1994; Jellinek et al., Biochemistry 34:11363-11372, 1995; Pagratis et al, Nature Biotechnol 15:68-73, 1997, each of which is incorporated herein by reference).
- the covalent bond linking the nucleotides of a polynucleotide generally is a phosphodiester bond.
- the covalent bond also can be any of numerous other bonds, including a thiodiester bond, a phosphorothioate bond, a peptide-like bond or any other bond known to those in the art as useful for linking nucleotides to produce synthetic polynucleotides (see, for example, Tam et al, Nucl Acids Res. 22:977-986, 1994; Ecker and Crooke, BioTechnology 13:351360, 1995, each of which is incorporated herein by reference).
- a polynucleotide comprising naturally occurring nucleotides and phosphodiester bonds can be chemically synthesized or can be produced using recombinant DNA methods, using an appropriate polynucleotide as a template.
- a polynucleotide comprising nucleotide analogs or covalent bonds other than phosphodiester bonds generally will be chemically synthesized, although an enzyme such as T7 polymerase can incorporate certain types of nucleotide analogs into a polynucleotide and, therefore, can be used to produce such a polynucleotide recombinantly from an appropriate template (Jellinek et al., supra, 1995).
- recombinant nucleic acid molecule is used herein to refer to a polynucleotide that is manipulated by human intervention.
- a recombinant nucleic acid molecule can contain two or more nucleotide sequences that are linked in a manner such that the product is not found in a cell in nature.
- the two or more nucleotide sequences can be operatively linked and, for example, can encode a fusion polypeptide, or can comprise an encoding nucleotide sequence and a regulatory element, particularly a first RBS operatively linked to a second RBS.
- a recombinant nucleic acid molecule also can be based on, but manipulated so as to be different, from a naturally occurring polynucleotide, for example, a polynucleotide having one or more nucleotide changes such that a first codon, which normally is found in the polynucleotide, is biased for chloroplast codon usage, or such that a sequence of interest is introduced into the polynucleotide, for example, a restriction endonuclease recognition site or a splice site, a promoter, a DNA origin of replication, or the like.
- an RBS positioned about 20 to 40 nucleotides upstream (5') of an initiation codon, for example, an AUG codon allows robust translation of coding sequence starting with the AUG codon (see Example 2).
- an RBS positioned about 20 to 40 nucleotides upstream of an AUG codon is considered "operatively linked" to the AUG codon.
- an RBS positioned about 5 to 15 nucleotides upstream from an initiation codon can direct translation of a coding sequence in prokaryotes and, as disclosed herein, such an RBS can be operatively linked to a second RBS positioned about 20 to 40 nucleotides upstream of the initiation codon to produce a translational regulatory element than can direct t ranslation in a prokaryote and in a chloroplast.
- a first and second RBS spaced apart by about 5 to 25 nucleotides are considered operatively linked with respect to each other.
- first, second, third, etc. when used herein in reference to an RBS or a polynucleotide or polypeptide or the like, are used only for convenience of discussion and, unless specifically indicated otherwise, do not imply an order, importance, or the like.
- first RBS that can direct translation in a prokaryote
- second RBS that can direct translation in a chloroplast
- Reference to an RBS having the ability to "direct translation” means that, when operatively linked to a coding sequence, which generally begins with an initiation codon, the RBS can be bound by a ribosome such that translation can occur beginning with the initiation codon.
- initiation codon refers to a ribonucleotide sequence or encoding deoxyribonucleotide sequence that is the first codon of a coding sequence.
- an initiation codon is an "initiator AUG codon” (in RNA) or an “initiator ATG codon” (in DNA), and encodes methionine, although other codons also can act as an initiation codons, including, for example, CUG.
- Codons of an encoding polynucleotide can be biased to reflect chloroplast codon usage (Example 1). Most amino acids are encoded by two or more different (degenerate) codons, and it is well recognized that various organisms utilize certain codons in preference to others. Such preferential codon usage, which also is utilized in chloroplasts, is referred to herein as "chloroplast codon usage”. Table 1 (below) shows the chloroplast codon usage for C. reinhardtii.
- bias when used in reference to a codon, means that the sequence of a codon in a polynucleotide has been changed such that the codon is one that is used preferentially in chloroplasts (see Table 1).
- a polynucleotide that is biased for chloroplast codon usage can be synthesized de novo, or can be genetically modified using routine recombinant DNA techniques, for example, by a site directed mutagenesis method, to change one or more codons such that they are biased for chloroplast codon usage (see Example 1).
- chloroplast codon bias can be variously skewed in different plants, including, for example, in alga chloroplasts as compared to tobacco.
- chloroplast codon bias selected for purposes of the present invention, including, for example, in preparing a synthetic polynucleotide as disclosed herein, TABLE 1 Chloroplast Codon Usage in Chlamydomonas reinhardtii -
- chloroplast codon usage of a plant chloroplast reflects chloroplast codon usage of a plant chloroplast, and includes a codon bias that, with respect to the third position of a codon, is skewed towards A/T, fore example, where the third position has greater than about 66% AT bias, particularly greater than about 70% AT bias.
- chloroplast codon biased for purposes of the present invention excludes the third position bias observed, for example, in Nicotiana tabacus (tobacco), which has 34.56% GC bias in the third codon position (see, for example, world wide web at URL "kazusa.or.jp/codon/", and the "chloroplast” link).
- the chloroplast codon usage is biased to reflect alga chloroplast codon usage, for example, C reinhardtii, which has about 74.6% AT bias in the third codon position.
- a method of the invention can be performed using a polynucleotide that encodes a first polypeptide and at least a second polypeptide.
- the polynucleotide can encode, for example, a first polypeptide and a second polypeptide; a first polypeptide, a second polypeptide, and a third polypeptide; etc.
- any or all of the encoded polypeptides can be the same or different.
- polypeptides expressed in chloroplasts of the microalga Chlamydomonas reinhardtii assembled to form functional polypeptides and protein complexes (see Examples 1 and 3).
- a method of the invention provides a means to produce functional protein complexes, including, for example, dimers, trimers, and tetramers, wherein the subunits of the complexes can be the same or different (e.g., homodimers or heterodimers, respectively) .
- a method of expressing functional polypeptides and protein complexes in chloroplasts is exemplified by the production of antibodies, and by the production of reporter proteins, including a green fluorescent protein and a luciferase (luxAB fusion protein; see Examples 1 and 4; see, also, SEQ ID NOS:l and 45, respectively), and of an antibodies expressed from polynucleotides biased for chloroplast codon usage (see Example 3; see, also, SEQ ID NOS: 15, 42, and 47).
- chloroplasts were transfected with a recombinant nucleic acid molecule comprising a polynucleotide encoding a single chain antibody having a complete heavy chain linked to a light chain variable region, wherein homodimers comprising two single chain antibodies that associated through a specific interaction of their heavy chain domains were produced.
- a method of the invention can be practiced using a first recombinant nucleic acid molecule, which includes a nucleotide sequence encoding an RBS that directs translation in chloroplasts, and, preferably, further encoding an operatively linked RBS that directs translation in a prokaryote, the nucleotide sequence being operatively linlced to at least one polynucleotide encoding at least a first polypeptide.
- the recombinant nucleic acid molecule can include a polynucleotide encoding an immunoglobulin heavy chain (H) or a variable region thereof (VH), and can further encode a second polypeptide, which is an immunoglobulin light chain (L) or a variable region thereof (V L ).
- a nucleotide sequence encoding an internal ribosome entry site can be positioned between the nucleotide sequences encoding the H and L chains such that expression of the second (downstream) encoded polypeptide is facilitated.
- a H chain Upon translation of the encoded H and L chains in the chloroplast, a H chain can associate with a L chain to form a monovalent antibody (i.e., an H:L complex), and two H:L complexes can further associate to produce a bivalent antibody.
- a monovalent antibody i.e., an H:L complex
- a method of the invention also can be practiced by introducing into a plant chloroplast a first recombinant nucleic acid molecule, which includes a polynucleotide encoding, for example, a H chain or a V H chain, and further introducing into the chloroplast as second recombinant nucleic acid molecule, which includes a polynucleotide encoding a L chain or a V chain, wherein each recombinant nucleic acid molecule includes a nucleotide sequence encoding a first RBS operatively linked to a nucleotide sequence encoding a second RBS, wherein the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast, and wherein the nucleotide sequence encoding the two RBS is operatively linlced to the encoding polynucleotide sequence.
- the H chains and L chains can associate to form an H:L complex, and the H:L complexes can further associate to produce a bivalent antibody.
- a recombinant nucleic acid molecule comprising a polynucleotide encoding a polypeptide can further contain, operatively linked to the coding sequence, a peptide tag such as a His-6 tag or the like, which can facilitate identification of expression of the polypeptide in a cell.
- a polyhistidine tag peptide such as His-6 can be detected using a divalent cation such as nickel ion, cobalt ion, or the like.
- Additional peptide tags include, for example, a FLAG epitope, which can be detected using an anti-FLAG antibody (see, for example, Hopp et al, BioTechnology 6:1204 (1988); U.S. Pat. No.
- tags can provide the additional advantage that they can facilitate isolation of the operatively linked polypeptide, for example, where it is desired to obtain a substantially purified polypeptide.
- a recombinant nucleic acid molecule useful in a method of the invention can be contained in a vector. Furthermore, where the method is performed using a second (or more) recombinant nucleic acid molecules, the second recombinant nucleic acid molecule also can be contained in a vector, which can, but need not, be the same vector as that containing the first recombinant nucleic acid molecule.
- the vector can be any vector useful for introducing a polynucleotide into a chloroplast and, preferably, includes a nucleotide sequence of chloroplast genomic DNA that is sufficient to undergo homologous recombination with chloroplast genomic DNA, for example, a nucleotide sequence comprising about 400 to 1500 or more substantially contiguous nucleotides of chloroplast genomic DNA.
- Chloroplast vectors and methods for selecting regions of a chloroplast genome for use as a vector are well known (see, for example, Bock, J Mol. Biol. 312:425- 438, 2001; see, also, Staub and Maliga, Plant Cell 4:39-45, 1992; Kavanagh et al., Genetics 152:1111-1122, 1999, each of which is incorporated herein by reference).
- the nucleotide sequence of the chloroplast genomic DNA is selected such that it is not a portion of a gene, including a regulatory sequence or coding sequence, particularly a gene that, if disrupted due to the homologous recombination event, would produce a deleterious effect with respect to the chloroplast, for example, for replication of the chloroplast genome, or to a plant cell containing the chloroplast.
- the website containing the C. reinhardtii chloroplast genome sequence also provides maps showing coding and non-coding regions of the chloroplast genome, thus facilitating selection of a sequence useful for constructing a vector of the invention.
- the chloroplast vector, p322 which was used in experiments disclosed herein, is a clone extending from the Eco (Eco Rl) site at about position 143.1 kb to the Xho (Xho I) site at about position 148.5 kb (see, world wide web, at the URL
- the vector also can contain any additional nucleotide sequences that facilitate use or manipulation of the vector, for example, one or more transcriptional regulatory elements, a sequence encoding a selectable markers, one or more cloning sites, and the like.
- the chloroplast vector contains a prokaryote origin of replication (ori), for example, an E. coli ori, thus providing a shuttle vector that can be passaged and manipulated in a prokaryote host cell as well as in a chloroplast.
- ori prokaryote origin of replication
- microalga C. reinhardtii
- the use of microalgae to express a polypeptide or protein complex according to a method of the invention provides the advantage that large populations of the microalgae can be grown, including commercially (Cyanotech Corp.; Kailua-Kona HI), thus allowing for production and, if desired, isolation of large amounts of a desired product.
- the ability to express, for example, functional mammalian polypeptides, including protein complexes, in the chloroplasts of any plant allows for production of crops of such plants and, therefore, the ability to conveniently produce large amounts of the polypeptides.
- the methods of the invention can be practiced using any plant having chloroplasts, including, for example, macroalgae, for example, marine algae and seaweeds, as well as plants that grow in soil, for example, corn (Zea mays), Brassica sp. (e.g., B. napus, B. rapa, B.
- chloroplasts including, for example, macroalgae, for example, marine algae and seaweeds, as well as plants that grow in soil, for example, corn (Zea mays), Brassica sp. (e.g., B. napus, B. rapa, B.
- juncea particularly those Brassica species useful as sources of seed oil, alfalfa (Medicago sativa), rice (Oryza sativa), rye (Secale cereale), sorghum (Sorghum bicolor, Sorghum vulgare), millet (e.g., pearl millet (Pennisetum glaucum), proso millet (Panicum miliaceum), foxtail millet (Setaria italica), finger millet (Eleusine coracana)), sunflower (Helianthus annuus), safflower (Carthamus tinctorius), wheat (Triticum aestivum), soybean (Glycine max), tobacco (Nicotiana tabacum), potato (Solanum tuberosum), peanuts (Arachis hypogaea), cotton (Gossypium barbadense, Gossypium hirsutum), sweet potato (Ipomoea batatus), cassava (Manihot esculent
- cantalupensis cantalupensis
- musk melon C. melo
- Ornamentals such as azalea (Rhododendron spp.), hydrangea (Macrophylla hydrangea), hibiscus (Hibiscus rosasanensis), roses (Rosa spp.), tulips (Tulipa spp.), daffodils (Narcissus spp.), petunias (Petunia hybrida), carnation (Dianthus caryophyllus), poinsettia (Euphorbia pulcherrima), and chrysanthemum are also included.
- Additional ornamentals useful for practicing a method of the invention include impatiens, Begonia, Pelargonium, Viola, Cyclamen, Verbena, Vinca, Tagetes, Primula, Saint Paulia, Agertum, Amaranthus, Antihirrhinum, Aquilegia, Cineraria, Clover, Cosmo, Cowpea, Dahlia, Datura, Delphinium, Gerbera, Gladiolus, Gloxinia, Hippeastrum, Mesembryanthemum, Salpiglossos, and Zinnia.
- Conifers that may be employed in practicing the present invention include, for example, pines such as loblolly pine (Pinus taeda), slash pine (Pinus elliotii), ponderosa pine (Pinus ponderosa), lodgepole pine (Pinus contorta), and Monterey pine (Pinus radiatd), Douglas- fir (Pseudotsuga menziesii); Western hemlock (Tsuga ultilane); Sitka spruce (Picea glauc ⁇ ); redwood (Sequoia sempervirens); true firs such as silver fir (Abies amabilis) and balsam fir (Abies balsamea); and cedars such as Western red cedar (Thuja plicata) and Alaska yellow-cedar (Chamaecyparis nootkatensis).
- pines such as loblolly pine (Pinus taeda), slash
- Leguminous plants useful for practicing a method of the invention include beans and peas.
- Beans include guar, locust bean, fenugreek, soybean, garden beans, cowpea, mung bean, lima bean, fava bean, lentils, chickpea, etc.
- Legumes include, but are not limited to, Arachis, e.g., peanuts, Vicia, e.g., crown vetch, hairy vetch, adzuki bean, mung bean, and chickpea, Lupinus, e.g., lupine, trifolium, Phaseolus, e.g., common bean and lima bean, Pisum, e.g., field bean, Melilotus, e.g., clover, Medicago, e.g., alfalfa, Lotus, e.g., trefoil, lens, e.g., lentil, and false indigo.
- Arachis e.g., peanuts
- Vicia e.g., crown vetch, hairy vetch, adzuki bean, mung bean, and chickpea
- Lupinus e.g., lupine, trifolium
- Phaseolus e.g., common bean and lim
- Preferred forage and turf grass for use in the methods of the invention include alfalfa, orchard grass, tall fescue, perennial ryegrass, creeping bent grass, and redtop.
- Other plants useful in the invention include Acacia, aneth, artichoke, arugula, blackberry, canola, cilantro, Clementines, escarole, eucalyptus, fennel, grapefruit, honey dew, jicama, kiwifruit, lemon, lime, mushroom, nut, okra, orange, parsley, persimmon, plantain, pomegranate, poplar, radiata pine, radicchio, Southern pine, sweetgum, tangerine, triticale, vine, yams, apple, pear, quince, cherry, apricot, melon, hemp, buckwheat, grape, raspberry, chenopodium, blueberry, nectarine, peach, plum, strawberry, watermelon,
- a method of the invention can generate a plant containing chloroplasts that are genetically modified to contain a stably integrated polynucleotide (i.e., transplastomes; see, for example, Hager and Bock, Appl. Microbiol. Biotechnol. 54:302-310, 2000, which is incorporated herein by reference; see, also, Bock, supra, 2001).
- the integrated polynucleotide can comprise, for example, an encoding polynucleotide operatively linked to a first and second RBS as defined herein.
- the present invention further provides a transgenic (transplastomic) plant, which comprises one or more chloroplasts containing a polynucleotide encoding one or more heterologous polypeptides, including polypeptides that can specifically associate to form a functional protein complex.
- a transgenic plant comprising a transplastome provides advantages over transgenic plants having a polynucleotide integrated in the nuclear genome. For example, in most crop species, chloroplasts are strictly maternally inherited through the egg; the pollen (sperm) lacks chloroplasts (see, for example, Hager and Bock, supra, 2000). As such, a transgenic plant comprising a transplastome is unable to cross-pollinate other plants, including native plants that may be in the vicinity of the transgenic plant, thus reducing any potential ecological risk associated with the growth of transgenic plants in the environment.
- plant is used broadly herein to refer to a eukaryotic organism containing plastids, particularly chloroplasts, and includes any such organism at any stage of development, or to part of a plant, including a plant cutting, a plant cell, a plant cell culture, a plant organ, a plant seed, and a plantlet.
- a plant cell is the structural and physiological unit of the plant, comprising a protoplast and a cell wall.
- a plant cell can be in the form of an isolated single cell or a cultured cell, or can be part of higher organized unit, for example, a plant tissue, plant organ, or plant.
- a plant cell can be a protoplast, a gamete producing cell, or a cell or collection of cells that can regenerate into a whole plant.
- a seed which comprises multiple plant cells and is capable of regenerating into a whole plant, is considered plant cell for purposes of this disclosure.
- a plant tissue or plant organ can be a seed, protoplast, callus, or any other groups of plant cells that is organized into a structural or functional unit.
- Particularly useful parts of a plant include harvestable parts and parts useful for propagation of progeny plants.
- a harvestable part of a plant can be any useful part of a plant, for example, flowers, pollen, seedlings, tubers, leaves, stems, fruit, seeds, roots, and the like.
- a part of a plant useful for propagation includes, for example, seeds, fruits, cuttings, seedlings, tubers, rootstocks, and the like.
- a transgenic plant can be regenerated from a transformed plant cell containing genetically modified chloroplasts.
- the term "regenerate” means growing a whole plant from a plant cell; a group of plant cells; a protoplast; a seed; or a piece of a plant such as a callus or tissue. Regeneration from protoplasts varies from species to species of plants. For example, a suspension of protoplasts can be made and, in certain species, embryo formation can be induced from the protoplast suspension, to the stage of ripening and germination.
- the culture media generally contains various components necessary for growth and regeneration, including, for example, hormones such as auxins and cytokinins; and amino acids such as glutamic acid and proline, depending on the particular plant species. Efficient regeneration will depend, in part, on the medium, the genotype, and the history of the culture. If these variables are controlled, however, regeneration is reproducible.
- hormones such as auxins and cytokinins
- amino acids such as glutamic acid and proline
- Regeneration can occur from plant callus, explants, organs or plant parts. Transformation can be performed in the context of organ or plant part regeneration, (see Meth. Enzymol Vol. 118; Klee et &l. Ann. Rev. Plant Physiol 38:467, 1987, which is incorporated herein by reference).
- leaf disk-transformation-regeneration method for example, disks are cultured on selective media, followed by shoot formation in about two to four weeks. Shoots that develop are excised from calli and transplanted to appropriate root-inducing selective medium. Rooted plantlets are transplanted to soil as soon as possible after roots appear. The plantlets can be repotted as required, until reaching maturity.
- the mature transgenic plants are propagated utilizing cuttings or tissue culture techniques to produce multiple identical plants. Selection of desirable transgenotes is made and new varieties are obtained and propagated vegetatively for commercial use.
- the mature transgenic plants can be self crossed to produce a homozygous inbred plant. The resulting inbred plant produces seeds that contain the introduced heterologous polynucleotide, and can be grown to produce plants that express a polypeptide encoded by the polynucleotide.
- the invention further provides seeds produced by a transgenic plant obtained by a method of the invention.
- transgenic plants of the invention containing chloroplasts that are genetically modified to express different heterologous polypeptides can be crossbred, thereby providing a means to obtain transgenic plants containing two or more different transgenes.
- Methods for breeding plants and selecting for crossbred plants having desirable characteristics or other characteristics of interest are well known in the art.
- a method of producing a heterologous polypeptide or protein complex in a chloroplast or in a transgenic plant of the invention can further include a step of isolating an expressed polypeptide or protein complex from the plant cell chloroplasts.
- isolated or “substantially purified” means that a polypeptide or polynucleotide being referred to is in a form that is relatively free of proteins, nucleic acids, lipids, carbohydrates or other materials with which it is naturally associated.
- an isolated polypeptide constitutes at least twenty percent of a sample, and usually constitutes at least about fifty percent of a sample, particularly at least about eighty percent of a sample, and more particularly about ninety percent or ninety-five percent or more of a sample.
- heterologous is used herein in a comparative sense to indicate that a nucleotide sequence (or polypeptide) being referred to is from a source other than a reference source, or is linked to a second nucleotide sequence (or polypeptide) with which it is not normally associated, or is modified such that it is in a form that is not normally associated with a reference material.
- a polynucleotide encoding an antibody is heterologous with respect to a nucleotide sequence of a plant chloroplast, as are the components of a recombinant nucleic acid molecule comprising, for example, a first nucleotide sequence operatively linked to a second nucleotide sequence, as is a mutated polynucleotide introduced into a chloroplast where the mutant polynucleotide is not normally found in the chloroplast.
- a polypeptide or protein complex can be isolated from chloroplasts using any method suitable for the particular polypeptide or protein complex, including, for example, salt fractionation methods and chromatography methods such as an affinity chromatography method using a ligand or receptor that specifically binds the polypeptide or protein complex.
- a determination that a polypeptide or protein complex produced according to a method of the invention is in an isolated form can be made using well known methods, for example, by performing electrophoresis and identifying the particular molecule as a relatively discrete band or the particular complex as one of a series of bands. Accordingly, the present invention also provides an isolated polypeptide or protein complex produced by a method of the invention.
- the present invention also provides compositions that can be used alone or in combination to obtain robust expression of heterologous polypeptides in a chloroplast.
- the invention provides a nucleotide sequence comprising (or encoding) a first RBS and a second RBS, wherein the first and second RBS are spaced apart such that one RBS directs translation in prokaryotic cells and the other RBS directs translation in plant chloroplasts.
- the nucleotide sequence also can contain (or encode) an initiation codon, for example, an initiator AUG (or ATG) codon, operatively linked to the first RBS and second RBS, or can contain a cloning site positioned so as to permit operative linkage of a coding sequence to the first and second RBS.
- the nucleotide sequence is contained in a vector, which, preferably, includes a nucleotide sequence of chloroplast genomic DNA that is sufficient to undergo site specific homologous recombination with a chloroplast genome.
- the vector is a shuttle vector that further contains a prokaryote origin of replication.
- codon selection is utilized to bias an encoding polynucleotide for chloroplast codon usage, thus providing a means to obtain robust expression of one or more encoded polypeptides in a chloroplast.
- the usefulness of codon selection to optimize polypeptide expression in chloroplasts is exemplified herein using the Aequeoria victoria green fluorescent protein (GFP; Example 1).
- GFP Aequeoria victoria green fluorescent protein
- the present invention also provides a polynucleotide encoding a GFP, wherein the polynucleotide has been codon optimized for expression in chloroplasts.
- the variant polynucleotide encodes a GFP that expressed in an amount making it useful as a reagent for detecting plant chloroplasts, including for examining gene expression in chloroplasts.
- the general usefulness of chloroplast codon optimization for expressing polypeptides is further demonstrated by the preparation of a synthetic polynucleotide encoding luciferase (Example 4), the expression of which can be detected in vivo or in vitro, and by polynucleotides encoding antibodies (Example 3).
- the exemplified compositions and methods demonstrate that functional fusion proteins can be expressed robustly in chloroplasts, including single chain antibodies and reporter polypeptides (see Examples 3 and 4).
- the chloroplasts of higher plants and algae likely originated by an endosymbiotic incorporation of a photosynthetic prokaryote into a eukaryotic host.
- genes were transferred from the chloroplast to the host nucleus (Gray, Curr. Opin. Gen. Devel. 9:678-687, 1999).
- proper photosynthetic function in the chloroplast requires both nuclear encoded proteins and plastid encoded proteins, as well as coordination of gene expression between the two genomes. Expression of nuclear and chloroplast encoded genes in plants is acutely coordinated in response to developmental and environmental factors.
- chloroplasts regulation of gene expression generally occurs after transcription, and often during translation initiation. This regulation is dependent upon the chloroplast translational apparatus, as well as nuclear-encoded regulatory factors (see Barkan and Goldsch idt-Clermont, Biochemie 82:559-572, 2000; Zerges, Biochemie 82:583-601, 2000; Bruick and Mayfield, supra, 1999).
- the chloroplast translational apparatus generally resembles that in bacteria; chloroplasts contain 70S ribosomes; have mRNAs that lack 5' caps and generally do not contain 3' poly-adenylated tails (Harris et al., Microbiol. Rev. 58:700-754, 1994); and translation is inhibited in chloroplasts and in bacteria by selective agents such as chloramphenicol.
- RNA elements that mediate proper translation initiation include an initiation codon, an RBS, a defined spacing between the RBS and the initiation codon, translational enhancer sequences, bias at the second codon, and secondary structures that affect RNA accessibility (Gold, Ann. Rev. Biochem. 57:199-233, 1988).
- ribosome binding and proper translation start site selection are mediated, at least in part, by cis-acting RNA elements (see Bruick and Mayfield, supra, 1999).
- chloroplast initiation codons affect the efficiency of translation initiation, but do not determine the location of the initiation site (Chen et al., Plant Cell 7:1295-1305, 1995), indicating that additional determinants are required for translation start site selection in chloroplasts.
- RNA elements that act as mediators of translational regulation have been identified within the 5'UTR's of chloroplast mRNAs (Alexander et al., Nucl. Acids Res. 26:2265-2272, 1998; Hirose and Sugiura, E 50 J. 15:1687-1695, 1996; Mayfield et al., J Cell Biol. 127:1537-1545, 1994; Sakamoto et al, Plant J. 6:503-512, 1994; Zerges et al., supra, 1997, each of which is incorporated herein by reference). These elements may interact with nuclear-encoded factors and generally do not resemble known prokaryotic regulatory sequences (McCarthy and Biimacombe, Trends Genet. 10:402-407, 1994).
- Consensus prokaryotic RBS elements feature a Shine-Dalgarno (SD) sequence, which is a sequence containing three to nine nucleotides, including generally about 4, 5 or 6 nucleotides that are complementary to the 3' end of the 16S rRNA.
- SD Shine-Dalgarno
- the 30S ribosomal subunit binds the mRNA at the SD sequence by virtue of the complementary anti-SD sequence within the 16S rRNA. Because the SD sequence in prokaryote mRNAs is located 5 to 15 nucleotides upstream of the initiation codon, the 3 OS ribosomal subunit is positioned such that the proper initiation codon resides within the ribosomal P site.
- chloroplast mRNAs contain elements resembling prokaryotic RBS elements (Bonham-Smith and Bourque, Nucl. Acids Res. 17:2057-2080, 1989; Ruf and K ⁇ ssel, FEBSLett. 240:41-44, 1988, each of which is incorporated herein by reference).
- RBS sequences have been unclear because these elements are often located further upstream of the start codon than is typically observed in prokaryotes.
- alteration of a putative RBS in the 5'UTR's of chloroplast mRNAs was reported to affect translation (Betts and Spremulli, J Biol. Chem.
- RBS element Because a pre-initiation complex can form at this RBS element, it has the characteristics of a bonafide recognition site for the 3 OS ribosomal subunit. However, the RBS element is unable to correctly define the translational start site in the absence of additional factors, which include nuclear-encoded translational activator proteins (Danon and Mayfield, 1991; Yohn et al, 1998a; Yohn et al, 1998b). This result indicates that the additional distance between the RBS and the initiation codon in the psbA mRNA accommodates additional translation factors, as exemplified by function of the RBS elements in chloroplasts to promote translation initiation in conjunction with light- regulated trans-acting factors.
- additional factors include nuclear-encoded translational activator proteins
- the invention provides an isolated ribonucleotide sequence that includes a first RBS operatively linked to a second RBS.
- first and second RBS generally are spaced apart by about 5 to 25 nucleotides such that, when the ribonucleotide sequence is operatively linked to a polynucleotide encoding a polypeptide, the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast.
- RBS is active in translation in chloroplasts, including allowing polysome formation, when it is positioned at least about 19 nucleotides upstream (5') of the initiator AUG codon, whereas positioning the RBS closer to the AUG results a loss of translation activity in chloroplasts (see Figure 4).
- the RBS (SD sequence) of the psbA mRNA begins at position -27 (i.e., following position -27 upstream of the AUG codon).
- An isolated ribonucleotide sequence of the invention generally is about 11 to 50 nucleotides in length, and can be about 15 to 40 nucleotides in length or about 20 to 30 nucleotides. Such a length allows for two SD sequences, which generally are about 3 to 9 nucleotides in length, usually about 4 to 7 nucleotides in length, to be spaced apart by about 5 to 25 nucleotides (generally by about 10 to 20 nucleotides, and particularly by about 15 nucleotides).
- a ribonucleotide sequence of the invention can include a first RBS of 4 nucleotides, e.g., GGAG, spaced apart by 5 nucleotides from a second of about 4 nucleotides, e.g., GGAG, thus providing a ribonucleotide sequence of 13 nucleotides in length.
- GGAG a first RBS of 4 nucleotides
- GGAG spaced apart by 5 nucleotides from a second of about 4 nucleotides, e.g., GGAG, thus providing a ribonucleotide sequence of 13 nucleotides in length.
- Each of the first RBS and the second RBS independently can have any sequence characteristic of a SD sequence.
- an RBS useful for directing translation in a plant chloroplast is complementary to at least three, particularly, four, five, or six, or more, of the anti-SD sequence at the 3' end of 16S rRNA (3'-CUUCCUCCAC-5'; SEQ ID NO:29), particularly complementary to the central eight nucleotides of the anti-SD sequence.
- RBS sequences comprising GGAG, GGAGG, o ⁇ ACGAGA (nucleotides complementary to SEQ ID NO:29 in italics) directed translation in plant chloroplasts, when operatively linked to an encoded polypeptide.
- An RBS useful in preparing a composition of the invention or in practicing a method of the invention can be chemically synthesized, or can be isolated from a naturally occurring nucleic acid molecule.
- an RBS that directs translation in a chloroplast generally is present in the 5'UTR of a chloroplast gene and, therefore, can be isolated from a chloroplast gene.
- additional nucleotide sequences as are normally associated with the SD sequence in the gene.
- a 5'UTR can include transcriptional regulatory elements such as a promoter, thus facilitating construction of a recombinant nucleic acid molecule that can be transcribed and translated in a plant chloroplast.
- a ribonucleotide of the invention containing an RBS that directs translation in a chloroplast can further contain a 5'UTR of a chloroplast gene, for example, a 5'UTR of a chloroplast gene that encodes a soluble protein, or a 5'UTR of a gene encoding a membrane-bound chloroplast protein.
- 5'UTRs are well known in the art and include those encoded by chloroplast genes encoding soluble proteins, for example, an AtpA 5'UTR (SEQ ID NO:4) or a RbcL 5'UTR (SEQ ID NO:5), and those encoded by chloroplast genes encoding membrane bound proteins, for example, a PsbD 5'UTR (SEQ ID NO:6), or a PsbA 5'UTR (SEQ ID NO:7).
- SEQ ID NO:4 an AtpA 5'UTR
- RbcL 5'UTR SEQ ID NO:5
- membrane bound proteins for example, a PsbD 5'UTR (SEQ ID NO:6), or a PsbA 5'UTR (SEQ ID NO:7).
- a 16S rRNA 5'UTR (SEQ ID NO:8) can be used, for example, to direct transcription of an operatively linked heterologous polynucleotide, and can be modified at the sequence complementary to the anti-SD sequence to generate an RBS that is particularly useful for directing translation of a polypeptide encoded by the polynucleotide in plant chloroplasts.
- a ribonucleotide sequence of the invention can further include an initiation codon, for example, an initiator AUG codon, operatively linlced to the first and second RBS.
- Such an initiator AUG codon can further include adjacent nucleotides of a Kozak sequence, for example, ACCAUGG or GCCAUGG or CC(A/G)CCAUGG or the like (see Kozak, J. Mol. Biol. 196:947-950, 1987, which is incorporated herein by reference), which can facilitate translation of an encoded polypeptide in a cell.
- the ribonucleotide sequence of the invention can be operatively linked to a polynucleotide encoding a polypeptide, wherein the polynucleotide contains an initiation codon, which can, but need not, be an endogenous initiation codon, or can be modified to contain an initiation codon.
- An isolated ribonucleotide sequence of the invention can be chemically synthesized, or can be generated using an enzymatic method, for example, from a DNA or RNA template using a DNA dependent RNA polymerase or an RNA dependent RNA polymerase, respectively.
- a DNA template encoding the ribonucleotide of the invention can be chemically synthesized, can be isolated from a naturally occurring DNA molecule, or can be derived from a naturally occurring DNA sequence that is modified to have the required characteristics.
- a DNA sequence of a prokaryote gene normally has nucleotide sequence encoding an RBS positioned about 5 to 15 nucleotides upstream an initiation codon.
- nucleotide sequence can be isolated and modified using routine recombinant DNA methods to contain a second RBS appropriately position upstream (5') of the endogenous prokaryote RBS. Accordingly, the present invention provides a polynucleotide encoding an operatively linlced first RBS and second RBS as defined herein.
- a polynucleotide encoding a first RBS operatively linked to a second RBS, wherein the first RBS can direct translation in a prokaryote and the second RBS can direct translation in a chloroplast, can be DNA or RNA, and can be single stranded or double stranded.
- the polynucleotide also can include an initiation codon, e.g., ATG, operatively linlced to the nucleotide sequence encoding the first RBS and second RBS, i.e., an ATG codon positioned about 3 to 15 nucleotides, including about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 nucleotides, downstream (3') of the first RBS, which directs translation in a prokaryote.
- initiation codon e.g., ATG
- ATG operatively linlced to the nucleotide sequence encoding the first RBS and second RBS, i.e., an ATG codon positioned about 3 to 15 nucleotides, including about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 nucleotides, downstream (3') of the first RBS, which directs translation in a prokaryote.
- a polynucleotide of the invention also can include a cloning site that is positioned to allow operative linkage of an expressible polynucleotide, which can encode a polypeptide, to the first RBS and second RBS, and to an ATG codon if present, such that the polypeptide can be expressed in a chloroplast or in a prokaryote host cell.
- cloning site is used broadly to refer to any nucleotide or nucleotide sequence that facilitates linkage of a first polynucleotide to a second polynucleotide.
- a cloning site comprises one or a plurality restriction endonuclease recognition sites, for example, a multiple cloning site, or one or a plurality of recombinase recognition sites, for example, a loxP site or an att site, or a combination of such sites.
- the cloning site can be provided to facilitate insertion or linkage, which can be operative linkage, of the first and second polynucleotide, for example, a first polynucleotide encoding a first RBS operatively linked to a second RBS to a second polynucleotide encoding a polypeptide of interest, which is to be translated in a prokaryote or a chloroplast or both.
- a polynucleotide encoding a first and second RBS, as defined herein, can be operatively linlced to an expressible polynucleotide, which can encode at least one polypeptide, including a peptide or peptide portion of a polypeptide.
- the expressible polynucleotide can encode only a first polypeptide, or can encode two or more polypeptides, which can be the same or different as the first polypeptide.
- the expressible polynucleotide can encode a first polypeptide and a second polypeptide, which are different from each other, particularly a first and second polypeptide that can specifically associate to form a functional heterodimer such as an antibody; an enzyme; a cell surface receptor such as a T cell receptor, a growth factor receptor, a cannabinoid receptor; or the like.
- a first and second (or other) polypeptide can be expressed as a fusion protein, for example, single chain antibody comprising a H chain linlced to a L chain, or can be expressed as separate and discrete polypeptides, which can, but need not, have the ability to specifically associate to form a functional protein complex.
- polypeptides are to be expressed as separate entities, it can be useful to include a nucleotide sequence encoding an internal ribosome entry site (IRES) operatively linked between the coding sequence of the first polypeptide and the coding sequence of the second polypeptide, thus facilitating translation of the second (or downstream) polypeptide.
- IRS internal ribosome entry site
- a polynucleotide encoding a first RBS operatively linked to a second RBS can be a linear nucleotide sequence, and can be flanked at one end by a first cloning site and the second end by a second cloning site, thus providing a cassette that readily can be inserted into or linlced to a second polynucleotide.
- the flanking first and second cloning sites can be the same or different, and one or both independently can comprise a multiple cloning site.
- the polynucleotide can further include any other nucleotide sequences of interest, for example, an operatively linked initiator ATG codon.
- the present invention further provides a vector containing a polynucleotide encoding an first RBS operatively linked to a second RBS, as defined herein.
- the vector can be any vector useful for introducing a polynucleotide into a prokaryotic or eukaryotic cell, including a cloning vector or an expression vector.
- the vector comprises a nucleotide sequence of chloroplast genomic DNA sufficient to undergo homologous recombination with chloroplast genomic DNA, particularly a silent nucleotide sequence, which does not encode a chloroplast gene.
- chloroplast vectors are well known in the art and include, for example, p322 (see Example 1; see, also, Kindle et al, Proc. Natl. Acad. Sci, USA 88:1721-1725, 1991, which is incorporated herein by reference; Hager and Bock, supra, 2000; Bock, supra, 2001).
- a vector of the invention also can contain one or more additional nucleotide sequences that confer desirable characteristics on the vector, including, for example, sequences such as cloning sites that facilitate manipulation of the vector, regulatory elements that direct replication of the vector or transcription of nucleotide sequences contain therein, sequences that encode a selectable marker, and the like.
- the vector can contain, for example, one or more cloning sites such as a multiple cloning site, which can, but need not, be positioned such that a heterologous polynucleotide can be inserted into the vector and operatively linked to the first RBS and second RBS.
- the vector also can contain a prokaryote origin of replication (ori), for example, an E. coli ori or a cosmid ori, thus allowing passage of the vector in a prokaryote host cell, as well as in a plant chloroplast, as desired.
- ori prokaryote origin of replication
- regulatory element is used broadly herein to refer to a nucleotide sequence that regulates the transcription or translation of a polynucleotide or the localization of a polypeptide to which it is operatively linked.
- an expression control sequence can be a promoter, enhancer, transcription terminator, an initiation (start) codon, a splicing signal for intron excision and maintenance of a correct reading frame, a STOP codon, an amber or ochre codon, an IRES, or a sequence that targets a polypeptide to a particular location, for example, a cell compartmentalization signal, which can be useful for targeting a polypeptide to the cytosol, nucleus, plasma membrane, endoplasmic reticulum, mitochondrial membrane or matrix, chloroplast membrane or lumen, medial trans-Golgi cisternae, or a lysosome or endosome.
- Cell compartmentalization domains are well known in the art and include, for example, a peptide containing amino acid residues 1 to 81 of human type II membrane-anchored protein galactosyltransferase, or amino acid residues 1 to 12 of the presequence of subunit IV of cytochrome c oxidase (see, also, Hancock et al., EMBO J. 10:4033-4039, 1991; Buss et al., Mol. Cell. Biol. 8:3960-3963, 1988; U.S. Pat. No. 5,776,689, each of which is incorporated herein by reference).
- Inclusion of a cell compartmentalization domain in a polypeptide produced using a method of the invention can allow use of the polypeptide, which can comprise a protein complex, where it is desired to target the polypeptide to a particular cellular compartment in an individual.
- a vector or other recombinant nucleic acid molecule of the invention can include a nucleotide sequence encoding a reporter polypeptide or other selectable marker.
- reporter or selectable marker refers to a polynucleotide (or encoded polypeptide) that confers a detectable phenotype.
- a reporter generally encodes a detectable polypeptide, for example, a green fluorescent protein or an enzyme such as luciferase, which, when contacted with an appropriate agent (a particular wavelength of light or luciferin, respectively) generates a signal that can be detected by eye or using appropriate instrumentation (Giacomin, Plant Sci. 116:59-72, 1996; Scikantha, J Bacteriol.
- a selectable marker generally is a molecule that, when present or expressed in a cell, provides a selective advantage (or disadvantage) to the cell containing the marker, for example, the ability to grow in the presence of an agent that otherwise would kill the cell.
- a selectable marker can provide a means to obtain prokaryotic cells or plant cells or both that express the marker and, therefore, can be useful as a component of a vector of the invention (see, for example, Bock, supra, 2001).
- selectable markers include those that confer antimetabolite resistance, for example, dihydrofolate reductase, which confers resistance to methotrexate (Reiss, Plant Physiol. (Life Sci. Adv.) 13:143-149, 1994); neomycin phosphotransferase, which confers resistance to the aminoglycosides neomycin, kanamycin and paromycin (Henera-Estrella, EMBO J.
- dihydrofolate reductase which confers resistance to methotrexate
- methotrexate Reiss, Plant Physiol. (Life Sci. Adv.) 13:143-149, 1994
- neomycin phosphotransferase which confers resistance to the aminoglycosides neomycin, kanamycin and paromycin
- hygro which confers resistance to hygromycin
- trpB which allows cells to utilize indole in place of tryptophan
- hisD which allows cells to utilize histinol in place of histidine
- mannose-6-phosphate isomerase which allows cells to utilize mannose
- WO 94/20627 mannose-6-phosphate isomerase which allows cells to utilize mannose
- ornithine decarboxylase which confers resistance to the ornithine decarboxylase inhibitor, 2-(difluoromethyl)-DL-ornithine (DFMO; McConlogue, 1987, In: Current Communications in Molecular Biology, Cold Spring Harbor Laboratory ed.); and deaminase from Aspergillus terreus, which confers resistance to Blasticidin S (Tamura, Biosci Biotechnol. Biochem. 59:2336-2338, 1995).
- Additional selectable markers include those that confer herbicide resistance, for example, phosphinothricin acetyltransferase gene, which confers resistance to phosphinothricin (White et al., Nucl. Acids Res. 18:1062, 1990; Spencer et al., Theor. Appl. Genet.
- markers conferring resistance to an herbicide such as glufosinate include polynucleotides that confer dihydrofolate reductase (DHFR) or neomycin resistance for eukaryotic cells and tetracycline; ampicillin resistance for prokaryotes such as E.
- DHFR dihydrofolate reductase
- neomycin resistance for eukaryotic cells and tetracycline
- ampicillin resistance for prokaryotes such as E.
- compositions or a method of the invention can result in expression of a polypeptide in chloroplasts, it can be useful if a polypeptide conferring a selective advantage to a plant cell is operatively linked to a nucleotide sequence encoding a cellular localization motif such that the polypeptide is translocated to the cytosol, nucleus, or other subcellular organelle where, for example, a toxic effect due to the selectable marker is manifest (see, for example, Von Heijne et al., Plant Mol. Biol. Rep. 9: 104, 1991; Clark et al., J Biol. Chem. 264:17544, 1989; della Cioppa et al., Plant Physiol.
- a shuttle vector of the invention in a prokaryote allows for conveniently manipulating the vector.
- a reaction mixture containing the vector and putative inserted polynucleotides of interest can be transformed into prokaryote host cells such as E. coli, amplified and collected using routine methods, and examined to identify vectors containing an insert or construct of interest.
- the vector can be further manipulated, for example, by performing site directed mutagenesis of the inserted polynucleotide, then again amplifying and selecting vectors having a mutated polynucleotide of interest.
- the shuttle vector then can be introduced into plant cell chloroplasts, wherein a polypeptide of interest can be expressed and, if desired, isolated according to a method of the invention.
- a polynucleotide or recombinant nucleic acid molecule of the invention which can be contained in a vector, including a vector of the invention, can be introduced into plant chloroplasts using any method known in the art.
- the term "introducing” means transfening a polynucleotide into a cell, including a prokaryote or a plant cell, particularly a plant cell plastid.
- a polynucleotide can be introduced into a cell by a variety of methods, which are well known in the art and selected, in part, based on the particular host cell.
- the polynucleotide can be introduced into a plant cell using a direct gene transfer method such as electroporation or microprojectile mediated (biolistic) transformation using a particle gun, or the "glass bead method" (see, for example, Kindle et al., supra, 1991), or by pollen-mediated transformation, liposome- mediated transformation, transformation using wounded or enzyme-degraded immature embryos, or wounded or enzyme-degraded embryogenic callus (see Potrykus, Ann. Rev. Plant. Physiol. Plant Mol. Biol. 42:205-225, 1991, which is incorporated herein by reference).
- a direct gene transfer method such as electroporation or microprojectile mediated (biolistic) transformation using a particle gun, or the "glass bead method” (see, for example, Kindle et al., supra, 1991), or by pollen-mediated transformation, liposome- mediated transformation, transformation using wounded or enzyme-degraded immature embryos, or wounded or enzyme-degrade
- Plastid transformation is a routine and well known method for introducing a polynucleotide into a plant cell chloroplast (see U.S. Pat. Nos. 5,451,513, 5,545,817, and 5,545,818; WO 95/16783; McBride et al., Proc. Natl. Acad. Sci, USA 91:7301-7305, 1994, each of which is incorporated herein by reference).
- Chloroplast transformation involves introducing regions of chloroplast DNA flanking a desired nucleotide sequence into a suitable target tissue, using, for example, a biolistic or protoplast transformation method (e.g., calcium chloride or PEG mediated transformation).
- One to 1.5 kb flanking nucleotide sequences of chloroplast genomic DNA allow homologous recombination of the vector with the chloroplast genome, and allow the replacement or modification of specific regions of the plastome.
- point mutations in the chloroplast 16S rRNA and rpsl2 genes which confer resistance to spectinomycin and streptomycin, can be utilized as selectable markers for transformation (Svab et al., Proc. Natl. Acad. Sci, USA 87:8526-8530, 1990; Staub and Maliga, supra, 1992), and can result in stable homoplasmic transformants, at a frequency of approximately one per 100 bombardments of target leaves.
- Plastid expression in which genes are inserted by homologous recombination into all of the several thousand copies of the circular plastid genome present in each plant cell, takes advantage of the enormous copy number advantage over nuclear-expressed genes to permit expression levels that can readily exceed 10%) of the total soluble plant protein.
- a direct gene transfer method such as electroporation also can be used to introduce a polynucleotide of the invention into a plant protoplast (Fromm et al., Proc. Natl. Acad. Sci, USA 82:5824, 1985, which is incorporated herein by reference). Electrical impulses of high field strength reversibly permeabilize membranes allowing the introduction of the polynucleotide. Electroporated plant protoplasts reform the cell wall, divide and form a plant callus. Microinjection can be performed as described in Potrykus and Spangenberg (eds.), Gene Transfer To Plants (Springer Verlag, Berlin, NY 1995). A transformed plant cell containing the introduced polynucleotide can be identified by detecting a phenotype due to the introduced polynucleotide, for example, expression of a reporter gene or a selectable marker.
- Microprojectile mediated transformation also can be used to introduce a polynucleotide into a plant cell chloroplast (Klein et al., Nature 327:70-73, 1987, which is incorporated herein by reference).
- This method utilizes microprojectiles such as gold or tungsten, which are coated with the desired polynucleotide by precipitation with calcium chloride, spermidine or polyethylene glycol.
- the microprojectile particles are accelerated at high speed into a plant tissue using a device such as the BIOLISTIC PD-1000 particle gun (BioRad; Hercules CA). Methods for the transformation using biolistic methods are well known (Wan, Plant Physiol.
- Microprojectile mediated transformation has been used, for example, to generate a variety of transgenic plant species, including cotton, tobacco, corn, hybrid poplar and papaya.
- Important cereal crops such as wheat, oat, barley, sorghum and rice also have been transformed using microprojectile mediated delivery (Duan et al., Nature Biotech. 14:494-498, 1996; Shimamoto, Curr. Opin. Biotech. 5:158-162, 1994).
- the transformation of most dicotyledonous plants is possible with the methods described above.
- Transformation of monocotyledonous plants also can be transformed using, for example, biolistic methods as described above, protoplast transformation, electroporation of partially permeabilized cells, introduction of DNA using glass fibers, the glass bead agitation method (Kindle et al., supra, 1991), and the like.
- the present invention also provides a vector that includes a nucleotide sequence encoding an RBS positioned about 20 to 40 nucleotides 5' to a cloning site.
- the cloning site can be any nucleotide sequence that facilitates insertion or linkage of a heterologous nucleotide sequence into the vector, for example, one or more restriction endonuclease recognition sites, one or more recombinase recognition sites, or a combination of such sites.
- the cloning site is a multiple cloning site, which includes a plurality of restriction endonuclease recognition sites or recombinase recognition sites, or a combination of at least one restriction endonuclease recognition site and at least one recombinase recognition site.
- the vector can further contain an initiation codon or a portion thereof adjacent and 5' to the cloning site, thus providing a translation start site (or cryptic start site) for a coding sequence that otherwise lacks an initiator ATG codon or contains a partial initiation codon due, for example, to cleavage by a restriction endonuclease.
- the vector also can contain a chloroplast gene 3'UTR positioned 3' to the cloning site, for example, PsbA 3'UTR (SEQ ID NO:9), a RbcL 3'UTR (SEQ ID NO: 10), an AtpA 3'UTR (SEQ ID NO: 11), a tRNA ⁇ 0 3'UTR (SEQ ID NO: 12), or a PsbD 3'UTR (see SEQ ID NO:30, beginning at position 1553; also showing insertion site for GFP construct encoding PsbD-GFP fusion protein).
- a chloroplast gene 3'UTR positioned 3' to the cloning site, for example, PsbA 3'UTR (SEQ ID NO:9), a RbcL 3'UTR (SEQ ID NO: 10), an AtpA 3'UTR (SEQ ID NO: 11), a tRNA ⁇ 0 3'UTR (SEQ ID NO: 12), or a PsbD 3
- a shuttle vector of the invention can be made, for example, by introducing into a nucleotide sequence of chloroplast genomic DNA sufficient to undergo homologous recombination with chloroplast genomic DNA, a nucleotide sequence comprising a prokaryote origin of replication; a nucleotide sequence encoding a first RBS; and a nucleotide sequence encoding a second RBS, wherein the first RBS and second RBS are spaced apart by about 5 to 25 nucleotides; and a cloning site, wherein the cloning site is positioned to allow operative linkage of a polynucleotide encoding a polypeptide to the first RBS and second RBS such that the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast.
- a method of making a chloroplast/prokaryote shuttle expression vector also can be performed by genetically modifying a nucleotide sequence of chloroplast genomic DNA, which is sufficient to undergo homologous recombination with chloroplast genomic DNA, to contain a prokaryote origin of replication, a nucleotide sequence encoding a first RBS spaced apart from a second RBS by about 5 to 25 nucleotides, and a cloning site positioned to allow operative linkage of a polynucleotide encoding a polypeptide to the first RBS and second RBS such that the first RBS can direct translation of the polypeptide in a prokaryote and the second RBS can direct translation of the polypeptide in a chloroplast.
- the present invention also provides a chloroplast/prokaryote shuttle vector produced by a method as disclosed herein.
- the invention also provides a recombinant nucleic acid molecule, which includes a first nucleotide sequence encoding chloroplast RBS operatively linked to a second nucleotide sequence encoding a polypeptide, wherein the first nucleotide sequence is heterologous with respect to the second nucleotide sequence.
- An operatively linlced RBS generally is positioned about 20 to 40 nucleotides 5' (upstream) to an initiation codon, which, in turn, is operatively linked to the nucleotide sequence encoding the polypeptide.
- the first nucleotide sequence comprises an ATG codon positioned about 20 to 40 nucleotides 3' of nucleotide sequence encoding the RBS.
- a recombinant nucleic acid molecule of the invention can further include other regulatory elements or encoding polynucleotides of interest, as exemplified herein or otherwise known in the art.
- Reporter genes have been successfully used in chloroplasts of higher plants, and high levels of recombinant protein expression have been reported. In addition, reporter genes have been used in the chloroplast of C. reinhardtii, but, in most cases very low amounts of protein were produced. Reporter genes greatly enhance the ability to monitor gene expression in a number of biological organisms. In chloroplasts of higher plants, ⁇ -glucuronidase (uidA, Staub and Maliga, EMBO J. 12:601-606, 1993), neomycin phosphotransferase (nptll, Carrer et al., Mol. Gen. Genet.
- heterologous proteins have been expressed in the chloroplasts of higher plants such as Bacillus thuringiensis Cry toxins, conferring resistance to insect herbivores (Kota et al, Proc. Natl. Acad. Sci, USA 96:1840-1845, 1999), or human somatotropin (Staub et al., Nat. Biotechnol. 18:333-338, 2000), a potential biopharmaceutical.
- Several reporter genes have been expressed in the chloroplast of the eukaryotic green alga, C. reinhardtii, although with varying degrees of success. These include aadA (Goldschmidt-Clermont, Nucl. Acids Res.
- codon bias on heterologous protein expression is well documented in both prokaryotic and eukaryotic organisms, and even viral genes display a codon bias that can affect their temporal, and tissue specific expression.
- codon usage is conelated with the level of iso-accepting tRNAs.
- genes encoding highly expressed proteins tend to utilize codons whose levels of cognate tRNAs are particularly abundant (Duret, supra, 2000; Kanaya et al., Gene 238:143-155, 1999).
- the C. reinhardtii chloroplast genome displays a strong codon bias, with adenine or uracil (or thymine) preferred at the third position (Nakamura et al., supra, 1999).
- the role of chloroplast codon usage in expression of recombinant polypeptides in the C. reinhardtii chloroplasts was examined by synthesizing de novo a polynucleotide that encodes GFP and is biased for chloroplast codon usage of the major C. reinhardtii chloroplast encoded proteins (Example 1). GFP accumulation was monitored in C.
- the present invention provides an isolated synthetic polynucleotide encoding a fluorescent protein or a mutant or variant thereof, wherein codons of the polynucleotide are biased to reflect chloroplast codon usage.
- the synthetic polynucleotide can be DNA or RNA, can be single stranded or double stranded, and can be a linear polynucleotide containing a cloning site at one or both ends.
- the polynucleotide which can be contained in a vector, also can be operatively linked to a polynucleotide encoding a first RBS and a second RBS that are spaced apart by about 5 to 25 nucleotides, such that the fluorescent protein conveniently can be translated in a prokaryote and in a chloroplast.
- Table 1 exemplifies codons that are preferentially used in alga chloroplast genes.
- chloroplast codon usage is used herein to refer to such codons, and is used in a comparative sense with respect to degenerate codons that encode the same amino acid but are less likely to be found as a codon in a chloroplast gene.
- bias when used in reference to chloroplast codon usage, refers to the manipulation of a polynucleotide such that one or more nucleotides of one or more codons is changed, resulting in a codon that is preferentially used in chloroplasts.
- Chloroplast codon bias is exemplified herein by the alga chloroplast codon bias as set forth in Table 1.
- the chloroplast codon bias can, but need not, be selected based on a particular plant in which a synthetic polynucleotide is to be expressed.
- the manipulation can be a change to a codon, for example, by a method such as site directed mutagenesis, by a method such as PCR using a primer that is mismatched for the nucleotide(s) to be changed such that the amplification product is biased to reflect chloroplast codon usage, or can be the de novo synthesis of polynucleotide sequence such that the change (bias) is introduced as a consequence of the synthesis procedure.
- chloroplast codon bias As a means to provide efficient translation of a polypeptide, it will be recognized that an alternative means for obtaining efficient translation of a polypeptide in a chloroplast to re-engineer the chloroplast genome (e.g., a C. reinhardtii chloroplast genome) for the expression of tRNAs not otherwise expressed by in the chloroplast genome.
- a chloroplast genome e.g., a C. reinhardtii chloroplast genome
- Such an engineered algae expressing one or more heterologous tRNA molecules provides the advantage that it would obviate a requirement to modify every polynucleotide of interest that is to be introduced into and expressed from a chloroplast genome; instead, algae such as C.
- reinhardtii that comprise a genetically modified chloroplast genome can be provided and utilized for efficient translation of a polypeptide according to a method of the invention.
- Correlations between tRNA abundance and codon usage in highly expressed genes is well known (Franklin et al., Plant J. 30:733-744, 2002; Dong et al, J. Mol Biol. 260:649-663, 1996; Duret, Trends Genet. 16:287-289, 2000; Goldman et. al, J Mol. Biol. 245:467-473, 1995; Komar et. al., Biol. Chem. 379:1295-1300, 1998, each of which is incorporated herein by reference.
- coli for example, re-engineering of strains to express underutilized tRNAs resulted in enhanced expression of genes which utilize these codons (see Novy et al., inNovations 12:1-3, 2001, which is incoiporated herein by reference).
- site directed mutagenesis can be used to make a synthetic tRNA gene, which can be introduced into chloroplasts to complement rare or unused tRNA genes in a chloroplast genome such as a C. reinhardtii chloroplast genome.
- One or more codons encoding a fluorescent protein of the invention can be biased, for example, to contain an adenine or a thymine at position three, thus facilitating translation of the fluorescent protein in a chloroplast.
- the polynucleotide encoding Aequorea victoria GFP was biased by de novo synthesis of an encoding sequence having 121 synonymous codon changes, including 66 changes that represent a modest shift toward chloroplast codon usage and 54 changes that resulted in an infrequently used codon being shifted toward chloroplast codon usage (Example 1).
- polynucleotide set forth as SEQ ID NO:l which encodes a modified GFP (SEQ ID NO:2)
- SEQ ID NO:2 provides an example of a polynucleotide of the invention
- polynucleotides that encode SEQ ID NO:2 but have fewer biased codons provide additional examples.
- the modified GFP having an amino acid sequence as set forth in SEQ ID NO:2.
- GFPs are well known in the ait and have been isolated from the Pacific Northwest jellyfish, Aequorea victoria, the sea pansy, Renilla reniformis, and Phialidium gregarium (Ward et al., Photochem. Photobiol. 35:803-808, 1982; Levine et al., Comp. Biochem. Physiol. 72B:77-85, 1982, each of which is incorporated herein by reference).
- red fluorescent proteins are known and have been isolated from the coral, Discosoma (Matz et al., Nature Biotechnoh 17:969-973, 1999, which is incorporated herein by reference).
- Cultivation of C. reinhardtii transformants for expression of GFP was carried out in TAP medium (Gorman and Levine, supra, 1965) at 23 °C under constant illumination of 5,000 Lux on a rotary shaker set at 100 rpm, unless stated otherwise. Cultures were maintained at a density of 1 x 10 7 cells per ml for at least 48 hr prior to harvest.
- the sequence for the 5' GFPncb was 5'-CATATGAGTAAAGGAGAAGAAC-3' (SEQ ID NO:17); the sequence for the 3' GFPct primer was 5'-TCTAGATTATTTGTATAGTTCATCC-3' (SEQ ID NO: 18).
- the coding region of the GFPct gene was synthesized de novo as described by Stemmer et al., Gene 164:49-53, 1995, which is incorporated herein by reference) from a pool of primers, each 40 nucleotides in length.
- the 5' terminal and 3' terminal primers contained restriction sites for Nde I and Xba I, respectively.
- the resulting 717 bp PCR products containing the GFPct and GFPncb genes were cloned into plasmid pCR2.1 TOPO (Invitrogen, Inc.) according to the manufacturers protocol to generate plasmids pCrG Pct and pCrGFPncb respectively.
- the rbcL 3' UTR was generated via PCR using a 1.6 kb Hind III fragment of C. reinhardtii chloroplast genomic DNA, cloned into plasmid pUC19, as the template.
- the sequence of the PCR primer, corresponding to the 5' end of the rbcL 3' UTR and a portion of the pUC19 polylinker, including the Xba I site was 5'-TCTAGAGTCGACCTGCAG-3' (SEQ ID NO.T9).
- the sequence of the PCR primer, corresponding to the 3' end of the rbcL 3' UTR was 5'-GGATCCGTCGACGTATG-3* (SEQ ID NO:20), and includes a Bam HI restriction site for subsequent cloning.
- the resulting 433 bp product was cloned into plasmid pCR2.1 TOPO to generate plasmid p3rbcL.
- the rbcL 5' UTR was generated by PCR using C. reinhardtii genomic DNA as template.
- the sequence of the PCR primer, complementary to the 5' end of the rbcL gene beginning at position -189 relative to the translational start site was 5'-GAATTCATATACCTAAAGGCCCTTTCTATGC-3' (SEQ ID NO:21), and contains an Eco Rl restriction site.
- the PCR primer complementary to the 3' end of the rbcL 5'UTR begins at the translation initiation site and had the sequence 5'-CATATGTATAAATAAATGTAACTTC-3' (SEQ ID NO:22), and contains a Nde I restriction site.
- the resulting 241 bp PCR product was cloned into the pCR2.1 TOPO vector to generate plasmid p5rbcL.
- the plasmid p5rbcL was digested with Bam HI and Nde I and the resulting fragment was ligated into either pCrGFPct or pCr GFPncb digested with Bam HI and Nde I to generate plasmids p5CrGFPct and p5CxGFPncb respectively.
- p5CrGFPct and ⁇ p5CtGFPncb were digested with Bam HI and Xba I and the resulting 958 bp fragments were ligated into p3rbcL, also digested with Bam HI and Xba I, to generate plasmids ⁇ 3vGFPct and p53rGFPncb.
- Both p53rGFPct and ⁇ 53 ⁇ GFPncb were digested with Nde I and Bam HI and the 1.2 kb fragments were ligated into pET19b (Novagen) to generate plasmids pETG ct and x E ⁇ GFPncb, respectively, for expression in E. coli.
- p53rGFPct and p53xGFPncb were next digested with Bam HI and the 1.43 kb fragments were ligated into the C. reinhardtii chloroplast transformation vector, p322 (Chlamydomonas Genetics Center, Duke University) to form plasmids pExGFPct and pExGFPncb.
- the p322 vector is based on the nucleotide sequence of the C. reinhardtii chloroplast genomic DNA sequence extending from the Eco (Eco Rl) site beginning at position 143,073 to the Xho (Xho I) site beginning at position 148,561 (see, world wide web, at the URL "biology.duke.edu/chlamy_genome/chloro.html", and clicking on "view complete genome as text file”; see, also, “maps of the chloroplast genome” link, then "140-150 kb” link for Eco site at about 143.1 kb and Xho site at about 148.5 kb).
- the Eco/Xho chloroplast genome sequence was inserted into Eco Rl Xho I digested the pBS plasmid (Stratagene Corp., La Jolla CA).
- the Bam HI site in p322 corresponds to that beginning at position 146522 of the chloroplast genomic DNA sequence.
- Southern blots and 32 P labeling of DNA for use as probes were carried out as described in Sambrook et al., supra, 1989).
- Radioactive probes used on Southern blots included the 2.2 b Bam Hl/Pst I fragment of p322 (probe 5' ⁇ 322), the 2.0 kb Bam Hl/Xho I fragment of p322 (probe 3' p322) and the 717 bp Nde I/Xba I fragments from p53xGFPct (probe GFPct) ox p53xGFPncb (probe GFPncb).
- Plasmids pETG Pct and pETGFPncb were transformed into E. coli strain BL21 and 6 His-tagged GFPct or GFPncb protein expression induced by IPTG according to the manufacturer's protocol (Novagen). Purification of His-tagged proteins was carried out using Ni-agarose affinity chromatography (Qiagen). Western blots were carried out as described in Cohen et al. (supra, 1998) using a mouse anti GFP primary antibody (Clontech) and an alkaline phosphatase labeled anti-mouse secondary antibody (Sigma). Fluorescence gels were run as for gels intended for Coomassie staining or western transfer, except that proteins were not boiled prior to loading. GFP was visualized in gels by viewing with a Berthold Night Owl CCD camera, model LB 981, equipped with 485 nm excitation and 535 nm emission filters (Chroma Corp.). Images were generated using WinLight software.
- Excitation spectra were generated with affinity purified GFPct or GFPncb proteins on a Perkin Elmer Luminescence Spectrometer Model LS50. Recombinant proteins were diluted in 50 mM NaH 2 PO 4 , 300 mM NaCl, 250 mM imidazole, pH 8.0, prior to reading on the spectrometer. Excitation spectra were generated by scanning illumination from 350 to 550 nm, while monitoring emission at 510 nm.
- the second change a serine to threonine change at amino acid position 65 was made to enhance the amplitude of excitation at 485 nm relative to native GFP (approximately 6 fold), while at the same time reducing excitation at 395 nm (Heim et al., Nature 373:663-664, 1995, which is incorporated herein by reference).
- This change was introduced into the GFPct coding sequence to improve fluorescent detection using visible light.
- E. coli cell lysates prepared from cells transformed with either pETGFPct or pETGFPncb were examined.
- Ni affinity chromatography of E. coli lysates produced proteins of the conect molecular mass for GFP.
- Direct fluorescence assays of SDS PAGE separated E. coli produced proteins revealed that both proteins fluoresced under blue light illumination, and showed slightly different fluorescent properties consistent with the introduced amino acid changes.
- the S65T alteration to the GFPct protein resulted in greatly enhanced level of fluorescence at 485nm (only 1/5 the amount of E.
- GFPct protein was used in this assay relative to GFPncb protein), while its fluorescence at 395nm excitation is greatly reduced (see Figure 2).
- Western blot analysis using a mouse polyclonal antibody raised against native GFP showed a similar signal for both GFPct and GFPncb. This result is particularly important given that the spectral qualities of the GFPct protein was intentionally enhanced relative to the GFPncb protein.
- fluorescence detection based upon excitation in the visible (485nm), would favor GFPct detection, immunolabeling is nondiscriminatory, allowing for the direct comparison of GFPct and GFPncb protein accumulation in C. reinhardtii chloroplasts.
- C. reinhardtii chloroplasts were transformed with pExGFPct and pExGFPncb.
- the cells were cotransformed with the selectable marker plasmid, p228, which confers resistance to spectinomycin.
- Primary transformants were screened by PCR followed by Southern blot analysis, and positive transformants were taken through additional rounds of selection to isolate homoplasmic lines, in which all copies of the chloroplast genome contained the introduced GFP gene.
- the filters were stripped and re-probed with the GFPct and GFPncb specific probes. Strains 5.8 and 12.1 accumulated GFPncb mRNA, while strains 18.3 and 21.2 accumulated GFPct mRNA. No GFP signal was observed in wt cells, as expected. All four cDNA probes were labeled to approximately the same specific activity, and while the GFPct and GFPncb signals were similar, both GFP probed filters required longer exposures (approximately four times) to obtain a similar signal to the rbcL probe. These results indicate that the GFP mRNAs accumulate to roughly one quarter the level of the endogenous rbcL mRNA.
- GFP was measured by both fluorescence and western blot analysis. Comparison of GFP accumulation in C. reinhardtii transgenic strain 21.2 expressing GFPct, and strains 5.8 and 12.1, both expressing GFPncb. Cells were grown to a density of 1 x 10 7 cells per ml under continuous light (5,000 lux), conditions known to allow maximal accumulation of GFP. Total soluble protein was subjected to SDS-PAGE, followed by western blot analysis with anti-GFP antisera. Twenty ⁇ g of total soluble protein was loaded for GFPncb transgenic strains 5.8 and 12.1, while 250 ng (1/80) to 20 ⁇ g (1/1) total soluble protein was loaded for GFPct transgenic 21.2.
- C. reinhardtii GFPct transgenic strain 21.2 was maintained under constant illumination at a density of 1 x 10 6 cells per ml at either 5,000 lux (high light) or 450 lux (low light), prior to harvesting.
- Western blot analysis was carried out on 1 ⁇ g tsp from each treatment. The effect of light intensity on accumulation of GFPct in C. reinhardtii was examined. Prior to harvest, C.
- reinhardtii transgenic line 21.2 was maintained at either 1 x 10 6 cells per ml or 1 x 10 7 cells per ml for at least 48 hr under constant illumination at the indicated light intensity.
- Total soluble protein (1 ⁇ g) was subjected to 12% SDS-PAGE and western blotting with anti-GFP primary antibody.
- heterologous genes have been employed as reporters of chloroplast gene expression in C. reinhardtii, but their utility has been limited due to low levels of protein expression.
- the promoters used to drive transcription of these genes may result in low levels of transcription.
- some of these reporter mRNAs may be inherently unstable, resulting in low levels of mRNA accumulation.
- RNA elements required for translation may be lacking from these chimeric mRNAs. Strong codon bias in C. reinhardtii chloroplast genes also may preclude the translation of heterologous mRNAs.
- GFP coding region of GFP was engineered to match the codon usage of protein coding sequences from the C. reinhardtii chloroplast genome. Expression of this GFPct gene, as well as a native GFP gene (GFPncb), was placed under the control of the C. reinhardtii chloroplast rbcL 5' and 3' UTRs. Both the GFPncb gene and the GFPct gene were transcribed and accumulated mRNA to similar levels in transgenic C. reinhardtii chloroplasts.
- Transgenic strains expressing GFPct accumulated approximately 80 fold more GFP than those expressing GFPncb.
- the GFPct producing strain 21.2 accumulated GFP to approximately 0.5 % of the total soluble protein, under optimal growth conditions. This level of protein expression allows for analysis of GFP expression by fluorescence assays of total cellular proteins. Previous reports of uidA (GUS)expression in C.
- reinhardtii chloroplast under the control of the rbcL 5' and 3' UTRs showed low levels of protein expression, approximately 0.01% of soluble protein; this level of GUS accumulation was similar to the level of GFP accumulation obtained with the GFPncb gene using the same rbcL control elements (Ishikura et al., supra, 1999, also reporting relatively low levels of rbcL-GUS mRNA accumulation) (similar to the low levels for rbcL GFP mRNA, as disclosed herein).
- the results disclosed herein demonstrate that optimizing codon usage can facilitate translation and expression of a polypeptide, as exemplified by the optimized GFPct gene, which was used as a reporter of chloroplast gene expression C. reinhardtii.
- the demonstration that codon optimization can be used to achieve high levels of recombinant protein expression in C. reinhardtii indicates that codon optimization generally can contribute to translation efficiency of other heterologous polypeptides in plant chloroplasts.
- the relatively low levels of GFP mRNA accumulation as compared to the endogenous rbcL mRNA indicates that optimizing promoter activity and mRNA stability of GFPct can provide a means to enhance the signal of GFPct to even higher or more desirable levels.
- the GFPct gene provides a tool that is useful to conveniently optimize transcription, mRNA stability and translation of GFP in plant chloroplasts, including in C. reinhardtii chloroplasts.
- the pellets were resuspended in TKMD buffer containing 100 mM KCl and frozen in liquid nitrogen for storage at -70°C.
- the degree of cross contamination of the 3 OS and 5 OS subunits was assayed using RNA blot analysis (Cohen et al, supra, 1998).
- heparin-agarose purified protein (Cohen et al., 1998) was incubated with 0.4 units PRIME RNase Inhibitor (5 Prime ⁇ -3 Prime, Inc.) for 10 min at room temperature in a total volume of 8 ⁇ l dialysis buffer (20 mM Tris-HCl (pH 7.5), 100 mM KOAc, 0.2 mM EDTA (pH 8.0), 2 mM DTT, 20% glycerol, 4 mM MgCl 2 ).
- the reaction was incubated at room temperature for 10 min upon addition of 0.04 pmol of in vitro transcribed ( 32 P)-labeled/7,s ⁇ /4 RNA, spanning the positions -90 to +171 relative to the translation start codon, 20 ⁇ g of wheat germ tRNA (Sigma), and 3 ⁇ g of FuD7 (a C. reinhardtii strain lacking psbA mRNA) total RNA.
- 10 pmol unlabeled in vitro transcribed xmlabe ⁇ e ⁇ psbA RNA was added as a competitor. RNA/protein complexes were separated in a 5% non-denaturing polyacrylamide gel.
- Chloroplast 30S ribosomal subunits recognize a Shine-Delgarno ribosome binding sequence in the psbA 5'UTR
- RNA elements required for chloroplast mRNA translation variant psbA genes containing site-specific mutations within the 5'UTR were introduced into chloroplasts of a psbA-deficient strain of C. reinhardtii (Mayfield et al., supra, 1994).
- a potential RBS within the psbA 5'UTR located 27 nucleotides upstream of the start codon was identified based on its potential to recognize the anti-SD sequence within the chloroplast 16S rRNA.
- SD sequences within RBS elements promote the initiation of translation from prokaryotic transcripts by pairing to a complementary sequence (anti-SD sequence) at the 3' end of the 16S rRNA of the 3 OS small ribosomal subunit (Voorma, In Translational Control(ed. Hershey et al., Cold Spring Harbor Laboratory Press 1996), which is incorporated herein by reference). This interaction has been measured in vitro using purified 30S ribosomal subunits added to prokaryotic transcripts (Hartz et al., supra, 1991). Bound 30S subunits block extension of a downstream oligonucleotide primer on the mRNA resulting in a ribosomal "toeprint”.
- the psbA-e icoded Dl protein failed to accumulate in the 16S-1470/71 mutant, and accumulated to only 20%) of wild type levels in the 16S- 1467/68 mutant.
- the psbD- encoded D2 protein showed a similar pattern, accumulating to less than 10% of wild type in the 16S-1470/71 mutant and to about 25% in the 16S-1467/68 mutant. Accumulation of the chloroplast ATPase was also impaired in the 16S-1470/71 mutant (50% of wild type levels), although present at near wild type levels in the 16S- 1467/68 mutant. Conversely, accumulation of the soluble chloroplast-encoded large subunit of Rubisco (Lsu) was largely unaffected in either 16S mutant strain.
- the RBS-Alt mutation eliminates the SD base-pairing potential between the psbA mRNA and the 3' terminus of the 16S rRNA, without disrupting the relative location of other elements within the 5'UTR (see Figure 4).
- the DI protein failed to accumulate in RBS-Alt. This result demonstrates that the GGAG sequence is required for psbA expression as expected for an authentic RBS.
- the putative SD sequence in the psbA mRNA which is located 27 nucleotides from the psbA initiation codon, should be unable to direct translation initiation at the proper start codon.
- a series of deletions were introduced into the 5'UTR to position the RBS element closer to the initiation codon ( Figure 4).
- each of the strains that contained an RBS element had significant (>50% wild type levels) psbA mRNA association with ribosomes, even in strains that fail to accumulate the DI protein. Failure to accumulate DI protein would indicate that the ribosome-associated RNA in the RBS-15 and RBS-11 mutants primarily consisted of RNA bound to monoribosomes rather than polyribosomes.
- chimeric genes were constructed that contained the bacterial luciferase coding region placed behind the wild type or mutant psbA 5'UTR. The chimeric genes were transformed into E. coli and translation of the luciferase mRNA was measured by luminescence activity. The luciferase expression pattern in E. coli was inverse to that observed for DI expression in C. reinhardtii. Mutations that position the psbA SD sequence closer to the initiation codon were newly competent for translation in bacteria.
- the coding regions of the bacterial luciferase genes (lux AB)from Vibrio harveyi were fused to either wild type (wt) or mutant psbA 5'UTR's and ligated into plasmids containing the wild type psbA promoter and 3'UTR.
- the plasmids were transformed into E. coli strain BL21 (DE3) and translation of luciferase was monitored by photon counting using a video camera (Welsh and Kay, Curr. Opin. Biotech. 5:617-622, 1997, which is incorporated herein by reference) in the presence of the luciferase substrate n-decyl aldehyde.
- the percentage of optimal expression (RBS-11) was determined for each strain. Luciferase was efficiently translated in bacteria from the constructs containing an RBS positioned 11 to 15 nucleotides upstream of the initiation codon, but was poorly translated when the RBS was positioned greater than 19 nucleotides upstream. This result contrasts with that reported for the 5'UTR of the atpB mRNA from C. reinhardtii, which was reported to drive translation in either bacteria or chloroplast at similar levels (Fargo et al., supra, 1998).
- a nuclear-encoded protein complex specifically recognizes the psbA 5'UTR and dramatically enhances DI protein synthesis by stimulating translation initiation (Danon and Mayfield, supra, 1991; Yohn et al., supra, 1998a; Yohn et al., supra, 1998b).
- RNA binding affinity was measured for each of the mutant RNAs using an in vitro gel shift analysis. Gel shift analysis of binding of the psbA-specific complex to the psbA 5'UTR was performed.
- Radiolabeled RNA fragments conesponding to the wild type psbA 5'-terminus were transcribed in vitro and incubated in the presence of heparin-agarose purified proteins. RNA protein interactions resulted in the retardation of the RNA on nondenaturing PAGE. A 250-fold excess of unlabeled competitor RNA also was added to some reactions. Excess unlabeled RNA corresponding to the psbA 5'UTR from each mutant was used to compete the binding of the protein complex to labeled RNA conesponding to the wild type psbA 5' UTR.
- Chloroplast promoters contain elements similar to those of bacteria, and plastid promoters are capable of driving transcription in E. coli.
- the ribosomes of the chloroplast are clearly related to those of bacteria, and chloroplast ribosomal RNAs and ribosomal proteins show a high degree of conservation with their bacterial counterparts (Harris et al, supra, 1994).
- Chloroplast mRNAs also resemble prokaryotic mRNAs in that they are uncapped, generally not poly-adenylated, and can contain polycistronic messages.
- PRB2 Based on the loss of psbD translation in the 16S mutations and the position of the PRB2 element relative to the SD element of psbA, PRB2 likely is a SD element for the psbD mRNA.
- the location of the RBS element within the psbA mRNA is indicative of a novel mechanism in chloroplasts to promote migration of the early initiation complex from the RBS to the start codon. Secondary structures can shorten the distance between atypically positioned RBS elements in some prokaryotic messages. However, the nucleotides between the psbA RBS and the initiation codon can be substantially altered without loss of psbA translation, and this region is predicted to be relatively unstructured. A scanning mechanism, observed during translation initiation in eukaryotes, also was proposed for chloroplast mRNA, but requires ATP as an energy source for helicase activity, a characteristic not yet ascribed to chloroplast translation.
- chloroplast mRNAs may use protein factors to bring the 30S subunit, bound at the RBS sequence, into register with the initiation codon.
- One specific protein factor that binds to the 5'UTR of the psbA mRNA has homology with a eukaryotic protein known to interact with translation initiation factors (Yohn et al., supra, 1998a).
- Such eukaryotic-like proteins can bring the translation initiation complex to the conect initiation codon, thus functioning as translational regulators in the chloroplast.
- the additional spacing required between the RBS and the initiation codon can accommodate these protein factors, as most of the mutations examined herein did not prevent binding of the these factors in vitro.
- Analogous distal SD sequences also have been identified in the psbA 5'UTR of higher plants, indicating that such SD elements are characteristic for plant chloroplast mRNA.
- Samples of total soluble protein from two transformants (10.6 and 11.3) were collected in the absence or presence of the reducing agent, dithiothreitol (DTT), separated by 10% SDS-PAGE using the Laemmli buffer system, and transferred to nitrocellulose filters (Cohen et al., supra, 1998) for western blot analysis.
- the HSV8 antibody which contains an operatively linked FLAG peptide tag, was visualized using an anti-FLAG peptide tag antibody (M2 monoclonal antibody; Sigma) and an anti-mouse alkaline phosphatase conjugated antibody (Sigma).
- HSV8 single chain antibody expressed in the two different transformants migrated at the expected apparent molecular mass (about 65 kDa).
- HSV8 antibodies isolated in the absence of DTT migrated as a dimer.
- This lsc antibody directed against glycoprotein D of the herpes simplex virus, is produced in a soluble form by the alga and assembles into higher order complexes, in vivo. Aside from dimerization by disulfide bond formation, the antibody undergoes no detectable post-translational modification. Further, the results demonstrate that accumulation of the antibody can be modulated by the specific growth regime used to culture the alga, and by the choice of 5' and 3' elements used to drive expression of the antibody gene. These results demonstrate the utility of alga as an expression platform for recombinant proteins, and describe a new type of single chain antibody containing the entire heavy chain protein, including the Fc domain.
- mAbs Monoclonal antibodies
- the length of time required from the initial transformation event to having usable (mg to gram) quantities of recombinant protein on hand can be as long as three years for species such as corn.
- a second concern sunounding the expression of human therapeutics in food crops, is the potential for gene flow (via pollen) to sunounding crops (6), as occurred between transgenic corn expressing Bacillus thuringiensis insecticidal proteins and native landraces (7). These concerns raise the possibility that regulatory agencies will prohibit the open cultivation of transgenic food plants (like corn, rice and soybean) expressing human therapeutics.
- human monoclonal antibodies and fragments thereof can be expressed in transgenic algae chloroplasts.
- a large single chain antibody gene was engineered in C. reinhardtii chloroplast codon bias, and utilized the C. reinhardtii chloroplast atpA or rbcL promoters and 5' untranslated regions to drive expression.
- This antibody is directed against herpes simplex virus glycoprotein D (15), and contains the entire IgA heavy chain protein fused to the variable region of the light chain by a flexible linker peptide.
- the lsc antibody accumulates as a soluble protein in transgenic chloroplasts, and binds herpes virus proteins, as determined by ELISA assays.
- C. reinhardtii HSV8-lsc were isolated in TRIS buffered saline (TBS; 25 mM TRIS ph 7.4, 150 mM NaCl) containing complete protease inhibitor cocktail tablets (Roche, Inc.) and phenylmethylsuifonyl fluoride MSF) at 1 mM final concentration. Extracts were purified using anti Flag M2 agarose beads (Sigma) according to the manufacturer's protocol. ELISA assays were canied out on volumes of lOO ⁇ l volumes in 96 well microtiter plates (Costar) coated with 100 ⁇ l of HSV proteins.
- Samples for use in ELISA were diluted in blocking buffer comprised of phosphate buffered saline (PBS; 137 mM NaCl, 2.7 mM KCl, 1.8 mM K 2 HPO 4 , 10 mM Na 2 HPO , pH 7.4) and 5% nonfat dry milk. Incubations were carried out for 8 hr at 4°C with rocking. Plates were then rinsed with PBS plus 0.5% Tween 20 three times, then incubated with anti-Flag antibody for 8 hr at 4°C.
- PBS phosphate buffered saline
- Protein concentrations were determined using a BioRad Protein assay reagent. Western blots were canied out as described (23) using a murine anti-Flag primary antibody (Sigma) and an alkaline phosphatase conjugated goat anti-mouse secondary antibody (Santa Cruz Biotechnology). RESULTS
- a single chain antibody gene was synthesized using codons optimized to reflect abundantly translated C. reinhardtii chloroplast mRNAs.
- the engineered antibody was derived from a human antibody library displayed on phage, and identified by panning with herpes simplex virus proteins (15). This antibody, termed HSV8, was previously shown to bind the viral surface antigen glycoprotein D (16), and both Fab or IgGl versions of this antibody act as efficient neutralizing antibodies, in vivo and in vitro (15, 16).
- the chimeric antibody genes were ligated into the Bam HI site of p322 to create plasmid p322/atpA- HSV8 and plasmid p322/rbcL-HSV8.
- the p322/HSV8 constructs were co-transformed into C. reinhardtii chloroplasts via particle bombardment (17), along with plasmid p228, containing a 16S ribosomal gene confe ing spectinomycin resistance.
- HSV8 positive transformants were taken through additional rounds of selection to isolate homoplasmic lines in which all copies of the chloroplast genome contained the introduced HSV8-lsc gene.
- Two homoplasmic transformants were selected, one 10-6-3, containing the atpA promoter driving HSV8-lsc and the other, 20-4-4, containing the rbcL promoter driving HSV8-lsc.
- Genomic DNA from wt and the two HSV8-lsc transformants was digested with Eco Rl and Xho I, separated on agarose gels, and subjected to Southern blot analysis.
- C. reinhardtii DNA was prepared as described in Example 3, digested with Eco Rl and Xho I, and filters were hybridized with the radioactive probes indicated by the double anowheads in Figure 5C.
- Hybridization with a 32 P labeled Nde I/Xba I fragment of the HSV8 coding region identified a 6.0 kb band in both the atpA-HSV8 and rbcL-HSV8 transgenic strains, while no detectable band was observed in the wt lane, as expected.
- a 32 P labeled 1.5 kb Eco Rl to Pst I fragment from the 5' end of p322 a 5.7 kb fragment was visualized in the wt sample, while a slightly larger 6.0 kb fragment was identified in the two transgenic strains.
- RNA from wt and the two transgenic lines was separated on denaturing agarose gels and blotted to nylon membrane.
- Duplicate filters were stained with methylene blue and hybridized with either a 32 P labeled psbA cDNA probe, or an HSV8 specific probe.
- Ribosomal RNA andpsbA mRNA accumulate to similar levels in wt and each of the transgenic strains, demonstrating that equal amounts of RNA were loaded, and that introduction of the transgene does not alter endogenous mRNA accumulation.
- Hybridization with an HSV8 specific probe showed that strains 10-6-3 and 20-4-4 accumulate HSV8-lsc mRNA of the conect size, while no HSV8 signal was detected in the wt lane, as expected.
- HSV8-lsc antibody levels were measured by Western blot analysis using an anti-flag antibody to determine if HSV8-lsc protein accumulated in the transgenic lines. Twenty ⁇ g of total protein from an E. coli strain expressing HSV8-lsc from a pET vector, and 20 ⁇ g of total protein from C. reinhardtii wt and the two transgenic lines, was separated by SDS-PAGE and either stained with Coomassie blue, or subjected to western blot analysis with anti-Flag antisera. For bacterial expression, the Nde I/Bam HI fragment of codon optimized HSV8-lsc gene was ligated into a pET vector and expression was induced by addition of IPTG.
- the Coomassie stained gel indicated that equal amounts of protein were loaded in each lane, and that overall protein accumulation was normal in the transgenic lines.
- Western blot analysis of the same samples using an anti-Flag antibody showed a robust signal of the conect molecular weight in both of the HSV8-lsc transgenic strains and E. coli, but no signal in the C. reinhardtii wt lane, as expected.
- HSV8-lsc that accumulated in C. reinhardtii chloroplast was functional
- the chloroplast expressed protein was characterized along with that of the bacterial expressed HSV8-lsc.
- HSV8-lsc transgenic bacteria and algae were resuspended in TBS, and the cells lysed by sonication. Soluble proteins were separated from insoluble proteins by centrifugation. Equal amounts of protein from the soluble fractions and from the insoluble pellets were separated by SDS-PAGE, and HSV8-lsc proteins visualized by western blot analysis.
- any disulfide bonds formed between the two heavy chain moieties of the antibody should remain intact allowing the antibody to migrate as a larger species.
- chloroplast expressed HSV8-lsc runs as a much larger protein of approximately 140 kDa, the size expected of a dimmer. Treatment with Bme, to reduce disulfide bonds, results in the migration of the chloroplast HSV8-lsc proteins at the predicted molecular weight of the monomer at 68 kDa.
- HSV8-lsc The ability of chloroplast expressed HSV8-lsc to bind HS V8 proteins was examined to confirm that the HSV8-lsc accumulating in the transgenic chloroplast was functional.
- HSV8-lsc was purified from transgenic chloroplast using an anti-flag affinity resin. As shown in Figure 6, the chloroplast produced antibody recognized HSV8 proteins in ELISA assays in a robust manner.
- a human monoclonal antibody was expressed in the chloroplast of green algae. High levels of recombinant protein expression were achieved by optimizing the codon usage within the antibody coding sequence to reflect the codon usage of abundant chloroplast proteins, and by driving expression of the chimeric gene using the chloroplast atpA or rbcL promoters and 5' UTRs.
- This large single chain (lsc) antibody contains the entire IgA heavy chain fused to the variable region of the light chain by a flexible linker, and accumulated as a fully soluble protein in chloroplast. The antibody was directed against glycoprotein D of herpes simplex virus, and the alga expressed antibody bound to herpes proteins as determined through ELISA.
- This lsc antibody contains the Fc portion of the heavy chain, which is the site normally involved in intermolecular disulfide bond formation leading to dimerization of the antibody.
- the chloroplast expressed antibody assembled into higher order complexes that are susceptible to reduction by Bme, indicating that the chloroplast expressed antibody forms dimers in vivo. Formation of disulfide bonds in recombinant proteins expressed in chloroplast has been shown for human somatotropin expressed in tobacco chloroplast (8), and was somewhat expected due to the presence of protein disulfide isomerase in algal chloroplasts (18).
- This lsc antibody also contains putative sites for N-linked glycosylation.
- Chloroplast encoded proteins are not known to be glycosylated and, indeed, there was no evidence of glycosylation of the chloroplast expressed antibody based upon mass spectral analysis.
- the transgenic strains generated showed differential accumulation of antibody depending upon the promoter used to drive expression, as well as the cell density and light conditions under which they are cultured. The reasons for these large fluctuations in antibody accumulation likely arise from a variety of factors including stability and translational competence of the chimeric mRNAs, and turnover of the antibody protein.
- Recombinant proteins can be produced in a variety of protein expression systems.
- Complex therapeutic proteins like monoclonal antibodies (mAbs) are primarily produced by culture of transgenic mammalian cells. Costs for mAb production in cultured mammalian cells averages approximately $150/gram for raw materials, while in plant systems mAb production has been estimated to cost $0.05/ gram (1). Costs for production of mAbs in algal systems are expected to rival those in terrestrial plants, given that media costs for algae are quite reasonable ($0.002/liter). In addition, algae can be grown in continuous culture and their growth medium recycled.
- transgenic algae can be generated quickly, requiring only a few weeks between the generation of initial transformants and their scale up to production volumes.
- both the chloroplast and nuclear genome of algae can be genetically transformed, opening the possibility of producing a variety of transgenic proteins in a single organism, a requirement if multimeric protein complexes such as secretory antibodies are to be produced.
- algae have the ability to be grown on scales ranging from a few milliliters to 500,000 liters, in a cost effective manner.
- This Example confirms the robust expression in chloroplasts of a luciferase fusion protein encoded by a chloroplast codon biased synthetic polynucleotide.
- Luciferase reporter genes have been successfully used in a variety of organisms to examine gene expression in living cells, but have yet to be successfully developed for use in chloroplast. As disclosed in Example 1, a green fluorescent protein (gfp) has been expressed from a chloroplast codon biased polynucleotide and was useful as a reporter of chloroplast gene expression.
- gfp green fluorescent protein
- a luciferase reporter protein encoded by a chloroplast codon biased polynucleotide was developed a luciferase reporter by synthesizing the two subunit bacterial luciferase, luxAB, as a single fusion protein in C. reinhardtii chloroplast codon bias.
- the chloroplast luciferase gene, luxCt was expressed in C. reinhardtii chloroplasts under the control of the atpA promoter and 5' UTR and rbcL 3'UTR.
- the luxCt is a sensitive reporter of chloroplast gene expression, allowing luciferase activity to be measured in vivo using a CCD camera or in vitro using a luminometer. Furthermore, luxCt protein accumulation, as measured by western blot analysis, is proportional to luminescence as determined both in vivo and in vitro.
- Reporter genes have greatly enhanced the ability to monitor gene expression in a number of biological organisms.
- ⁇ -glucuronidase (uidA, Staub and Maliga, 1993)
- neomycin phosphotransferase nptll, Carrer et al., 1993
- aadA adenosyl-3-adenyltransferase
- gfp of Aequorea aequorea Sidorov et al., 1999; Reed et al, 2001 have been used as reporter genes (Heifetz, 2000).
- reporter genes have also been expressed in the chloroplast of the eukaryotic green alga, C. reinhardtii, including aadA (Goldschmidt-Clennont, 1991; Zerges and Rochaix, 1994), uidA (Sakamoto et al, 1993, Ishikura et al., 1999), aphA6 (Bateman and Purton, 2000) and Renilla luciferase (Minico et al., 1999). Unfortunately, these initial reporter gene cassettes produced very low levels of protein accumulation, making them poor reporters for quantitative analysis of gene expression.
- a bacterial luciferase gene was synthesized having C. reinhardtii chloroplast codon bias.
- the de novo synthesized lux gene was based on the bacterial luxAB gene of Vibrio harveyi (Baldwin et al., 1984, Johnson et al., 1986).
- the luciferase coding sequence was synthesized such that the luciferase A and B subunits were expressed as a single coding region by linking the A and B subunits with a flexible peptide linker (Kirchener et al, 1989, Olsson et al., 1989, Almashanu et al, 1990).
- the chloroplast optimized luciferase (luxCt) gene was placed in a cassette containing the atpA promoter and 5'UTR and the rbcL 3' UTR.
- Transgenic lines containing the luxCt gene accumulated luxCt mRNA and LUXCt protein, as judged by northern and western blot analysis, respectively (see below).
- Luminescence from transgenic lines expressing luxCt was easily visualized with a CCD camera, when cells were treated with decanal, the bacterial luciferase substrate, while wt cells showed no luminescence in the same assays.
- Expression of luxCt as judged by western blot analysis, was proportional to expression of luxCt, as judge by luminescence assays using a CCD camera, and by in vitro luminometer assays. Luciferase activity in transgenic lines could be measured over several orders of magnitude, demonstrating the sensitivity and utility of luxCt as a reporter of chloroplast gene expression in living cells.
- Transformations were canied out on C. reinhardtii strain 137c (mt+), or in the psbA deficient strain cc744 (REF).
- Cells were grown to late log phase (approximately 7 days) in the presence of 40 M 5-fluorodeoxyuridine in TAP medium (Gorman and Levine, 1965) at 23° C under constant illumination of 4,000 lux (high light) on a rotary shaker set at 100 rpm. Fifty ml of cells were harvested by centrifugation at 4,000 x g at 4°C for 5 min.
- the atpA promoter and 5' UTR and the rbcL 3' UTR were generated as described (Mayfield et al., 2002).
- Chloroplast transformation plasmid p322 was constructed as described (Franklin et al, 2002).
- Plasmids pluxAB and pluxCt were transformed into E. coli strain BL21 and cells grown overnight in liquid media.
- proteins were isolated from E. coli or from C. reinhardtii utilizing a buffer containing 750 mM Tris-Cl, pH 8.0, 15% sucrose (wt/vol), 100 mM Bme, 1 mM PMSF. Samples were centrifuged for 30 min at 13,000 x g at 4°C with the resulting supernatant used in western blot analysis.
- reinhardtii proteins for use in in vitro luminescence assays were prepared in 50 mM Na 2 HPO , pH 7.0, 50 mM Bme, 400 mM sucrose buffer, and the crude lysate was centrifuged for 30 min at 13,000 x g at 4°C with the resulting supernatant used in luciferase assays.
- 96 well microtiter assays were adapted from the bacterial luciferase method (Langridge and Szalay, 1994).
- reinhardtii soluble proteins were diluted in luciferase extraction buffer to 100 ⁇ l per sample, to which 125 ⁇ l of 500 ⁇ M NADH in 50 mM Tris-Cl, pH 8.0, and 0.025 U of diaphorase in 50 mM Na 2 HPO 4 , 50 mM Bme, 1%) bovine serum albumin buffer were added.
- 125 ⁇ l of 500 ⁇ M NADH in 50 mM Tris-Cl, pH 8.0, and 0.025 U of diaphorase in 50 mM Na 2 HPO 4 , 50 mM Bme, 1%) bovine serum albumin buffer were added.
- 130 ⁇ l of a solution containing 125 ⁇ l 100 ⁇ M FMN " in 200 mM Tricine, pH 7.0 and 5 ⁇ l 0.1% decanal sonicated for 10s in 50 mM Na HPO 4 , pH 7.4 was added.
- a luciferase gene was synthesized using codons optimized to reflect abundantly expressed genes of the C. reinhardtii chloroplast (Example 1; Franklin et al., 2002).
- the luciferase gene, luxCt ( Figure 7, was designed based on the bacterial luciferase AB gene of Vibrio harveyi (luxAB; Baldwin et al., 1984).
- the two subunits of luxAB were linlced into a single coding sequence by eliminating the stop codon of the A subunit and linking the B subunit, in the conect reading frame, with a flexible peptide sequence to create a single fusion protein (Figure 7).
- the V. harveyi luxAB sequence was obtained from the GenBank database and a series of oligonucleotides were designed based on the amino acid sequence, but changing codon usage to reflect those of highly expressed C. reinhardtii chloroplast genes. The gene was assembled by the method of Stemmer et al. (1995). PCR products were cloned into E.
- coli plasmids the synthetic gene sequenced, and errors corrected by site directed mutagenesis.
- An Nde I site was placed at the initiation codon and an Xba I site placed immediately downstream of the stop codon, for ease in subsequent cloning.
- the resulting gene, luxCt was cloned into an E. coli expression cassette and luciferase expression was assayed by luminescence imaging with a CCD camera. Surprisingly, no luminescence was detected in bacteria containing the luxCt gene, although high luminescence could be detected in bacteria transformed with the bacterial luxAB gene (Kondo et al., 1993).
- both the luxCt and the bacterial luxAB genes contained in the E. coli expression plasmids were sequenced. No errors were detected in the luxCt gene compared to the desired sequence, but a number of differences were identified in the luxAB sequence from the plasmid used to express luxAB in bacteria (Kondo et al, 1993), and the luxAB sequence reported in the GenBank database (Ace. No. E12410). Alignment of luxAB proteins from several different bacterial species (Johnson et al.
- the luxCt fusion protein gene produces a functional luciferase in bacteria
- the alpha (A) and beta (B) subunits of luxAB were identified, as was the single fusion protein (FP) of luxCt.
- FP single fusion protein
- luciferase expression was determined in E. coli was grown overnight on agar media and treated with decanol vapor. Untransformed E. coli cells or cells expressing either the luxAB or luxCt genes were photographed with reflect light (photograph), or visualized by luminescent imaging with a CCD camera (luminescence). When E. coli cells were treated with decanal and imaged with a CCD • camera, both luciferase strains luminesced, while untransformed E. coli showed no light signal, as expected.
- Wild type C. reinhardtii cells were transfonned with the p322-atpA /r ⁇ :Ct plasmid and the selectable marker plasmid p228, confening resistance to spectinomycin.
- Primary transformants were screened for the presence of the luxCt gene by luminescent assays on the CCD camera, and positive transformants were confirmed by Southern blot analysis. Transformants were taken through additional rounds of selection to isolate homoplasmic lines in which all copies of the chloroplast genome contained the introduced luxCt gene.
- FIG. 8 shows the luxCt constructs with relevant restriction sites indicated.
- Conect integration of the 8.7 kb Eco/Xho region of plasmid p322-atpA luxCt into the chloroplast genome was ascertained using either the Nde I - Xba I fragment of luxCt, or the Bam HI - Xho I fragment of plasmid p322, as indicated in Figure 8.
- Southern blot analysis of luxCt C. reinhardtii chloroplast transformants was performed.
- reinhardtii DNA was prepared as described in Example 4, digested simultaneously with Eco Rl and Xho I and subjected to Southern blot analysis. Filters were hybridized with the radioactive probe indicated in Figure 8B. The two transformants contained luxCt hybridizing bands, while the wild type strain showed no signal with this luxCt coding region probe. Two bands were identified in the transgenic lines because the luxCt gene contains a single Eco Rl site in the middle of the gene. Hybridization with the Bam HI - Xho I fragment from the p322 plasmid identified a single band in wt and a different sized band in the two transformants, as expected. Each of these bands was of the correct predicted size for both the wt and transformant lines. These results demonstrate that the two transgenic lines are homoplasmic.
- luxCt as a reporter of chloroplast gene expression in living cells
- luxCt gene As a reporter of chloroplast gene expression in living C. reinhardtii cells, luminescence was measured for wt and transgenic cells grown on agar plates. Cells were plated on solid media and maintained for seven days under continuous light (1000 lux). Decanal, the substrate for luxAB, was swabbed onto the Petri plate lids, and the plates were placed under a CCD camera. The transgenic lines appeared similar to wt cells when visualized under ambient light. Imaging with the CCD camera, after 5 min of dark adaptation to eliminate chlorophyll fluorescence, showed a bright luminescent signal for the two transgenic lines, and no signal for the wt strain. The signal from the luxCt transgenic lines was sufficient to visualize even small individual colonies in vivo.
- the cassette was transformed into a psbA deficient strain of C. reinhardtii (cc744, Chlamydomonas Genetics Center, http://www.botany.duke.edu/chlamy/). Again, primary transformants were screened by luminescent assays with the CCD camera, and positive transgenic lines were taken through several rounds of selection to obtain homoplasmic lines. Luminescence from the cc744/luxCt strain was much higher than from the 137c/luxCt strain.
- luxCt protein accumulation and luciferase activity were measured in the 137c and cc744 transgenic lines. Wild type and luxCt transgenic lines luxCtl37c and luxCtcc744 were grown on agar plates and treated with decanal. Cells were either photographed under reflective light (photograph), or visualized on a CCD camera (luminescence). Proteins were extracted from the cells and subjected to western blot analysis (western anti-luxAB) or quantitated by luminometer assays (luminometer).
- luxCt gene is a robust reporter of chloroplast gene expression, and that measurement of lux activity in vivo conesponded to luciferase accumulation as measured by both western blot analysis and in vitro luminescence assays.
- Luciferases have been used in a number of organisms as reporter genes (Greer and Szalay, 2002; Langeridge et al, 1994; Kondo et al., 1993; Kay, 1993) due to their high level of sensitivity and because luciferase can be readily visualized in living cells with little perturbation of the organism.
- This Example demonstrates the construction of a luciferase reporter gene for chloroplast expression by synthesizing the two subunit bacterial luciferase, luxAB, as a single fusion protein, and by optimizing the codon usage of this synthetic luciferase gene to reflect highly expressed genes from the C. reinhardtii chloroplast.
- Example 2 extends the results of Example 1 , which demonstrated that codon usage dramatically effected the expression of heterologous proteins in C. reinhardtii chloroplast by synthesizing a gfp in chloroplast-optimized codons (see, also, Franklin et al, 2002).
- the cgfp accumulated to 0.5% of total soluble protein within transgenic chloroplast, and could be visualized by fluorescent analysis of chloroplast extracts. However, even that relatively high level of GFP accumulation was insufficient to visualize the reporter in vivo.
- Komine et al reported visualization of gfp in transgenic C.
- the transgenic strains expressing luxCt accumulated sufficient levels of luciferase to be easily visualized by luminescence assays in vivo using a CCD camera.
- LuxCt protein accumulation as measured by western blot analysis, was proportional to luciferase activity as measured by CCD camera luminescence assays or in vitro luminometer assays.
- C. reinhardtii has been referred to as "green yeast", a well deserved term given the excellent genetic characteristic of this organism that have allowed its use to dissect a number of cellular processes, most notably in the biogenesis of flagella and the photosynthetic apparatus. What has clearly been lacking, however, is a facile means to assay gene expression, especially in the chloroplast.
- the present results demonstrate the utility of the optimized luxCt gene as a reporter of chloroplast gene expression in vivo.
- the present results also demonstrate that luxCt is a sensitive reporter capable of monitoring gene expression even in small colonies of cells, making luxCt the reporter of choice for any high throughput analysis of chloroplast gene expression.
Landscapes
- Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Plant Pathology (AREA)
- Biochemistry (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Immunology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Peptides Or Proteins (AREA)
- Breeding Of Plants And Reproduction By Means Of Culturing (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
Abstract
Description
Claims
Priority Applications (8)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2003239182A AU2003239182B2 (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same |
JP2003587949A JP2005537784A (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts and compositions and methods for expressing the polypeptides |
CA2483337A CA2483337C (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same |
EP03733900A EP1576101A4 (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same |
KR1020047017113A KR101107380B1 (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same |
MXPA04010432A MXPA04010432A (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same. |
CN03812392.4A CN1886512B (en) | 2002-04-23 | 2003-04-23 | The expression of polypeptide in chloroplast(id) and for the composition of express polypeptide and method |
IL164549A IL164549A (en) | 2002-04-23 | 2004-10-13 | Method of producing a functional polypeptide in a microalgal plastid |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US37512902P | 2002-04-23 | 2002-04-23 | |
US60/375,129 | 2002-04-23 | ||
US43495702P | 2002-12-19 | 2002-12-19 | |
US60/434,957 | 2002-12-19 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2003091413A2 true WO2003091413A2 (en) | 2003-11-06 |
WO2003091413A3 WO2003091413A3 (en) | 2006-06-29 |
Family
ID=29273018
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2003/012997 WO2003091413A2 (en) | 2002-04-23 | 2003-04-23 | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same |
Country Status (10)
Country | Link |
---|---|
US (1) | US20040014174A1 (en) |
EP (1) | EP1576101A4 (en) |
JP (1) | JP2005537784A (en) |
KR (1) | KR101107380B1 (en) |
CN (1) | CN1886512B (en) |
AU (1) | AU2003239182B2 (en) |
CA (1) | CA2483337C (en) |
IL (1) | IL164549A (en) |
MX (1) | MXPA04010432A (en) |
WO (1) | WO2003091413A2 (en) |
Cited By (23)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2006211977A (en) * | 2005-02-04 | 2006-08-17 | Univ Nagoya | Firefly luciferase gene functioning in chloroplast and real-time monitoring of chloroplast gene expression using the same |
US20090280545A1 (en) * | 2007-09-11 | 2009-11-12 | Sapphire Energy | Molecule production by photosynthetic organisms |
WO2010068821A1 (en) | 2008-12-10 | 2010-06-17 | Synthetic Genomics, Inc. | Production of branched-chain alcohols by photosynthetic microoraganisms |
EP2215247A1 (en) * | 2007-11-13 | 2010-08-11 | The Scripps Research Institute | Production of cytotoxic antibody-toxin fusion in eukaryotic algae |
EP2248825A1 (en) * | 2009-05-04 | 2010-11-10 | Ribovax Biotechnologies SA | Antigen binding fragments of an antibody for use in treating and diagnosing ocular diseases |
WO2011008565A1 (en) | 2009-06-29 | 2011-01-20 | Synthetic Genomics, Inc. | Acyl-acp thioesterase genes and uses therefor |
WO2011019858A1 (en) | 2009-08-11 | 2011-02-17 | Synthetic Genomics, Inc. | Microbial production of fatty alcohols |
US8143039B2 (en) | 2007-06-01 | 2012-03-27 | Sapphire Energy, Inc. | Use of genetically modified organisms to generate biomass degrading enzymes |
WO2012087673A1 (en) | 2010-12-23 | 2012-06-28 | Exxonmobil Research And Engineering Company | Prokaryotic acyl-acp thioesterases for producing fatty acids in genetically engineered microorganisms |
WO2013162648A1 (en) | 2012-04-23 | 2013-10-31 | Exxonmobil Research And Engineering Company | Cell systems and methods for improving fatty acid synthesis by expression of dehydrogenases |
EP2660323A1 (en) | 2012-05-02 | 2013-11-06 | Algenics | Production of secreted therapeutic antibodies in phaeodactylum tricornutum microalgae |
EP2660324A1 (en) | 2012-05-02 | 2013-11-06 | Algenics | Production of secreted therapeutic antibodies in microalgae |
WO2013164095A1 (en) | 2012-05-02 | 2013-11-07 | Algenics | Production of secreted therapeutic antibodies in microalgae |
WO2014062163A1 (en) | 2012-10-16 | 2014-04-24 | Exxonmobil Research And Engineering Company | Dgat genes and methods of use for triglyceride production in recombinant microorganisms |
WO2014089533A2 (en) | 2012-12-06 | 2014-06-12 | Synthetic Genomics, Inc. | Algal mutants having a locked-in high light acclimated phenotype |
US8835149B2 (en) | 2012-12-06 | 2014-09-16 | Exxonmobil Research And Engineering Company | DGAT genes comprising pleckstrin homology domains and methods of use for triglyceride production in recombinant microorganisms |
US8846370B2 (en) | 2010-12-23 | 2014-09-30 | Exxonmobil Research And Engineering Company | Genetically engineered microorganisms comprising 4-hydroxybenzoyl-coa thioesterases and methods of using the same for producing free fatty acids and fatty acid derivatives |
US8940508B2 (en) | 2010-12-31 | 2015-01-27 | Exxonmobil Research And Engineering Company | Enhancement of biomass production by disruption of light energy dissipation pathways |
US9096834B2 (en) | 2012-02-24 | 2015-08-04 | Exxonmobil Research And Engineering Company | Recombinant microorganisms comprising thioesterase and lysophosphatidic acid acyltransferase genes for fatty acid production |
US9175256B2 (en) | 2010-12-23 | 2015-11-03 | Exxonmobil Research And Engineering Company | Production of fatty acids and fatty acid derivatives by recombinant microorganisms expressing polypeptides having lipolytic activity |
US9718883B2 (en) | 2003-09-10 | 2017-08-01 | Amgen Fremont Inc. | Antibodies to M-CSF |
WO2017160573A1 (en) | 2016-03-18 | 2017-09-21 | Exxonmobil Research And Engineering Company | Chlorophyllase overproduction to enhance photosynthetic efficiency |
EP3579845A4 (en) * | 2017-02-09 | 2020-12-30 | Dow Agrosciences LLC | Novel eukaryotic cell-free expression system that does not require an artificial energy regeneration system |
Families Citing this family (18)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
AR042145A1 (en) * | 2002-11-27 | 2005-06-08 | Dow Agrociences Llc | IMMUNOGLOBULIN PRODUCTION IN PLANTS WITH A REDUCED FUCOCILATION |
WO2007133558A2 (en) * | 2006-05-09 | 2007-11-22 | The Scripps Research Institute | Robust expression of a bioactive mammalian protein in chlamydomonas chloroplast |
JP2010517558A (en) * | 2007-02-09 | 2010-05-27 | サントル ナショナル ドゥ ラ ルシェルシュ スィヤンティフィック(セーエヌエルエス) | Expression of polypeptides from the nuclear genome of the species Ostreococcus |
EP2203542A1 (en) * | 2007-09-11 | 2010-07-07 | Sapphire Energy, Inc. | Methods of producing organic products with photosynthetic organisms and products and compositions thereof |
US20090176272A1 (en) * | 2007-09-12 | 2009-07-09 | Kuehnle Agrosystems, Inc. | Expression of nucleic acid sequences for production of biofuels and other products in algae and cyanobacteria |
US8314222B2 (en) * | 2007-10-05 | 2012-11-20 | Sapphire Energy, Inc. | System for capturing and modifying large pieces of genomic DNA and constructing organisms with chloroplasts |
US20090269816A1 (en) * | 2007-10-05 | 2009-10-29 | Sapphire Energy, Inc. | System for capturing and modifying large pieces of genomic DNA and constructing organisms with synthetic chloroplasts |
US20100050301A1 (en) * | 2007-10-05 | 2010-02-25 | Sapphire Energy, Inc. | System for capturing and modifying large pieces of genomic dna and constructing vascular plants with synthetic chloroplast genomes |
EP2220125A4 (en) * | 2007-11-13 | 2010-12-29 | Sapphire Energy Inc | Production of fc-fusion polypeptides in eukaryotic algae |
MX2010013917A (en) | 2008-06-27 | 2011-03-03 | Sapphire Energy Inc | Induction of flocculation in photosynthetic organisms. |
AU2009333021B2 (en) * | 2008-12-31 | 2015-09-10 | Sapphire Energy, Inc. | Genetically engineered herbicide resistant algae |
BRPI1008958A2 (en) * | 2009-03-11 | 2015-09-01 | Sapphire Energy Inc | Biofuel production in prokaryotes and eukaryotes |
JP2013529058A (en) * | 2010-02-25 | 2013-07-18 | バイオグロウ インク. | Self-luminous plant containing bacterial LUX operon and method for producing the same |
JP2011217662A (en) * | 2010-04-08 | 2011-11-04 | Senshukai Co Ltd | Chloroplast transformed plant mass-expressing modified luciferase |
KR101633159B1 (en) * | 2013-11-28 | 2016-06-23 | 부경대학교 산학협력단 | Composition for treating or preventing gastrointestinal disorders comprsing peptide derived from chlamidomonas sp. |
CN109071609A (en) * | 2016-03-24 | 2018-12-21 | 马凯特大学 | Quantitative flagellum fluorescent marker and standard items |
EP3544994B1 (en) | 2016-11-27 | 2024-09-25 | Triton Algae Innovations, Inc. | Method of purification of recombinant osteopontin from microalgae |
CN115948450B (en) * | 2022-08-01 | 2024-02-13 | 深圳大学 | Chlamydomonas reinhardtii chloroplast-saccharomyces cerevisiae-escherichia coli shuttle vector and construction method and application thereof |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6156517A (en) * | 1997-01-17 | 2000-12-05 | The Scripps Research Institute | RNA binding protein and binding site useful for expression of recombinant molecules |
US6271444B1 (en) * | 1998-07-10 | 2001-08-07 | Calgene Llc | Enhancer elements for increased translation in plant plastids |
WO2001064929A1 (en) * | 2000-02-29 | 2001-09-07 | Auburn University | Production of antibodies in transgenic plastids |
Family Cites Families (12)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5567600A (en) * | 1983-09-26 | 1996-10-22 | Mycogen Plant Sciences, Inc. | Synthetic insecticidal crystal protein gene |
GB2200118A (en) * | 1987-01-23 | 1988-07-27 | Allelix Inc | Synthetic chymosin-and prochymosin-encoding DNA segments |
US5082767A (en) * | 1989-02-27 | 1992-01-21 | Hatfield G Wesley | Codon pair utilization |
US5661017A (en) * | 1993-09-14 | 1997-08-26 | Dunahay; Terri Goodman | Method to transform algae, materials therefor, and products produced thereby |
US5545818A (en) * | 1994-03-11 | 1996-08-13 | Calgene Inc. | Expression of Bacillus thuringiensis cry proteins in plant plastids |
US5545817A (en) * | 1994-03-11 | 1996-08-13 | Calgene, Inc. | Enhanced expression in a plant plastid |
US6110668A (en) * | 1996-10-07 | 2000-08-29 | Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. | Gene synthesis method |
PL343460A1 (en) * | 1998-03-11 | 2001-08-13 | Novartis Ag | Expression of trehalose biosynthetic genes in plants |
ATE360072T1 (en) * | 1998-07-10 | 2007-05-15 | Calgene Llc | EXPRESSION OF EUKAROTIC PEPTIDES IN PLANT PLASTIDS |
MXPA01001365A (en) * | 1998-08-03 | 2002-11-29 | Univ Rutgers | Translation control elements for high-level protein expression in the plastids of higher plants and methods of use thereof. |
CN1330718A (en) * | 1998-10-07 | 2002-01-09 | 辛根塔参与股份公司 | Therapeutically active proteins in plants |
EP2080803B1 (en) * | 2000-03-01 | 2011-05-25 | Auburn University | Plastid transformation vectors for expressing proinsulin fused to the cholera toxin B-subunit in plants |
-
2003
- 2003-04-23 CN CN03812392.4A patent/CN1886512B/en not_active Expired - Fee Related
- 2003-04-23 WO PCT/US2003/012997 patent/WO2003091413A2/en active Application Filing
- 2003-04-23 AU AU2003239182A patent/AU2003239182B2/en not_active Ceased
- 2003-04-23 US US10/422,628 patent/US20040014174A1/en not_active Abandoned
- 2003-04-23 MX MXPA04010432A patent/MXPA04010432A/en active IP Right Grant
- 2003-04-23 EP EP03733900A patent/EP1576101A4/en not_active Withdrawn
- 2003-04-23 CA CA2483337A patent/CA2483337C/en not_active Expired - Fee Related
- 2003-04-23 KR KR1020047017113A patent/KR101107380B1/en not_active IP Right Cessation
- 2003-04-23 JP JP2003587949A patent/JP2005537784A/en active Pending
-
2004
- 2004-10-13 IL IL164549A patent/IL164549A/en not_active IP Right Cessation
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6156517A (en) * | 1997-01-17 | 2000-12-05 | The Scripps Research Institute | RNA binding protein and binding site useful for expression of recombinant molecules |
US6271444B1 (en) * | 1998-07-10 | 2001-08-07 | Calgene Llc | Enhancer elements for increased translation in plant plastids |
WO2001064929A1 (en) * | 2000-02-29 | 2001-09-07 | Auburn University | Production of antibodies in transgenic plastids |
Non-Patent Citations (2)
Title |
---|
See also references of EP1576101A2 * |
SIDOROV ET AL: 'Stable chloroplast transformation in potato: use of green fluorescent protein as a plasmid marker.' PLANT J. vol. 19, no. 2, 1999, pages 209 - 216, XP000886694 * |
Cited By (34)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9718883B2 (en) | 2003-09-10 | 2017-08-01 | Amgen Fremont Inc. | Antibodies to M-CSF |
US10280219B2 (en) | 2003-09-10 | 2019-05-07 | Amgen Fremont Inc. | Antibodies to M-CSF |
JP2006211977A (en) * | 2005-02-04 | 2006-08-17 | Univ Nagoya | Firefly luciferase gene functioning in chloroplast and real-time monitoring of chloroplast gene expression using the same |
US8143039B2 (en) | 2007-06-01 | 2012-03-27 | Sapphire Energy, Inc. | Use of genetically modified organisms to generate biomass degrading enzymes |
US8669059B2 (en) | 2007-06-01 | 2014-03-11 | The Scripps Research Institute | High throughput screening of genetically modified photosynthetic organisms |
US8318436B2 (en) | 2007-06-01 | 2012-11-27 | Sapphire Energy, Inc. | Use of genetically modified organisms to generate biomass degrading enzymes |
US8268553B2 (en) | 2007-06-01 | 2012-09-18 | Sapphire Energy, Inc. | High throughput screening of genetically modified photosynthetic organisms |
US20090280545A1 (en) * | 2007-09-11 | 2009-11-12 | Sapphire Energy | Molecule production by photosynthetic organisms |
EP2215247A4 (en) * | 2007-11-13 | 2010-11-10 | Scripps Research Inst | Production of cytotoxic antibody-toxin fusion in eukaryotic algae |
EP2215247A1 (en) * | 2007-11-13 | 2010-08-11 | The Scripps Research Institute | Production of cytotoxic antibody-toxin fusion in eukaryotic algae |
WO2010068821A1 (en) | 2008-12-10 | 2010-06-17 | Synthetic Genomics, Inc. | Production of branched-chain alcohols by photosynthetic microoraganisms |
WO2010128053A1 (en) * | 2009-05-04 | 2010-11-11 | Ribovax Biotechnologies Sa | Antigen binding fragments of an antibody for use in treating or diagnosing ocular diseases |
US8747856B2 (en) | 2009-05-04 | 2014-06-10 | Ribovax Biotechnologies S.A. | Antigen binding fragments of an antibody for use in treating or diagnosing ocular diseases |
EP2248825A1 (en) * | 2009-05-04 | 2010-11-10 | Ribovax Biotechnologies SA | Antigen binding fragments of an antibody for use in treating and diagnosing ocular diseases |
WO2011008565A1 (en) | 2009-06-29 | 2011-01-20 | Synthetic Genomics, Inc. | Acyl-acp thioesterase genes and uses therefor |
WO2011019858A1 (en) | 2009-08-11 | 2011-02-17 | Synthetic Genomics, Inc. | Microbial production of fatty alcohols |
WO2012087673A1 (en) | 2010-12-23 | 2012-06-28 | Exxonmobil Research And Engineering Company | Prokaryotic acyl-acp thioesterases for producing fatty acids in genetically engineered microorganisms |
US8530207B2 (en) | 2010-12-23 | 2013-09-10 | Exxonmobil Research And Engineering Company | Photosynthetic microorganisms comprising exogenous prokaryotic acyl-ACP thioesterases and methods for producing fatty acids |
US9175256B2 (en) | 2010-12-23 | 2015-11-03 | Exxonmobil Research And Engineering Company | Production of fatty acids and fatty acid derivatives by recombinant microorganisms expressing polypeptides having lipolytic activity |
US8846370B2 (en) | 2010-12-23 | 2014-09-30 | Exxonmobil Research And Engineering Company | Genetically engineered microorganisms comprising 4-hydroxybenzoyl-coa thioesterases and methods of using the same for producing free fatty acids and fatty acid derivatives |
US8940508B2 (en) | 2010-12-31 | 2015-01-27 | Exxonmobil Research And Engineering Company | Enhancement of biomass production by disruption of light energy dissipation pathways |
US9096834B2 (en) | 2012-02-24 | 2015-08-04 | Exxonmobil Research And Engineering Company | Recombinant microorganisms comprising thioesterase and lysophosphatidic acid acyltransferase genes for fatty acid production |
WO2013162648A1 (en) | 2012-04-23 | 2013-10-31 | Exxonmobil Research And Engineering Company | Cell systems and methods for improving fatty acid synthesis by expression of dehydrogenases |
US9181568B2 (en) | 2012-04-23 | 2015-11-10 | Exxonmobil Research And Engineering Company | Cell systems and methods for improving fatty acid synthesis by expression of dehydrogenases |
WO2013164095A1 (en) | 2012-05-02 | 2013-11-07 | Algenics | Production of secreted therapeutic antibodies in microalgae |
EP2660324A1 (en) | 2012-05-02 | 2013-11-06 | Algenics | Production of secreted therapeutic antibodies in microalgae |
EP2660323A1 (en) | 2012-05-02 | 2013-11-06 | Algenics | Production of secreted therapeutic antibodies in phaeodactylum tricornutum microalgae |
WO2014062163A1 (en) | 2012-10-16 | 2014-04-24 | Exxonmobil Research And Engineering Company | Dgat genes and methods of use for triglyceride production in recombinant microorganisms |
US9328336B2 (en) | 2012-10-16 | 2016-05-03 | Exxonmobil Research And Engineering Company | DGAT genes and methods of use for triglyceride production in recombinant microorganisms |
WO2014089533A2 (en) | 2012-12-06 | 2014-06-12 | Synthetic Genomics, Inc. | Algal mutants having a locked-in high light acclimated phenotype |
EP3401388A1 (en) | 2012-12-06 | 2018-11-14 | Synthetic Genomics, Inc. | Algal mutants having a locked-in high light acclimated phenotype |
US8835149B2 (en) | 2012-12-06 | 2014-09-16 | Exxonmobil Research And Engineering Company | DGAT genes comprising pleckstrin homology domains and methods of use for triglyceride production in recombinant microorganisms |
WO2017160573A1 (en) | 2016-03-18 | 2017-09-21 | Exxonmobil Research And Engineering Company | Chlorophyllase overproduction to enhance photosynthetic efficiency |
EP3579845A4 (en) * | 2017-02-09 | 2020-12-30 | Dow Agrosciences LLC | Novel eukaryotic cell-free expression system that does not require an artificial energy regeneration system |
Also Published As
Publication number | Publication date |
---|---|
CA2483337A1 (en) | 2003-11-06 |
KR20040106384A (en) | 2004-12-17 |
MXPA04010432A (en) | 2005-05-27 |
CN1886512B (en) | 2015-11-25 |
EP1576101A2 (en) | 2005-09-21 |
JP2005537784A (en) | 2005-12-15 |
EP1576101A4 (en) | 2007-10-31 |
WO2003091413A3 (en) | 2006-06-29 |
AU2003239182B2 (en) | 2008-11-06 |
CA2483337C (en) | 2015-10-27 |
IL164549A0 (en) | 2005-12-18 |
IL164549A (en) | 2010-12-30 |
AU2003239182A1 (en) | 2003-11-10 |
US20040014174A1 (en) | 2004-01-22 |
KR101107380B1 (en) | 2012-01-19 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2003239182B2 (en) | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same | |
US8932576B2 (en) | Robust expression of a bioactive mammalian protein in Chlamydomonas chloroplast | |
KR102010609B1 (en) | Insect resistant and herbicide tolerant soybean event 9582.814.19.1 | |
RU2707527C2 (en) | Maize plant dbn9936 and method of using in detection nucleic acid sequence thereof | |
CA2843172C (en) | Soybean event pdab9582.814.19.1 detection method | |
RU2679510C2 (en) | Fluorescence activated cell sorting (facs) enrichment to generate plants | |
KR101439568B1 (en) | Zinc finger nuclease-mediated homologous recombination | |
US9139842B2 (en) | Methods and compositions for targeting sequences of interest to the chloroplast | |
CN102939384B (en) | Genes conferring stress tolerance in plants and uses thereof | |
US20090205075A1 (en) | Use of plastid transit peptides derived from glaucocystophytes | |
CN105264067A (en) | Fad3 performance loci and corresponding target site specific binding proteins capable of inducing targeted breaks | |
US9284569B2 (en) | Autoluminescent phytosensor plants and uses thereof | |
CN104219949A (en) | Synthetic chloroplast transit peptides | |
CN105263312A (en) | Methods and compositions for integration of an exogenous sequence within the genome of plants | |
MX2014009113A (en) | Pathogen resistant citrus compositions, organisms, systems, and methods. | |
US20140366219A1 (en) | Increasing Soybean Defense Against Pests | |
US20150101081A1 (en) | Plant transformant, transformation method for plant, and vector used in said method | |
AU2008202053B2 (en) | Expression of polypeptides in chloroplasts, and compositions and methods for expressing same | |
CA3236641A1 (en) | Error prone dna polymerase for organelle mutation | |
CN108473997B (en) | Plant promoters for transgene expression | |
RU2818368C2 (en) | Nucleic acid sequence for detecting soya plant dbn8002 and method for detecting same | |
CN109415420A (en) | For the plant promoter of transgene expression and 3 ' UTR |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
WWE | Wipo information: entry into national phase |
Ref document number: 3159/DELNP/2004 Country of ref document: IN |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2004/08498 Country of ref document: ZA Ref document number: 200408498 Country of ref document: ZA |
|
REEP | Request for entry into the european phase |
Ref document number: 2003733900 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2003733900 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: PA/a/2004/010432 Country of ref document: MX Ref document number: 2483337 Country of ref document: CA Ref document number: 1020047017113 Country of ref document: KR |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2003587949 Country of ref document: JP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2003239182 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 20038123924 Country of ref document: CN |
|
WWP | Wipo information: published in national office |
Ref document number: 1020047017113 Country of ref document: KR |
|
WWP | Wipo information: published in national office |
Ref document number: 2003733900 Country of ref document: EP |