WO2003048364A2 - Ovomucoid promoter and methods of use - Google Patents

Ovomucoid promoter and methods of use Download PDF

Info

Publication number
WO2003048364A2
WO2003048364A2 PCT/US2002/038413 US0238413W WO03048364A2 WO 2003048364 A2 WO2003048364 A2 WO 2003048364A2 US 0238413 W US0238413 W US 0238413W WO 03048364 A2 WO03048364 A2 WO 03048364A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
avian
cell
sequence
polypeptide
Prior art date
Application number
PCT/US2002/038413
Other languages
French (fr)
Other versions
WO2003048364A3 (en
Inventor
Alex J. Harvey
Youliang Wang
Original Assignee
Avigenics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP2003549541A priority Critical patent/JP2005536984A/en
Priority to CA2467648A priority patent/CA2467648C/en
Priority to US10/496,731 priority patent/US7375258B2/en
Priority to AU2002351192A priority patent/AU2002351192B2/en
Priority to EP02786838A priority patent/EP1461430A4/en
Application filed by Avigenics, Inc. filed Critical Avigenics, Inc.
Publication of WO2003048364A2 publication Critical patent/WO2003048364A2/en
Publication of WO2003048364A3 publication Critical patent/WO2003048364A3/en
Priority to US10/856,218 priority patent/US7294507B2/en
Priority to US11/047,184 priority patent/US7335761B2/en
Priority to US11/649,543 priority patent/US7507873B2/en
Priority to US12/313,064 priority patent/US7812215B2/en
Priority to US12/799,605 priority patent/US20100333219A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8135Kazal type inhibitors, e.g. pancreatic secretory inhibitor, ovomucoid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/30Bird
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid

Definitions

  • the present invention relates generally to the identification of an avian ovomucoid gene expression control region, particularly from the chicken. More specifically, the invention relates to recombinant nucleic acids and expression vectors, transfected cells and transgenic animals, especially chickens, that comprise the avian ovomucoid gene expression control region operably linked to a heterologous polypeptide-encoding nucleic acid.
  • transgenics The field of transgenics was initially developed to understand the action of a single gene in the context of the whole animal and the phenomena of gene activation, expression, and interaction. This technology has also been used to produce models for various diseases in humans and other animals and is amongst the most powerful tools available for the study of genetics, and the understanding of genetic mechanisms and function. From an economic perspective, the use of transgenic technology for the production of specific proteins or other substances of pharmaceutical interest (Gordon et al, Biotechnology 5: 1183-1187 (1987); Wilmut et al, Theriogenology 33: 113-123 (1990)) offers significant advantages over more conventional methods of protein production by gene expression.
  • Heterologous nucleic acids have been engineered so that an expressed protein may be joined to a protein or peptide that will allow secretion of the transgenic expression product into milk or urine, from which the protein may then be recovered. These procedures have had limited success and may require lactating animals, with the attendant costs of maintaining individual animals or herds of large species, including cows, sheep, or goats. Historically, transgenic animals have been produced almost exclusively by microinjection of the fertilized egg. The pronuclei of fertilized eggs are microinjected in vitro with foreign, i.e., xenogeneic or allogeneic, heterologous DNA or hybrid DNA molecules.
  • the microinjected fertilized eggs are then transferred to the genital tract of a pseudopregnant female (See e.g., Krimpenfort et al, in U.S. Pat. No. 5,175,384).
  • a pseudopregnant female See e.g., Krimpenfort et al, in U.S. Pat. No. 5,175,384.
  • One system that holds potential is the avian reproductive system.
  • the production of an avian egg begins with formation of a large yolk in the ovary of the hen.
  • the unfertilized oocyte or ovum is positioned on top of the yolk sac. After ovulation, the ovum passes into the infundibulum of the oviduct where it is fertilized, if sperm are present, and then moves into the magnum of the oviduct which is lined with tubular gland cells.
  • the hen oviduct offers outstanding potential as a protein bioreactor because of the high levels of protein production, the promise of proper folding and post-translation modification of the target protein, the ease of product recovery, and the shorter developmental period of chickens compared to other potential animal species. As a result, efforts have been made to create transgenic chickens expressing heterologous proteins in the oviduct.
  • Chicken oviduct cells when stimulated by steroid hormones during egg-laying, secrete three principal polypeptides, ovalbumin, ovomucoid and lysozyme (Tsai et al, Biochemistry 17: 5773-5779 (1978)).
  • the mRNA transcript encoding ovalbumin constitutes about 50% of the total mRNA of these cells.
  • Ovomucoid and lysozyme mRNAs contribute about 6.6% and 3.4% respectively of the total mRNA of the steroid stimulated cells. (Hynes et al, Cell 11:923 932 (1977)).
  • ovomucoid-encoding sequence includes seven intronic sequences (Lindenmaier et al, Nuc. Acid Res. 7;1221-1232 (1979); Catterall et al, Nature 278: 323-327 (1979); Lai et al, Cell 18:829-842 (1979)). Short stretches of the 5' flanking region of the ovomucoid gene have been sequenced (Lai et al, Cell 18: 829-842 (1979); Genbank Accession No. J00897), but extending only 579 bases upstream of the recognized transcription start site.
  • the ovoinhibitor-encoding cDNA and the attached 3 '-untranslated region, which extends into the 10 kb ovoinhibitor-ovomucoid region, were also sequenced (Scott et al. J. Biol. Chem. 262: 5899-5907 (1987)).
  • the chicken ovomucoid gene therefore, is highly expressed in the tubular glands of the mature hen oviduct and represents a suitable candidate for an efficient promoter for heterologous protein production in transgenic animals, especially animals.
  • the regulatory region of the ovomucoid locus extends over a nucleic acid region of about 10 kb of DNA 5' upstream of the transcription start site, and comprises at least one recognized element, the CR1.
  • the present invention relates to nucleic acids comprising an avian ovomucoid gene expression control region, which is useful for the expression of nucleotide sequences encoding a polypeptide of interest in a transfected avian cell such as, for example, an oviduct cell.
  • the polypeptide is heterologous, i.e., not the ovomucoid protein product, and more preferably, is a mammalian, most preferably, a human polypeptide.
  • One aspect of the present invention provides a nucleic acid isolated from a region immediately 5' upstream of a transcription start site of the chicken (or other avian) ovomucoid gene locus.
  • the nucleic acid comprises an avian nucleic acid sequence comprising an ovomucoid gene expression control region comprising at least one avian CR1 repeat element, and a proximal ovomucoid promoter. Interspersed between these constituent elements may be stretches of nucleic acid that may serve at least to organize the gene regulatory elements in an ordered array relative to a polypeptide-encoding region.
  • the ovomucoid gene expression control region is isolated from a chicken.
  • the ovomucoid gene expression control region has a nucleotide sequence of SEQ ID NO:26.
  • the ovomucoid gene expression control region is at least 75%, at least 95 %, or at least 99% identical to SEQ ID NO:26 and directs expression of a polypeptide encoding nucleotide sequence in an avian oviduct cell.
  • the avian ovomucoid gene expression control region of the present invention is useful for directing tissue-specific expression of a polypeptide-encoding nucleic acid.
  • the avian ovomucoid gene expression control region may be operably linked with a selected nucleic acid insert, wherein the nucleic acid insert encodes a polypeptide, preferably heterologous, desired to be expressed in a transfected cell.
  • the nucleic acid insert may be placed in frame with a nucleotide sequence encoding a signal peptide. Translation initiation may start with the signal peptide and continue through the nucleic acid insert, thereby producing an expressed polypeptide having the desired amino acid sequence.
  • the recombinant DNA of the present invention may further comprise a polyadenylation signal sequence that will allow the transcript directed by the ovomucoid gene expression control region of the invention to proceed beyond the nucleic acid insert encoding a heterologous polypeptide (i.e., not the ovomucoid protein that is expressed from the endogenous gene containing the ovomucoid gene expression control region) and allow the transcript to further comprise a 3 ' untranslated region and a polyadenylated tail.
  • Any functional polyadenylation signal sequence may be linked to the 3' end of the nucleic acid insert including the SV40 polyadenylation signal sequence, bovine growth hormone adenylation sequence or the like.
  • the nucleic acid of the invention may also comprise gene expression control elements, e.g. promoters, enhancers, IRES's, from other than an ovomucoid gene and may even be from a non-avian gene.
  • the sequence of the expressed nucleic acid insert may be optimized for codon usage by a host cell. This may be determined from the codon usage of at least one, and preferably more than one, protein expressed in a chicken cell. For example, the codon usage may be determined from the nucleic acid sequences encoding the proteins ovalbumin, ovomucoid, ovomucin and ovotransferrin of chicken.
  • the expression vector of the present invention may comprise an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a non- ovomucoid polypeptide, and optionally, a polyadenylation signal sequence.
  • the expression vector may further comprise a bacterial plasmid sequence, a viral nucleic acid sequence, or fragments or variants thereof that may allow for replication of the vector in a suitable host.
  • the nucleic acid is a YAC, BAC, bacteriophage-derived artificial chromosome (BBPAC), cosmid or PI derived artificial chromosome (PAC).
  • the present invention further relates to nucleic acid vectors and transgenes inserted therein that incorporate multiple polypeptide-encoding regions, wherein a first polypeptide- encoding region is operatively linked to a transcription promoter and a second polypeptide- encoding region is operatively linked to an Internal Ribosome Entry Sequence (IRES).
  • the vector may contain coding sequences for two different heterologous proteins (e.g., the heavy and light chains of an immunoglobulin).
  • nucleic acid constructs when inserted into the genome of a bird and expressed therein, will generate individual polypeptides that may be post-translationally modified, for example, glycosylated or, in certain embodiments, be present as complexes, such as heterodimers with each other.
  • Another aspect of the present invention is a method of expressing a heterologous polypeptide in a eukaryotic cell by transfecting the cell with a recombinant DNA comprising an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding the heterologous polypeptide and, optionally, a polyadenylation signal sequence, and culturing the transfected cell in a medium suitable for expression of the heterologous polypeptide under the control of the avian ovomucoid gene expression control region.
  • the polypeptide is a cytokine, growth factor, enzyme, structural protein, and more preferably, an immunoglobulin, or subunit thereof.
  • the polypeptide is a mammalian, preferably a human polypeptide or derived from a human or mammalian polypeptide.
  • the level of expression of the heterologous protein is greater than 5 ⁇ g, 10 ⁇ g, 50 ⁇ g, 100 ⁇ g, 250 ⁇ g, 500 ⁇ g, or 750 ⁇ g, more preferably greater than 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, 50 mg, 100 mg, 200 mg, 500 mg, 700 mg, 1 gram, 2 grams, 3 grams, 4 grams or 5 grams in an egg (preferably the egg white) produced by the transgenic avian of the invention.
  • the transformed cell is a chicken oviduct cell and the nucleic acid comprises the chicken ovomucoid gene expression control region, a nucleic acid insert encoding a heterologous polypeptide of interest, e.g. human interferon ⁇ 2, which optionally is codon optimized for expression in an avian cell, and an SV40 polyadenylation sequence.
  • a heterologous polypeptide of interest e.g. human interferon ⁇ 2
  • an SV40 polyadenylation sequence e.g. human interferon ⁇ 2
  • animal is used herein to include all vertebrate animals, including humans. It also includes an individual animal in all stages of development, including embryonic and fetal stages.
  • avian refers to any species, subspecies or race of organism of the taxonomic class ava, such as, but not limited to, such organisms as chicken, turkey, duck, goose, quail, pheasants, parrots, finches, hawks, crows and ratites including ostrich, emu and cassowary.
  • nucleic acid refers to any natural and synthetic linear and sequential arrays of nucleotides and nucleosides, for example cDNA, genomic DNA, mRNA, tRNA, oligonucleotides, oligonucleosides and derivatives thereof.
  • nucleic acids of the present invention include bacterial plasmid vectors including expression, cloning, cosmid and transformation vectors such as, but not limited to, pBR322, animal viral vectors such as, but not limited to, modified adenovirus, influenza virus, polio virus, pox virus, retrovirus, and the like, vectors derived from bacteriophage nucleic acid, e.g., plasmids and cosmids, artificial chromosomes, such as but not limited to, Yeast Artificial Chromosomes (YACs) and Bacterial Artificial Chromosomes (BACs), and synthetic oligonucleotides like chemically synthesized DNA or RNA.
  • YACs Yeast Artificial Chromosomes
  • BACs Bacterial Artificial Chromosomes
  • nucleic acid further includes modified or derivatised nucleotides and nucleosides such as, but not limited to, halogenated nucleotides such as, but not only, 5-bromouracil, and derivatised nucleotides such as biotin-labeled nucleotides.
  • isolated nucleic acid refers to a nucleic acid that has been removed from other components of the cell containing the nucleic acid or from other components of chemical/synthetic reaction used to generate the nucleic acid. In specific embodiments, the nucleic acid is 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100% pure.
  • isolated nucleic acid does not include nucleic acids that are members of a library, e.g. cDNA or genomic library, unless identified and separated from the other members of the library.
  • nucleic acids and proteins of the present invention are well known to those of skill in the art and standard molecular biology and biochemical manuals may be consulted to select suitable protocols without undue experimentation. See, for example, Sambrook et al, 2001, Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Press; the content of which is herein incorporated by reference in its entirety.
  • enriched in reference to nucleic acid it is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction of the total DNA or RNA present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken.
  • Enriched does not imply that there are no other DNA or RNA sequences present, just that the relative amount of the sequence of interest has been significantly increased, for example, by 1 fold, 2 fold, 5 fold, 10 fold, 50 fold, 100 fold, 500 fold, 1000 fold, 10,000 fold, 100,000 fold, or 1,000,000 fold.
  • the other DNA may, for example, be derived from a yeast or bacterial genome, or a cloning vector, such as a plasmid or a viral vector. It is advantageous for some purposes that a nucleotide sequence is in purified form.
  • nucleic acid in reference to nucleic acid represents that the sequence has increased purity relative to the natural environment, preferably, 50%, 60%, 70%, 80%, 90%, 95%, or 99% pure.
  • polynucleotide and nucleic acid sequence are used interchangeably herein and include, but are not limited to, coding sequences (polynucleotide(s) or nucleic acid sequence(s) which are transcribed and translated into polypeptide in vitro or in vivo when placed under the control of appropriate regulatory or control sequences); control sequences (e.g., translational start and stop codons, promoter sequences, ribosome binding sites, polyadenylation signals, transcription factor binding sites, transcription termination sequences, upstream and downstream regulatory domains, enhancers, silencers, and the like); and regulatory sequences (DNA sequences to which a transcription factor(s) binds and alters the activity of a gene's promoter either positively (induction) or negatively (repression)).
  • control sequences e.g., translational start and stop codons, promoter sequences, ribosome binding sites, polyadenylation signals, transcription factor binding sites, transcription termination sequences, upstream and downstream regulatory domains
  • polypeptide and protein refer to a polymer of amino acids of three or more amino acids in a serial array, linked through peptide bonds.
  • polypeptide includes proteins, protein fragments, protein analogues, oligopeptides and the like.
  • polypeptides contemplates polypeptides as defined above that are encoded by nucleic acids, produced through recombinant technology (isolated from an appropriate source such as a bird), or synthesized.
  • polypeptides further contemplates polypeptides as defined above that include chemically modified amino acids or amino acids covalently or noncovalently linked to labeling ligands.
  • fragment refers to an at least 10, 20, 50, 75, 100, 150, 200, 250, 300, 500, 1000, 2000 or 5000 nucleotide long portion of a nucleic acid (e.g., cDNA) that has been constructed artificially (e.g., by chemical synthesis) or by cleaving a natural product into multiple pieces, using restriction endonucleases or mechanical shearing, or enzymatically, for example, by PCR or any other polymerizing technique known in the art, or expressed in a host cell by recombinant nucleic acid technology known to one of skill in the art.
  • a nucleic acid e.g., cDNA
  • fragment as used herein may also refer to an at least 5, 10, 20, 30, 40, 50, 75, 100, 150, 200, 250, 300, 400, 500, 1000, 2000, or 5000 amino acid portion of a polypeptide, which portion is cleaved from a naturally occurring polypeptide by proteolytic cleavage by at least one protease, or is a portion of the naturally occurring polypeptide synthesized by chemical methods or using recombinant DNA technology (e.g., expressed from a portion of the nucleotide sequence encoding the naturally occurring polypeptide) known to one of skill in the art.
  • gene refers to nucleic acid sequences (including both RNA or DNA) that encode genetic information for the synthesis of a whole RNA, a whole protein, or any portion of such whole RNA or whole protein. Genes that are not naturally part of a particular organism's genome are referred to as “foreign genes,” “heterologous genes” or “exogenous genes” and genes that are naturally a part of a particular organism's genome are referred to as “endogenous genes”.
  • gene product refers to RNAs or proteins that are encoded by the gene.
  • RNA or proteins encoded by “foreign genes” are RNA or proteins encoded by "foreign genes” and “endogenous gene products” are RNA or proteins encoded by endogenous genes.
  • endogenous gene products are RNA or proteins encoded by endogenous genes.
  • heterologous gene products are RNAs or proteins encoded by “foreign, heterologous or exogenous genes” and are, therefore, not naturally expressed in the cell.
  • RNA nucleic acid molecule at least complementary in part to a region of one of the two nucleic acid strands of the gene.
  • expression also refers to the translation from said RNA nucleic acid molecule to give a protein, a polypeptide or a portion thereof.
  • locus refers to the site of a gene on a chromosome. Pairs of genes control hereditary traits, each in the same position on a pair of chromosomes. These gene pairs, or alleles, may both be dominant or both be recessive in expression of that trait. In either case, the individual is said to be homozygous for the trait controlled by that gene pair. If the gene pair (alleles) consists of one dominant and one recessive trait, the individual is heterozygous for the trait controlled by the gene pair.
  • allelic variants Natural variation in genes or nucleic acid molecules caused by, for example, recombination events or resulting from mutation, gives rise to allelic variants with similar, but not identical, nucleotide sequences. Such allelic variants typically encode proteins with similar activity to that of the protein encoded by the gene to which they are compared, because natural selection typically selects against variations that alter function. Allelic variants can also comprise alterations in the untranslated regions of the gene as, for example, in the 3' or 5' untranslated regions or can involve alternate splicing of a nascent transcript, resulting in alternative exons being positioned adjacently.
  • the terms "operably linked” or "operatively linked” refer to the configuration of the coding and control sequences so as to perform the desired function.
  • control sequences operably linked to a coding sequence are capable of effecting the expression of the coding sequence and regulating in which tissues, at what developmental timepoints, or in response to which signals, etc., a gene is expressed.
  • a coding sequence is operably linked to or under the control of transcriptional regulatory regions in a cell when DNA polymerase will bind the promoter sequence and transcribe the coding sequence into mRNA that can be translated into the encoded protein.
  • the control sequences need not be contiguous with the coding sequence, so long as they function to direct the expression thereof. Thus, for example, intervening untranslated yet transcribed sequences, can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered "operably linked" to the coding sequence.
  • transcription regulatory sequences and “gene expression control regions” as used herein refer to nucleotide sequences that are associated with a gene nucleic acid sequence and which regulate the transcriptional expression of the gene.
  • exemplary transcription regulatory sequences include enhancer elements, hormone response elements, steroid response elements, negative regulatory elements, and the like.
  • the "transcription regulatory sequences” may be isolated and incorporated into a nucleic acid vector to enable regulated transcription in appropriate cells of portions of the vector DNA.
  • transcription regulatory sequence may precede, but is not limited to, the region of a nucleic acid sequence that is in the region 5' of the end of a protein coding sequence that may be transcribed into mRNA.
  • Transcriptional regulatory sequences may also be located within a protein coding region, in regions of a gene that are identified as "intron” regions, or may be in regions of nucleic acid sequence that are in the region of nucleic acid.
  • promoter refers to the DNA sequence that determines the site of transcription initiation by an RNA polymerase.
  • a "promoter-proximal element” may be a regulatory sequence within about 200 base pairs of the transcription start site.
  • a "magnum-specific” promoter as used herein, is a promoter that is primarily or exclusively active in the tubular gland cells of the avian magnum. Useful promoters also include exogenously inducible promoters. These are promoters that can be “turned on” in response to an exogenously supplied agent or stimulus, which is generally not an endogenous metabolite or cytokine.
  • antibiotic-inducible promoter such as a tetracycline- inducible promoter, a heat-inducible promoter, a light- inducible promoter, or a laser inducible promoter
  • a tetracycline- inducible promoter such as a tetracycline- inducible promoter, a heat-inducible promoter, a light- inducible promoter, or a laser inducible promoter
  • Halloran et al 2000, Development 127: 1953-1960
  • coding region refers to a continuous linear arrangement of nucleotides which may be translated into a protein.
  • a full length coding region is translated into a full length protein; that is, a complete protein as would be translated in its natural state absent any post-translational modifications.
  • a full length coding region may also include any leader protein sequence or any other region of the protein that may be excised naturally from the translated protein.
  • complementary refers to two nucleic acid molecules that can form specific interactions with one another.
  • an adenine base within one strand of a nucleic acid can form two hydrogen bonds with thymine within a second nucleic acid strand when the two nucleic acid strands are in opposing polarities.
  • a guanine base within one strand of a nucleic acid can form three hydrogen bonds with cytosine within a second nucleic acid strand when the two nucleic acid strands are in opposing polarities.
  • Complementary nucleic acids as referred to herein may further comprise modified bases wherein a modified adenine may form hydrogen bonds with a thymine or modified thymine, and a modified cytosine may form hydrogen bonds with a guanine or a modified guanine.
  • probe when referring to a nucleic acid, refers to a nucleotide sequence that can be used to hybridize with and thereby identify the presence of a complementary sequence, or a complementary sequence differing from the probe sequence but not to a degree that prevents hybridization under the hybridization stringency conditions used.
  • the probe may be modified with labels such as, but not only, radioactive groups, biotin, and the like that are well known in the art.
  • hybridizing under stringent conditions refers to annealing a first nucleic acid to a second nucleic acid under stringent conditions as defined below.
  • Stringent hybridization conditions typically permit the hybridization of nucleic acid molecules having at least 70% nucleic acid sequence identity with the nucleic acid molecule being used as a probe in the hybridization reaction.
  • the first nucleic acid may be a test sample or probe
  • the second nucleic acid may be the sense or antisense strand of a ovomucoid gene expression control region or a fragment thereof.
  • Hybridization of the first and second nucleic acids may be conducted under stringent conditions, e.g., high temperature and/or low salt content that tend to disfavor hybridization of dissimilar nucleotide sequences.
  • hybridization of the first and second nucleic acid maybe conducted under stringent conditions, e.g., high temperature and/or low salt content that tend to disfavor hybridization of dissimilar nucleotide sequences.
  • hybridization of the first and second nucleic acid maybe conducted under stringent conditions, e.g., high temperature and/or low salt content that tend to disfavor hybridization of dissimilar nucleotide sequences.
  • hybridization of the first and second nucleic acid maybe conducted under
  • 5 reduced stringency conditions e.g. low temperature and/or high salt content that tend to favor hybridization of dissimilar nucleotide sequences.
  • Low stringency hybridization conditions may be followed by high stringency conditions or intermediate medium stringency conditions to increase the selectivity of the binding of the first and second nucleic acids.
  • the hybridization conditions may further include reagents such as, but not limited to, dimethyl
  • a suitable hybridization protocol may, for example, involve hybridization in 6X SSC (wherein IX SSC comprises 0.015 M sodium citrate and 0.15 M sodium chloride), at 65° C in an aqueous solution, followed by washing with IX SSC at 65° C.
  • 6X SSC wherein IX SSC comprises 0.015 M sodium citrate and 0.15 M sodium chloride
  • IX SSC comprises 0.015 M sodium citrate and 0.15 M sodium chloride
  • stringent conditions will be those in which the salt concentration is less than about 1.5 M sodium ion, typically about 0.01 to 1.0 M Na ion concentration (or other
  • exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCl, 1% SDS (sodium dodecyl sulphate) at
  • Exemplary moderate stringency conditions include hybridization in 40 to 45 % formamide, 1 M NaCl, 1% SDS at 37° Celsius, and a wash in 0.5x to lx SSC at 55 to 60° Celsius.
  • Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° Celsius, and a wash in 0.1 x SSC at 60 to 65° Celsius.
  • 5 nucleic acid region as referred to herein is a nucleotide sequence present in two or more nucleic acid sequences, to which a particular nucleic acid sequence can hybridize under low, medium or high stringency conditions.
  • percent sequence identity or “percent sequence similarity” as used herein refer to the degree of sequence identity between two nucleic acid sequences or two amino acid sequences as determined using the algorithm of Karlin & Attschul (1990) Proc. Natl. Acad. Sci. 87: 2264-2268, modified as in Karlin & Attschul (1993) Proc. Natl. Acad. Sci. 90:
  • NBLAST and XBLAST programs of Attschul et al. (1990) T. Mol. Biol. Q15: 403-410.
  • Gapped BLAST is utilized as described in Attschul et al. (1997) Nucl. Acids Res. 25: 3389-3402.
  • the default parameters of the respective programs e.g. XBLAST and NBLAST
  • Other algorithms, programs and default settings may also be suitable such as, but not only, the 5 GCG-Sequence Analysis Package of the U.K. Human Genome Mapping Project Resource Centre that includes programs for nucleotide or amino acid sequence comparisons.
  • sense strand refers to a single stranded DNA molecule from a genomic DNA that may be transcribed into RNA and translated into the natural polypeptide product of the gene.
  • antisense strand refers to the single strand 0 DNA molecule of a genomic DNA that is complementary with the sense strand of the gene.
  • antisense DNA refers to a gene sequence DNA that has a nucleotide sequence complementary to the "sense strand" of a gene when read in reverse orientation, i.e., DNA read into RNA in a 3' to 5' direction rather than in the 5' to 3' direction.
  • antisense RNA is used to mean an RNA nucleotide sequence (for example that 5 encoded by an antisense DNA or synthesized complementary with the antisense DNA). Antisense RNA is capable of hybridizing under stringent conditions with an antisense DNA.
  • the antisense RNA of the invention is useful for regulating expression of a "target gene" either at the transcriptional or translational level.
  • transcription of the subject nucleic acids may produce antisense transcripts that are capable of inhibiting transcription by inhibiting initiation of transcription or by competing for limiting transcription factors; the antisense transcripts may inhibit transport of the "target RNA", or, the antisense transcripts may inhibit translation of "target RNA”.
  • nucleic acid vector refers to a natural or synthetic single or double stranded plasmid or viral nucleic acid molecule, or any other nucleic acid molecule, such as but not limited to YACs, BACs, bacteriophage-derived artificial chromosome (BBPAC), cosmid or PI derived artificial chromosome (PAC), that can be transfected or transformed into cells and replicate independently of, or within, the host cell genome.
  • a circular double stranded vector can be linearized by treatment with an appropriate restriction enzyme based on the nucleotide sequence of the vector.
  • a nucleic acid can be inserted into a vector by cutting the vector with restriction enzymes and ligating the pieces together.
  • the nucleic acid molecule can be RNA or DNA.
  • expression vector refers to a nucleic acid vector that comprises the ovomucoid gene expression control region operably linked to a nucleotide sequence coding at least one polypeptide.
  • regulatory sequences includes promoters, enhancers, and other elements that may control gene expression. Standard molecular biology textbooks such as Sambrook et al. eds "Molecular Cloning: A Laboratory Manual” 3rd ed., Cold Spring Harbor Press (2001) maybe consulted to design suitable expression vectors that may further include an origin of replication and selectable gene markers. It should be recognized, however, that the choice of a suitable expression vector and the combination of functional elements therein depends upon multiple factors including the choice of the host cell to be transformed and/or the type of protein to be expressed.
  • transformation and “transfection” as used herein refer to the process of inserting a nucleic acid into a host.
  • Many techniques are well known to those skilled in the art to facilitate transformation or transfection of a nucleic acid into a prokaryotic or eukaryotic organism. These methods involve a variety of techniques, such as treating the cells with high concentrations of salt such as, but not only, a calcium or magnesium salt, an electric field, detergent, or liposome mediated transfection, to render the host cell competent for the uptake of the nucleic acid molecules, and by such methods as sperm-mediated and restriction-mediated integration.
  • salt such as, but not only, a calcium or magnesium salt, an electric field, detergent, or liposome mediated transfection
  • transfecting agent refers to a composition of matter added to the genetic material for enhancing the uptake of heterologous DNA segment(s) into a eukaryotic cell, preferably an avian cell. The enhancement is measured relative to the uptake in the absence of the transfecting agent.
  • transfecting agents include adenovirus-transferrin-polylysine-DNA complexes. These complexes generally augment the uptake of DNA into the cell and reduce its breakdown during its passage through the cytoplasm to the nucleus of the cell. These complexes can be targeted to, e.g., the male germ cells using specific ligands that are recognized by receptors on the cell surface of the germ cell, such as the c-kit ligand or modifications thereof.
  • transfecting agents include but are not limited to lipofectin, lipfectamine, DIMRIE C, Supeffect, and Effectin (Qiagen), unifectin, maxifectin, DOTMA, DOGS (Transfectam; dioctadecylamidoglycylspermine), DOPE (1,2-dioleoyl-sn-glycero- 3-phosphoethanolamine), DOTAP (l,2-dioleoyl-3-trimethylammonium propane), DDAB (dimethyl dioctadecytammonium bromide), DHDEAB (N,N-di-n-hexadecyl-N,N- dihydroxyethyl ammonium bromide), HDEAB (N-n-hexadecylN,N- dihydroxyethylammonium bromide), polybrene, or poly(ethylenimine) (PEI).
  • These non- viral agents have the advantage that they can facilitate stable integration of
  • recombinant cell refers to a cell that has a new combination of nucleic acid segments that are not covalently linked to each other in nature in that particular configuration.
  • a new configuration of nucleic acid segments can be introduced into an organism using a wide array of nucleic acid manipulation techniques available to those skilled in the art.
  • a recombinant cell can be a single eukaryotic cell, such as a mammalian or avian cell (including within a transgenic mammal or avian) or a single prokaryotic cell.
  • the recombinant cell may harbor a vector that is extragenomic. An extragenomic nucleic acid vector does not insert into the cell's genome.
  • a recombinant cell may further harbor a vector or a portion thereof (e.g., the portion containing the regulatory sequences and the coding sequence) that is intragenomic.
  • the term intragenomic defines a nucleic acid construct incorporated within the recombinant cell's genome.
  • nucleic acid and “recombinant DNA” as used herein refer a combination of at least two nucleic acids that is not naturally found in a eukaryotic or prokaryotic cell in that particular configuration.
  • the nucleic acids may include, but are not limited to, nucleic acid vectors, gene expression regulatory elements, origins of replication, suitable gene sequences that when expressed confer antibiotic resistance, protein-encoding sequences and the like.
  • recombinant polypeptide is meant to include a polypeptide produced by recombinant DNA techniques such that it is distinct from a naturally occurring polypeptide either in its location, purity or structure.
  • compositions comprising agents that will modulate the regulation of the expression of a polypeptide-encoding nucleic acid operably linked to a ovomucoid gene expression control region can be administered in dosages and by techniques well known to those skilled in the medical or veterinary arts, taking into consideration such factors as the age, sex, weight, species and condition of the recipient animal, and the route of administration.
  • the route of administration can be percutaneous, via mucosal administration (e.g., oral, nasal, anal, vaginal) or via a parenteral route (intradermal, intramuscular, subcutaneous, intravenous, or intraperitoneal).
  • compositions can be administered alone, or can be co-administered or sequentially administered with other treatments or therapies.
  • Forms of administration may include suspensions, syrups or elixirs, and preparations for parenteral, subcutaneous, intradermal, intramuscular or intravenous administration (e.g., injectable administration) such as sterile suspensions or emulsions.
  • Pharmaceutical compositions may be administered in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, or the like.
  • compositions can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, adjuvants, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, adjuvants, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • Standard pharmaceutical texts such as Remmington's Pharmaceutical Science, 17th edition, 1985 may be consulted to prepare suitable preparations, without undue experimentation. Dosages can generally range from a few hundred milligrams to a few grams.
  • a "transgenic animal” is any non-human animal, such as an avian species, including the chicken, in which one or more of the cells of the animal contain a heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art.
  • the nucleic acid is introduced into a cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus.
  • the term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA.
  • the transgene causes cells to express a recombinant form of the subject polypeptide, e.g. either agonistic or antagonistic forms, or in which the gene has been disrupted.
  • the genome of the animal has been modified such that a heterologous gene expression element is inserted so as to be operably linked to an endogenous coding sequence.
  • chimeric animal or “mosaic animal” are used herein to refer to animals in which the recombinant gene is found, or in which the recombinant gene is expressed in some but not all cells of the animal.
  • tissue-specific chimeric animal indicates that the recombinant gene is present and/or expressed in some tissues but not others.
  • transgene means a nucleic acid sequence (encoding, for example, a human interferon polypeptide) that is partly or entirely heterologous, i.e., foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted (e.g., it is inserted at a location that differs from that of the natural gene or its insertion results in a knockout).
  • a trangene also includes a regulatory sequence designed to be inserted into the genome such that it regulates the expression of an endogenous coding sequence, e.g., to increase expression and or to change the timing and or tissue specificity of expression, etc. (e.g., to effect "gene activation”).
  • the term "cytokine” as used herein refers to any secreted polypeptide that affects the functions of cells and is a molecule that modulates interactions between cells in the immune, inflammatory or hematopoietic responses.
  • a cytokine includes, but is not limited to, monokines and lymphokines regardless of which cells produce them.
  • a monokine is generally referred to as being produced and secreted by a mononuclear cell, such as a macrophage and/or monocyte.
  • a mononuclear cell such as a macrophage and/or monocyte.
  • Many other cells however also produce monokines, such as natural killer cells, fibroblasts, basophils, neutrophils, endothelial cells, brain astrocytes, bone marrow stromal cells, epideral keratinocytes and B-lymphocytes.
  • Lymphokines are generally referred to as being produced by lymphocyte cells.
  • cytokines include, but are not limited to, Interleukin-1 (IL-1), Interleukin-6 (IL-6), Interleukin-8 (IL-8), Tumor Necrosis Factor-alpha (TNF-alpha) and Tumor Necrosis Factor beta (TNF-beta).
  • IL-1 Interleukin-1
  • IL-6 Interleukin-6
  • IL-8 Interleukin-8
  • TNF-alpha Tumor Necrosis Factor-alpha
  • TNF-beta Tumor Necrosis Factor beta
  • antibody refers to polyclonal and monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof.
  • antibody refers to a homogeneous molecular entity, or a mixture such as a polyclonal serum product made up of a plurality of different molecular entities, and may further comprise any modified or derivatised variant thereof that retains the ability to specifically bind an epitope.
  • a monoclonal antibody is capable of selectively binding to a target antigen or epitope.
  • Antibodies may include, but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, camelized antibodies, single chain antibodies (scFvs), Fab fragments, F(ab') 2 fragments, disulfide-linked Fvs (sdFv) fragments, e.g., as produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, intrabodies, synthetic antibodies, and epitope-binding fragments of any of the above.
  • immunoglobulin polypeptide refers to a polypeptide derived from a constituent polypeptide of an immunoglobulin.
  • An “immunoglobulin polypeptide” may be, but is not limited to, an immunoglobulin (preferably an antibody) heavy or light chain and may include a variable region, a diversity region, joining region and a constant region or any combination, variant or truncated form thereof.
  • immunoglobulin polypeptides further includes single-chain antibodies comprised of, but not limited to, an immunoglobulin heavy chain variable region, an immunoglobulin light chain variable region and optionally a peptide linker.
  • Abbreviations used in the present specification include the following: aa, amino acid(s); bp, base pair(s); cDNA, DNA complementary to RNA; nt, nucleotide(s); SSC, sodium chloride-sodium citrate; DMSO, dimethyl sulfoxide.
  • FIG. 1 illustrates an agarose gel analysis of PCR products from PCR amplification of chicken genomic DNA using the primers OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2).
  • FIG. 2 illustrates the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site, and the positions and orientations of primers used to sequence this region.
  • FIG. 3 shows the PCR primers SEQ ID NOS: 1 - 25 used to PCR amplify and/or sequence the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site.
  • FIG. 4 shows the nucleic acid sequence SEQ ID NO: 26 of the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site.
  • FIG. 5 illustrates the 10 kb ovomucoid promoter linked to the luciferase or human IFN ⁇ -2b coding sequences.
  • FIG. 6 A shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a luciferase gene into HDl 1 cells, a chicken myeloid cell line.
  • FIG. 6B shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a luciferase gene into primary quail tubular gland cells isolated from the magnum portion of the oviduct of a laying quail hen.
  • FIG. 7 shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a interferon gene into primary quail tubular gland cells isolated from the magnum portion of the oviduct of a laying quail hen.
  • a series of PCR amplifications of template chicken genomic DNA were used to isolate the gene expression control region of the chicken ovomucoid locus.
  • the region of the chicken genome lying between the 3' end of the ovoinhibitor gene and the 5' transcription start site of the ovomucoid gene was PCR amplified using the primers OVINs 2,
  • SEQ ID NO: 26 The compiled nucleic acid sequence (SEQ ID NO: 26) of the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site is shown in FIG. 4.
  • SEQ ID NO: 26 includes the ovoinhibitor gene 3' untranslated region described by
  • any nucleic acid sequence encoding a polypeptide may be operably linked to the avian ovomucoid gene expression control region of the invention so as to be expressed in a transfected avian cell.
  • a plasmid construct contacting the cloned ovomucoid promoter region and a desired polypeptide-encoding nucleic acid sequence may be transfected into cultured quail or chicken oviduct cells, which may then be incubated to synthesize a polypeptide detectable with antibodies directed against the desired polypeptide.
  • the present invention can be used to express, in large yields and at low cost, a wide range of desired proteins including those used as human and animal pharmaceuticals, diagnostics, and livestock feed additives.
  • Proteins such as growth hormones, cytokines, structural proteins and enzymes, including human growth hormone, interferon, lysozyme, and ⁇ -casein, are examples of proteins that are desirably expressed in the oviduct and deposited in eggs according to the invention.
  • proteins to be produced include, but are not limited to, albumin, ⁇ -1 antitrypsin, antithrombin m, collagen, factors VIH, IX, X (and the like), fibrinogen, hyaluronic acid, insulin, lactoferrin, protein C, erythropoietin (EPO), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), tissue-type plasminogen activator (tPA), feed additive enzymes, somatotropin, and chymotrypsin Immunoglobulins and genetically engineered antibodies, including immunotoxins that bind to surface antigens on human tumor cells and destroy them, can also be expressed for use as pharmaceuticals or diagnostics.
  • immunoglobulin polypeptides expressed in avian cells following transfection by the methods of the present invention may include monomeric heavy and light chains, single-chain antibodies or multimeric immunoglobulins comprising variable heavy and light chain regions, i.e., antigen-binding domains, or intact heavy and light immunoglobulin chains.
  • the chicken ovomucoid gene expression control region of the present invention comprises the nucleotide elements that are positioned 5' upstream of the transcription start site of the native chicken ovomucoid locus and which are necessary for the regulated expression of a downstream polypeptide-encoding nucleic acid. It is contemplated that this region includes those transcription control regions regulatable by hormones including, for example, steroid hormones and the like.
  • One aspect of the present invention provides a novel isolated nucleic acid that comprises the nucleotide sequence SEQ ID NO: 26, shown in FIG. 4, (Genbank Accession No: AF 453747) and derivatives and variants thereof, that is located immediately 5' upstream of the transcription start site of the chicken ovomucoid gene locus.
  • the isolated nucleic acid may be isolated from an avian selected from the group consisting of a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird.
  • the isolated nucleic acid is obtained from a chicken.
  • the isolated nucleic acid has the sequence of SEQ ID NO: 26, as shown in FIG. 4, or a variant thereof. SEQ ID NO: 26 was cloned into pBluescript KS ⁇ (+/-) vector, as described in Example 2, and named pBS-OVMUP-10.
  • pBS-OVMUP-10 was deposited with American Type Culture Collection (ATCC) as ATCC No. on November 26, 2002 under the conditions set forth in the Budapest Treaty.
  • ATCC American Type Culture Collection
  • Another aspect of the invention provides nucleic acids that can hybridize under high, medium or low stringency conditions to an isolated nucleic acid comprising a chicken ovomucoid gene expression control region having all, a derivative of, or a portion of the nucleic acid sequence SEQ ID NO: 26 shown in FIG. 4 and direct expression of a polypeptide coding sequence in an avian oviduct cell.
  • nucleotide sequence determined from the isolation of the ovomucoid gene expression control region from a chicken will allow for the generation of probes designed for use in identifying ovomucoid gene expression control regions, or homo logs thereof in other avian species. Fragments of a nucleic acid comprising a portion of the subject ovomucoid gene expression control region are also within the scope of the invention.
  • a fragment of the nucleic acid comprising an active portion of a ovomucoid gene expression control region refers to a nucleotide sequence having fewer nucleotides than the nucleotide sequence comprising the entire nucleic acid sequence of the ovomucoid gene expression control region.
  • a fragment of the ovomucoid gene expression control region may contain one or more of the following elements: the ovoinhibitor gene 3' untranslated region from bases positions 1-255 as shown in FIG. 4, a CRl-like element located at base positions 2761-3024 as shown in FIG. 4, the region from base positions 9403-9920, as shown in FIG. 4 which includes a portion of the 5' untranslated region of the ovomucoid gene.
  • the fragment may be 10, 20, 50, 75, 100, 150, 200, 250, 300, 500, 1000, 2000, 4000, 5000, 6000, 7000, 8000 or 9000 nucleotides in length and be capable of directing expression of an operably linked heterologous gene sequence, particularly in an avian oviduct cell.
  • the nucleotide sequence of the isolated DNA molecule of the present invention may be used as a probe in nucleic acid hybridization assays for the detection of the ovomucoid gene expression control region.
  • the nucleotide sequence of the present invention may be used in any nucleic acid hybridization assay system known in the art, including, but not limited to, Southern blots (Southern, EM. J. Mol. Biol. 98: 508 (1975)), Northern blots (Thomas et al, Proc. Natl. Acad. Sci. 77: 5201-05 (1980)), and Colony blots (Grunstein et al, Proc. Natl. Acad. Sci.
  • the isolated DNA molecules of the present invention can be used in a gene amplification detection procedure such as a polymerase chain reaction (Erlich et al, Science 252: 1643-51 (1991), which is hereby incorporated by reference in its entirety) or in restriction fragment length polymorphism (RFLP) diagnostic techniques, as described in Watson et al, (2d ed. 1992), Recombinant DNA, Scientific American Books, 519-522, 545-547, which is hereby incorporated by reference.
  • RFLP restriction fragment length polymorphism
  • Nucleic acids constructed in accordance with the present invention can be labeled to provide a signal as a means of detection.
  • radioactive elements such as 32 P, 3 H, and 35 S or the like provide sufficient half-life to be useful as radioactive labels.
  • Other materials useful for labeling synthetic nucleotides include fluorescent compounds, enzymes and chemiluminescent moieties. Methods useful in selecting appropriate labels and binding protocols for binding the labels to the synthetic nucleotides are well known to those of skill in the art. Standard immunology manuals such as Promega: Protocol and Applications Guide. 2nd Edition, 1991 (Promega Corp., Madison, WI, the content of which is incorporated herein in its entirety) may be consulted to select an appropriate labeling protocol without undue experimentation.
  • an isolated nucleic acid molecule of the present invention includes a nucleic acid that is at least about 75%, preferably at least about 80%, more preferably at least about 85%, even more preferably at least about 90%, still more preferably at least about 95%, and even more preferably at least about 99%, identical to a chicken-derived ovomucoid gene expression control region -comprising nucleic acid molecule as depicted in SEQ ID NO: 26 and directs expression of a polypeptide encoding sequence in an avian oviduct cell, when operably linked to the polypeptide encoding sequence.
  • an isolated nucleic acid molecule of the present invention includes a nucleic acid that hybridizes to SEQ ID NO: 26 or the reverse complement thereof, or the insert in pBS-OVMUP-10, under high, moderate or low stringency hybridization conditions.
  • an avian ovomucoid gene expression control region gene or nucleic acid molecule can be an allelic variant of SEQ ID NO: 26 or a homolog from a different avian, e.g., quail, duck, etc.
  • the present invention also contemplates the use of antisense nucleic acid molecules that are designed to be complementary to a coding strand of a nucleic acid (i.e., complementary to an mRNA sequence) or, alternatively, complimentary to a 5' or 3' untranslated region of the mRNA.
  • a nucleic acid i.e., complementary to an mRNA sequence
  • Another use of synthetic nucleotides is as primers (DNA or RNA) for a polymerase chain reaction (PCR), ligase chain reaction (LCR), or the like. Synthesized oligonucleotides can be produced in variable lengths. The number of bases synthesized will depend upon a variety of factors, including the desired use for the probes or primers.
  • sense or anti-sense nucleic acids or oligonucleotides can be chemically synthesized using modified nucleotides to increase the biological stability of the molecule or of the binding complex formed between the anti-sense and sense nucleic acids.
  • nucleic acid sequence of a chicken ovomucoid gene expression control region nucleic acid molecule (SEQ ID NO: 26) of the present invention allows one skilled in the art to, for example, (a) make copies of those nucleic acid molecules by procedures such as, but not limited to, insertion into a cell for replication by the cell, by chemical synthesis or by procedures such as PCR or LCR, (b) obtain nucleic acid molecules which include at least a portion of such nucleic acid molecules, including full-length genes, full-length coding regions, regulatory control sequences, truncated coding regions and the like, (c) obtain ovomucoid gene expression control region nucleic acid homologs in other avian species such as, but not limited to, turkey, duck, goose, quail, pheasant, parrot, finch, ratites including ostrich, emu and cassowary and, (d) to obtain isolated nucleic acids capable of hybridizing to an avian ovo
  • nucleic acid homologs can be obtained in a variety of ways including by screening appropriate expression libraries with antibodies of the present invention, using traditional cloning techniques to screen appropriate libraries, amplifying appropriate libraries or DNA using oligonucleotide primers of the present invention in a polymerase chain reaction or other amplification method, and screening public and/or private databases containing genetic sequences using nucleic acid molecules of the present invention to identify targets.
  • preferred libraries to screen, or from which to amplify nucleic acid molecules include but are not limited to mammalian BAC libraries, genomic DNA libraries, and cDNA libraries.
  • preferred sequence databases useful for screening to identify sequences in other species homologous to chicken ovomucoid gene expression control region include, but are not limited to, GenBank and the mammalian Gene Index database of The Institute of Genomics Research (TIGR).
  • Codon-optimized proteins Another aspect of the present invention is a recombinant DNA molecule comprising the novel isolated avian ovomucoid gene expression control region of the present invention operably linked to a selected polypeptide-encoding nucleic acid insert, and which may express the nucleic acid insert when transfected to a suitable host cell, preferably an avian cell.
  • the nucleic acid insert may be placed in frame with a signal peptide sequence, whereby translation initiation from the transcript may start with the signal peptide and continue through the nucleic acid insert, thereby producing an expressed polypeptide having the desired amino acid sequence.
  • the recombinant DNA may further comprise a polyadenylation signal sequence that will allow the transcript directed by the novel ovomucoid gene expression control region to proceed beyond the nucleic acid insert encoding a polypeptide and allow the transcript to further comprise a 3' untranslated region and a polyadenylated tail.
  • Any functional polyadenylation signal sequence may be linked to the 3' end of the nucleic acid insert including the S V40 polyadenylation signal sequence, bovine growth hormone adenylation sequence or the like, or derivatives thereof.
  • Another aspect of the present invention is to provide nucleic acid sequences of a protein optimized for expression in avian cells, and derivatives and fragments thereof.
  • One embodiment of the present invention is a recombinant DNA molecule comprising the isolated avian ovomucoid gene expression control region of the present invention, operably linked to a nucleic acid insert encoding a polypeptide, and a polyadenylation signal sequence optionally operably linked thereto. It is contemplated that when the recombinant DNA is to be delivered to a recipient cell for expression therein, the sequence of the nucleic acid sequence may be modified so that the codons are optimized for the codon usage of the recipient species. When a heterologous nucleic acid is to be delivered to a recipient cell for expression therein, the sequence of the nucleic acid sequence may be modified so that the codons are optimized for the codon usage of the recipient species.
  • the sequence of the expressed nucleic acid insert is optimized for chicken codon usage. This may be determined from the codon usage of at least one, and preferably more than one, protein expressed in a chicken cell.
  • the codon usage may be determined from the nucleic acid sequences encoding the proteins ovalbumin, lysozyme, ovomucin and ovotransferrin of chicken.
  • the DNA sequence for the target protein may be optimized using the BACKTRANSLATE® program of the Wisconsin Package, version 9.1 (Genetics Computer Group, Inc., Madison, WI) with a codon usage table compiled from the chicken (Gallus gallus) ovalbumin, lysozyme, ovomucoid, and ovotransferrin proteins.
  • the template and primer oligonucleotides are then amplified, by any means known in the art, including but not limited to PCR with Pfu polymerase (STRATAGENE®, La Jolla CA).
  • a nucleic acid insert encoding the human interferon ⁇ 2b polypeptide optimized for codon-usage by the chicken is used. Optimization of the sequence for codon usage is useful in elevating the level of translation in avian eggs.
  • the heterologous polypeptide is encoded using the codon-usage of a chicken.
  • the recombinant DNA comprises
  • the isolated avian ovomucoid gene expression control region operably linked to a nucleic acid encoding a human interferon ⁇ 2b and the SV40 polyadenylation sequence.
  • the protein of the present invention may be produced in purified form by any known conventional technique.
  • the protein is purified from chicken eggs, preferably egg whites.
  • chicken cells may be homogenized and
  • the fraction containing the protein of the present invention is subjected to gel filtration in an appropriately sized dextran or polyacrylamide column to separate the proteins. If necessary, the protein fraction may be further purified by HPLC.
  • the invention in preferred embodiments, provides methods for producing multimeric proteins, preferably immunoglobulins, such as antibodies, and antigen binding fragments thereof.
  • the multimeric protein is an
  • first and second heterologous polypeptides are an immunoglobulin heavy and light chains respectively.
  • Illustrative examples of this and other aspects and embodiments of the present invention for the production of heterologous multimeric polypeptides in avian cells are fully disclosed in U.S. Patent Application No. 09/877,374, filed June 8, 2001, by Rapp, published as US-2002-0108132-A1 on August 8,
  • the multimeric protein is an immunoglobulin wherein the first and second heterologous polypeptides are an immunoglobulin heavy and light chain respectively. Accordingly, the invention provides immunoglobulin and other multimeric proteins that have been produced by transgenic avians of the invention.
  • an immunoglobulin polypeptide encoded by the transcriptional unit of at least one expression vector may be an immunoglobulin heavy chain polypeptide comprising a variable region or a variant thereof, and may further comprise a D region, a J region, a C region, or a combination thereof.
  • An immunoglobulin polypeptide encoded by the transcriptional unit of an expression vector may also be an immunoglobulin light chain polypeptide comprising a variable region or a variant thereof, and may further comprise a J region and a C region.
  • the immunoglobulin regions prefferably be derived from the same animal species, or a mixture of species including, but not only, human, mouse, rat, rabbit and chicken.
  • the antibodies are human or humanized.
  • the immunoglobulin polypeptide encoded by the transcriptional unit of at least one expression vector comprises an immunoglobulin heavy chain variable region, an immunoglobulin light chain variable region, and a linker peptide thereby forming a single-chain antibody capable of selectively binding an antigen.
  • Another aspect of the present invention provides a method for the production in an avian of an heterologous protein capable of forming an antibody suitable for selectively binding an antigen comprising the step of producing a transgenic avian incorporating at least one transgene, wherein the transgene encodes at least one heterologous polypeptide selected from an immunoglobulin heavy chain variable region, an immunoglobulin heavy chain comprising a variable region and a constant region, an immunoglobulin light chain variable region, an immunoglobulin light chain comprising a variable region and a constant region, and a single-chain antibody comprising two peptide-linked immunoglobulin variable regions.
  • the isolated heterologous protein is an antibody capable of selectively binding to an antigen.
  • the antibody may be generated by combining at least one immunoglobulin heavy chain variable region and at least one immunoglobulin light chain variable region, preferably cross-linked by at least one di-sulfide bridge.
  • the combination of the two variable regions will generate a binding site capable of binding an antigen using methods for antibody reconstitution that are well known in the art.
  • immunoglobulin heavy and light chains, or variants or derivatives thereof to be expressed in separate transgenic avians, and therefore isolated from separate media including serum or eggs, each isolate comprising a single species of immunoglobulin polypeptide.
  • the method may further comprise the step of combining a plurality of isolated heterologous immunoglobulin polypeptides, thereby producing an antibody capable of selectively binding to an antigen.
  • two individual transgenic avians maybe generated wherein one transgenic produces serum or eggs having an immunoglobulin heavy chain variable region, or a polypeptide comprising such, expressed therein.
  • a second transgenic animal having a second transgene, produces serum or eggs having an immunoglobulin light chain variable region, or a polypeptide comprising such, expressed therein.
  • the polypeptides may be isolated from their respective sera and eggs and combined in vitro to generate a binding site capable of binding an antigen.
  • therapeutic antibodies examples include but are not limited to HERCEPTIN® (Trastuzumab) (Genentech, CA) which is a humanized anti-HER2 monoclonal antibody for the treatment of patients with metastatic breast cancer; REOPRO® (abciximab) (Centocor) which is an anti-glycoprotein flb/ ⁇ ia receptor on the platelets for the prevention of clot formation; ZENAPAX® (daclizumab) (Roche Pharmaceuticals, Switzerland) which is an immunosuppressive, humanized anti- CD25 monoclonal antibody for the prevention of acute renal allograft rejection; PANOREXTM which is a niurine anti-17-IA cell surface antigen IgG2a antibody (Glaxo Wellcome/Centocor); BEC2 which is a murine anti-idiotype (GD3 epitope) IgG antibody (ImClone System); 1MC-C225 which is a chimeric
  • Another potentially useful application of the novel isolated ovomucoid gene expression control region of the present invention is the possibility of increasing the amount of a heterologous protein present in a bird, (especially the chicken) by gene transfer.
  • a heterologous polypeptide-encoding nucleic acid insert transferred into the recipient animal host will be operably linked with the ovomucoid gene expression control region to allow the cell to initiate and continue production of the genetic product protein.
  • a recombinant DNA molecule of the present invention can be transferred into the extra-chromosomal or genomic DNA of the host.
  • the recombinant DNA nucleic acid molecules of the present invention can be delivered to cells using conventional recombinant DNA technology.
  • the recombinant DNA molecule may be inserted into a cell to which the polypeptide-encoding nucleic acid insert of the recombinant DNA molecule is heterologous (i.e. not normally present).
  • the recombinant DNA molecule may be introduced into cells which normally contain the polypeptide-encoding nucleic acid insert of the recombinant DNA molecule, for example, to correct a deficiency in the expression of a polypeptide, or where over-expression of the polypeptide is desired.
  • the heterologous DNA molecule is inserted into the expression system or vector of the present invention in proper sense orientation and correct reading frame.
  • the vector contains the necessary elements for the transcription and translation of the inserted protein-coding sequences, including the novel isolated ovomucoid gene expression control region.
  • One aspect of the present invention is an expression vector suitable for delivery to a recipient cell for replication OR expression of a polypeptide-encoding nucleic acid of the vector therein. It is contemplated to be within the scope of the present invention for the expression vector to comprise an isolated avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a polypeptide, and optionally a polyadenylation signal sequence.
  • the expression vector of the present invention may further comprise a bacterial plasmid sequence, a viral nucleic acid sequence, or fragments or variants thereof that may allow for replication of the vector in a suitable host.
  • novel isolated avian ovomucoid gene expression control region of the present invention (SEQ ID NO: 26) and a polypeptide-encoding nucleic acid sequence operably linked thereto and optionally a polyadenylation signal sequence may be introduced into viruses such as vaccinia virus.
  • viruses such as vaccinia virus.
  • Methods for making a viral recombinant vector useful for expressing a protein under the control of the ovomucoid promoter are analogous to the methods disclosed in U.S. Patent Nos. 4,603,112; 4,769,330; 5,174,993; 5,505,941; 5,338,683; 5,494,807; 4,722,848; Paoletti, E. Proc. Natl. Acad. Sci.
  • Recombinant viruses can also be generated by transfection of plasmids into cells infected with virus.
  • Suitable vectors include, but are not limited to, viral vectors such as lambda vector system ⁇ gtl 1, ⁇ gt WES.tB, Charon 4, and plasmid vectors such as pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pKC37, pKClOl, SV 40, pBluescript II SK +/- or KS +/- (see "Stratagene Cloning Systems” Catalog (1993) from Stratagene, La Jolla, Calif, which is hereby incorporated by reference), pQE, pIH821, pGEX, pET series (see Studier, F.
  • DNA sequences are cloned into the vector using standard cloning procedures in the art, as described by Sambrook et al. Molecular Cloning: A Laboratory Manual, 3 rd ed., Cold Spring Harbor Laboratory, Cold Springs Harbor, N.Y. (2001), which is hereby incorporated by reference in its entirety.
  • the vectors of the invention comprise one or more nucleotide sequences encoding a heterologous protein desired to be expressed in the transgenic avian, as well as regulatory elements such as promoters, enhancers, Matrix Attachment Regions, IRES's and other translation control elements, transcriptional termination elements, polyadenylation sequences, etc.
  • the vector of the invention contains at least two nucleotide sequences coding for heterologous proteins, for example, but not limited to, the heavy and light chains of an immunoglobulin.
  • the present invention further relates to nucleic acid vectors and transgenes inserted therein, having the avian ovomucoid gene expression control region of the invention, that incorporate multiple polypeptide-encoding regions, wherein a first polypeptide-encoding region is operatively linked to a transcription promoter and a second polypeptide-encoding region is operatively linked to an IRES.
  • the vector may contain coding sequences for two different heterologous proteins (e.g., the heavy and light chains of an immunoglobulin).
  • nucleic acid constructs when inserted into the genome of a bird and expressed therein, will generate individual polypeptides that may be post-translationally modified, for example, glycosylated or, in certain embodiments, form complexes, such as heterodimers with each other in the white of the avian egg.
  • the expressed polypeptides may be isolated from an avian egg and combined in vitro, or expressed in a non-reproductive tissue such as serum.
  • two separate constructs each containing a coding sequence for one of the heterologous proteins operably linked to the ovomucoid gene expression control region of the invention are introduced into the avian cell.
  • two transgenic avians each containing one of the two heterologous proteins e.g., one transgenic avian having a transgene encoding the light chain of an antibody and a second transgenic avian having a transgene encoding the heavy chain of the antibody
  • the ovomucoid gene expression control region of the present invention Once the ovomucoid gene expression control region of the present invention has been cloned into a vector system, it is ready to be incorporated into a host cell. Such incorporation can be carried out by the various forms of transformation noted above, depending upon the vector/host cell system. Suitable host cells include, but are not limited to, bacteria, virus, yeast, mammalian or avian cells, and the like. Alternatively, it is contemplated that the incorporation of the DNA of the present invention into a recipient cell may be by any suitable method such as, but not limited to, viral transfer, electroporation, gene gun insertion, sperm mediated transfer to an ovum, microinjection, cytoplasmic injection, pronuclear injection and the like.
  • Another aspect of the present invention is a method of expressing a heterologous polypeptide in a eukaryotic cell by transfecting the cell with a recombinant DNA comprising an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a polypeptide and, optionally, a polyadenylation signal sequence, and culturing the transfected cell in a medium suitable for expression of the heterologous polypeptide under the control of the avian ovomucoid gene expression control region.
  • the ovomucoid gene expression control region directs a level of expression of the heterologous protein in avian eggs that is greater than 5 ⁇ g, 10 ⁇ g, 50 ⁇ g, 100 ⁇ g, 250 ⁇ g, 500 ⁇ g, or 750 ⁇ g, more preferably greater than 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, 50 mg, 100 mg, 200 mg, 500 mg, 700 mg, 1 gram, 2 grams, 3 grams, 4 grams or 5 grams per egg.
  • levels of expression can be obtained using the expression control regions of the invention.
  • the recipient eukaryotic cell is derived from an avian.
  • the avian is a chicken.
  • Yet another aspect of the present invention is a eukaryotic cell transformed with an expression vector according to the present invention and described above.
  • the transformed cell is a chicken oviduct cell and the nucleic acid insert comprises the chicken ovomucoid gene expression control region, a nucleic acid insert encoding a human interferon ⁇ 2d with codons optimized for expression in an avian cell, and an SV40 polyadenylation sequence.
  • the transfected cell according to the present invention may be transiently transfected, whereby the transfected recombinant DNA or expression vector may not be integrated into the genomic nucleic acid. It is further contemplated that the transfected recombinant DNA or expression vector may be stably integrated into the genomic DNA of the recipient cell, thereby replicating with the cell so that each daughter cell receives a copy of the transfected nucleic acid. It is still further contemplated for the scope of the present invention to include a transgenic animal producing a heterologous protein expressed from a transfected nucleic acid according to the present invention.
  • the transgenic animal is an avian selected from a turkey, duck, goose, quail, pheasant, ratite, an ornamental bird or a feral bird.
  • the avian is a chicken and the heterologous protein produced under the transcriptional control of the isolated avian ovomucoid gene expression control region according to the present invention is produced in the white of an egg.
  • Viral vector cell transformation An exemplary approach for the in vivo introduction of a polypeptide-encoding nucleic acid operably linked to the subject novel isolated ovomucoid gene expression control region into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA, encoding the gene product.
  • a viral vector containing nucleic acid e.g. a cDNA
  • Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid.
  • molecules encoded within the viral vector e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells that have taken up viral vector nucleic acid.
  • Retrovirus vectors and adeno-associated virus vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • Recombinant retrovirus can be constructed in the part of the retro viral coding sequence (gag, pol, env) that has been replaced by nucleic acid comprising a ovomucoid gene expression control region, thereby rendering the retrovirus replication defective.
  • Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel et al. (1989) (eds.) Greene Publishing Associates, Sections 9.10-9.14 and other standard laboratory manuals.
  • retroviruses examples include pLJ, pZIP, pWE and pEM which are all well known to those skilled in the art.
  • suitable packaging virus lines for preparing both ecotropic and amphotropic retroviral systems include psiCrip, psiCre, psi2 and psiAm.
  • retroviral-based vectors it is possible to limit the infection spectrum of retroviruses and consequently of retroviral-based vectors, by modifying the viral packaging proteins on the surface of the viral particle (see, for example PCT publications WO93/25234, WO94/06920, and WO94/11524).
  • strategies for the modification of the infection spectrum of retroviral vectors include coupling antibodies specific for cell surface antigens to the viral env protein (Roux et al., Proc. Natl. Acad. Sci. 86: 9079-9083 (1989); Julan et al, J. Gen. Virol.
  • Coupling can be in the form of the chemical cross-linking with a protein or other moiety (e.g. lactose to convert the env protein to an asialoglycoprotein), as well as by generating fusion proteins (e.g. single-chain antibody/env fusion proteins).
  • a protein or other moiety e.g. lactose to convert the env protein to an asialoglycoprotein
  • fusion proteins e.g. single-chain antibody/env fusion proteins
  • Another viral gene delivery system useful in the present invention utilizes adenovirus-derived vectors.
  • the genome of an adenovirus can be manipulated such that it encodes a gene product of interest, but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle (see, for example, Berkner et al, BioTechniques 6: 616 (1988); Rosenfeld et al, Science 252: 43 1434 (1991); and Rosenfeld et al, Cell 68: 143-155 (1992)), all of which are incorporated herein by reference in their entireties.
  • Suitable adeno viral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus are well known to those skilled in the art.
  • the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity.
  • introduced adeno viral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • adenoviral vectors currently in use and therefore favored by the present invention are deleted for all or parts of the viral El and E3 genes but retain as much as 80% of the adenoviral genetic material (see, e.g., Jones et al, Cell 16:683 (1979); Berkner et al, supra; and Graham et al, in Methods in Molecular Biology, E. J. Murray, (1991) Ed. (Humana, Clifton, N.J.) vol. 7. pp. 109-127), all of which are incorporated herein by reference in their entireties.
  • Expression of an inserted gene such as, for example, encoding the human interferon ⁇ 2b, can be under control of the exogenously added ovomucoid gene expression control region sequences.
  • Yet another viral vector system useful for delivery of, for example, the subject avian ovomucoid gene expression control region operably linked to a nucleic acid encoding a polypeptide is the adeno-associated virus (AAN).
  • AAN adeno-associated virus
  • Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate. Space for exogenous D ⁇ A is limited to
  • An AAV vector such as that described in Tratschin et al, Mol. Cell. Biol. 5:3251-3260 (1985) can be used to introduce D ⁇ A into cells.
  • a variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et al., Proc. ⁇ atl. Acad. Sci. 81:6466-6470 (1984); Tratschin et al, Mol. Cell. Biol. 4:2072-2081 (1985); Wondisford et al, Mol. Endocrinol. 2:32-39 (1988); Tratschin et al, J. Virol.
  • non- viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject ovomucoid gene expression control region and operably linked polypeptide-encoding nucleic acid by the targeted cell.
  • Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and 0 artificial viral envelopes.
  • a nucleic acid comprising the novel isolated ovomucoid gene expression control region of the present invention can be entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et ⁇ l., NO 5 Shinkei Geka 20:547-551 (1992); PCT publication WO91/06309; Japanese patent application 1047381 ; and European patent publication EP-A-43075), all of which are incorporated herein by reference in their entireties.
  • lipofectins e.g., lipofectins
  • the gene delivery system comprises an antibody or cell surface ligand that is cross-linked with a gene binding agent such as polylysine (see, for example, 0 PCT publications WO93/04701, WO92/22635, WO92/20316, WO92/19749, and
  • WO92/06180 all of which are incorporated herein by reference in their entireties. It will also be appreciated that effective delivery of the subject nucleic acid constructs via receptor-mediated endocytosis can be improved using agents which enhance escape of gene from the endosomal structures. For instance, whole adenovirus or fusogenic peptides of the 5 influenza HA gene product can be used as part of the delivery system to induce efficient disruption of DNA-containing endosomes (Mulligan et al, Science 260: 926 (1993); Wagner et al, Proc. Natl. Acad. Sci. 89:7934 (1992); and Christiano et al, Proc. Natl. Acad. Sci.
  • a recombinant DNA molecule comprising the novel isolated ovomucoid gene expression control region of the present invention may be delivered to a recipient host cell by other non- viral methods including by gene gun, microinjection, sperm-mediated transfer as described in PCT/US02/30156, filed September 23, 2002 and incorporated herein by reference in its entirely, nuclear transfer, or the like.
  • transgenic animals such as chickens that contain a transgene comprising the novel isolated ovomucoid gene expression control region of the present invention and which preferably (though optionally) express a heterologous gene in one or more cells in the animal.
  • Suitable methods for the generation of transgenic avians having heterologous DNA incorporated therein, for example, cytoplasmic injection and pronuclear injection, are described, for example, in U.S. Patent Application No: 10/251,364, entitled "Methods of Generating Transgenic Avians Using Microinjection into the Cytoplasm of an Avian Egg or Embryo", by Rapp et al., filed September 18, 2002, and incorporated herein by reference in its entirety.
  • the expression of the transgene may be restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences acting on the ovomucoid gene expression control region of the present invention and which control gene expression in the desired pattern.
  • Tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial patterns.
  • temporal patterns of expression can be provided by, for example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
  • transgenic avian having a heterologous polynucleotide sequence comprising a nucleic acid insert encoding the heterologous polypeptide and operably linked to the novel isolated avian ovomucoid gene expression control region.
  • the transgenic avian is selected from a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird.
  • the transgenic avian is a chicken.
  • the transgenic avian includes an avian ovomucoid gene expression control region comprising the nucleic acid sequence in SEQ ID NO: 26, or a degenerate variant thereof.
  • the transgenic avian further comprises a polyadenylation signal sequence.
  • the polyadenylation signal sequence is derived from the SV40 virus.
  • the nucleic acid insert encoding a polypeptide has a codon complement optimized for protein expression in an avian.
  • the transgenic avian produces the heterologous polypeptide in the serum or an egg white. In another embodiment of the transgenic avian of the present invention, the transgenic avian produces the heterologous polypeptide in an egg white.
  • Example 1 PCR amplification of Ovomucoid promoter
  • Sense primer OVINs2, 5'-TAGGCAGAGCAATAGGACTCTCAACCTCGT-3' (SEQ ID NO: 1) and the antisense primer, OVMUa2, 5'-AAGCTTCTGCAGCACTCTGGGAGTTACTCA-3' (SEQ ID NO: 2) were designed according to the sequences of chick ovoinhibitor exon 16 (Genbank Accession No: M16141) and a fragment of the chick ovomucoid promoter region (Genbank Accession No: J00897) respectively.
  • the template DNA for PCR amplification of the ovomucoid promoter region was prepared from white leghorn chick blood.
  • FIG. 2 A series of different PCR conditions were carried out to optimize synthesis of the approximately 10.0 kb product, the results of which are shown in FIG. 2.
  • the template DNA concentrations were 500 ng, 100 ng, 50 ng, or 10 ng.
  • the results of the tests were as shown in FIG. 2.
  • test reactions having 500 ng DNA template, the OVI sl (SEQ ID NO: 3) and OVMUal (SEQ ID NO: 4) primers, 60 mM Tris-SO 4 , pH9.1, 18 mM (NH 4 ) 2 SO 4 , 1.0 mM Mg 2* , and annealing temperatures between 50°C to 58°C gave no specific DNA product. Also, as shown in lanes 17 through 24 of FIG.
  • reaction conditions containing 100 ng DNA template, the OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) primers, 60 mM Tris-SO 4 , pH9.1, 18 mM (NH 4 ) 2 SO 4 , 2mM Mg 2+ and annealing temperatures between about 60 °C to about 68 °C gave an increased yield of the desired product.
  • the optimum DNA template concentration was between about 50 ng to 500 ng; the primers were OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2); the Mg 2+ concentration was 2 mM; the annealing temperature was at or between about 60 °C to about 68°C.
  • Each 50 ⁇ l PCR reaction consisted of 50 ng or 100 ng of template DNA, 0.1 ⁇ g each primer, 5 ⁇ l buffer B (from Elongase Enzyme Mix kit, Invitrogen Corp., Carlsbad, CA), 1 ml of 10 ⁇ M dNTP solution, and distilled deionized water.
  • the PCR protocol was one cycle at 94°C for 30 sees; thirty cycles at 94°C for 30 sees, 60 °C for 30 sees and 68 °C for 10 mins. One cycle was performed at 68 °C for 10 mins, 35 °C for 30 mins with a final hold at 4°C.
  • the PCR products were examined by 0.65% agarose gel analysis.
  • PCR products were purified by standard methods. Briefly, PCI (phenol: chloroform: isoamyl alcohol, 24:25:1) and chloroform extraction were performed once. The DNA was precipitated by adding 3M sodium acetate pH5.2 to a final concentration of 0.3M together with 2.5 volumes of 100% ethanol. The DNA pellet was dried and dissolved in distilled deionized water and then sequenced on a ABI3700 automatic sequencer (Applied Biosystems, Foster City, CA) using the primers OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) to confirm the identity of each PCR product.
  • PCI phenol: chloroform: isoamyl alcohol, 24:25:1
  • chloroform extraction were performed once.
  • the DNA was precipitated by adding 3M sodium acetate pH5.2 to a final concentration of 0.3M together with 2.5 volumes of 100% ethanol.
  • the DNA pellet was dried and dissolved in distilled deionized water and then sequenced on a ABI3700 automatic
  • the approximately 10 kb PCR product was treated with T4 polynucleotide kinase to add a phosphate to the 5' end. Mung bean nuclease removed any overhanging adenines from the ends of the PCR products, thereby producing a blunt end.
  • the PCR product was purified by PCI and chloroform extraction and precipitated by standard methods. This 10 kb product was then cleaved with Bam HI to give two fragments, of about 4.7 and about 5.5 kb respectively.
  • the vector plasmid pBluescript II KS (+/-) was cut by Bam HI and Eco RV and treated with calf intestinal alkaline phosphatase. DNA fragments to be ligated into the vector were analyzed by agarose gel electrophoresis and purified from agarose gel slices using a NucleoTrap Nucleic Acid Purification Kit (BD Biosciences Clontech, Palo Alto, CA). Fragments of 4.7 kb and 5.5 kb were inserted into the Bam HVEco RV-treated pBluescript to give the constructs pBS-OVMUP4.7 and pBS-OVMUP5.5 respectively.
  • Positive clones were screened by Xba VXho I digestion.
  • Clone pBS-OVMUP4.7 gave fragments of about 4.7 kb and 2.96 kb.
  • Clone pBS-OVMUP5.5 gave fragments of about 5.5 kb and 2.96 kb.
  • Apparent positive clones having the 4.7 kb insert were further confirmed by Xba VHind HI digestion that gave three fragments of 0.5 kb, 4.2 kb and 2.9 kb.
  • the apparent positive clones with an insert of about 5.5 kb insert were further confirmed by Xba VKpn I digestion that gave three fragments of 2 kb, 3.5 kb and 2.96 kb.
  • a construct, pBS-OVMUP-10, containing the entire 10 kb PCR product cloned into the pBluescript KS II (+/-) vector was made by taking a 4.7 kb Bam Hl/Xho I fragment from the pBS-OVMUP4.7 plasmid and inserting it into the Bam ⁇ UXba I cleaved sites of pBS-OVMUP5.5.
  • the Xho I and Xba I cut ends were blunt-ended by treating the digested fragments with Klenow enzyme and dNTPs at 25 °C for 15 mins before the digestion with Bam HI.
  • the plasmids pBS-OVMUP4.7 and pBS-OVMUP5.5 were sequenced from both ends of each insert as shown in FIG. 1.
  • the initial primers were T7 and T3 having the nucleic acid sequences 5'-TAATACGACTCACTATAGGG-3' (SEQ ID NO: 5) and
  • sequence of the parent fragment and no intervening Bam HI - Bam HI fragments were included in the final sequence SEQ ID NO: 26.
  • the sequence (SEQ DD NO: 26) of the region lying between the 3' end of the ovoinhibitor gene and the transcription start site of the ovomucoid-encoding region is shown in FIG. 4.
  • the Nco I site which overlaps 20 the second ATG was changed to a Pci I site as depicted below.
  • the wild type ovomucoid sequence at the start site of translation On the bottom, the second Nco I site was changed to a Pci I site.
  • the Pci I site in the Bluescript backbone of pBS-OVMUP-10 was destroyed by cutting with Pci I, filling in the ends with Klenow polymerase and religating, creating pOM- 35 10-alpha.
  • the proximal promoter region was PCR amplified with primers OM-5 (SEQ ID NO.:29) and OM-6 (SEQ ID NO.:30) and template pBS-ONMUP-10.
  • the resulting PCR product (SEQ ID ⁇ O.:31) was cut with Not I and TthU 1 1 and cloned into the 12059 bp Not
  • the 1 st and 2 nd ATGs of the ovomucoid sequence are shown underlined. Note that the ovomucoid coding sequence is in reverse. The underlined, bold A is not in the wildtype sequence but was incorporated into pOM-10-Pci due to a error in the oligo OM-5.
  • HDl 1 cells For expression in avian cells of non-magnum origin, HDl 1 cells, a chicken myeloid cell line was used. Cells were cultured as described in Beug, H., et al. (Chicken hematopoietic cells transformed by seven strains of defective avian leukemia viruses display three distinct phenotypes of differentiation. (1979) Cell, 18: 375-90, in which these cells were referred to as HBCI cells), herein incorporated by reference in its entirety. Plasmid DNA was transfected into HDl 1 cells with Lipofectamine 2000 (Invitrogen Corporation, Carlsbad, California) according to the manufacturer's instructions.
  • Lipofectamine 2000 Invitrogen Corporation, Carlsbad, California
  • HDl 1 cells are permissive for the CMN promoter and should be able to only weakly activate the ovomucoid promoter. Some expression of the luciferase gene linked to the 10 kb ovomucoid is evident.
  • avian oviduct cells For expression in avian oviduct cells, primary tubular gland cells were isolated as follows. The oviduct of a Japanese quail (Coturnix coturnixjaponica) was removed and the magnum portion minced and enzymatically dissociated with 0.8 mg/ml collagenase (Sigma Chemical Co., St. Louis, MO) and 1.0 mg/ml dispase (Roche Molecular Biochemicals, Indianapolis, IN) by shaking and titurating for 30 minutes at 37 ° C.
  • collagenase Sigma Chemical Co., St. Louis, MO
  • dispase Roche Molecular Biochemicals, Indianapolis, IN
  • the cell suspension was then filtered through sterile surgical gauze, washed three times with F-12 medium (Life Technologies, Grand Island, NY) by centrifugation at 200 x g, and resuspended in OPTEV ⁇ EMTM (Life Technologies) such that the OD 600 was approximately 2. 800 ⁇ l of the cell suspension was plated in each well of a 6-well dish. For each transfection, 4.0 ⁇ l of F-12 medium (Life Technologies, Grand Island, NY) by centrifugation at 200 x g, and resuspended in OPTEV ⁇ EMTM (Life Technologies) such that the OD 600 was approximately 2. 800 ⁇ l of the cell suspension was plated in each well of a 6-well dish. For each transfection, 4.0 ⁇ l of
  • the cells were scraped into the media with a rubber policeman. One milliliter was transferred to an eppendorf tube and the cells pelleted. The supernatant was removed and 20 ⁇ l of 10 mM Tris, ph 7.8, 1 mM EDTA (TE) was added. The cells were frozen at -80°C and thawed. 5 ⁇ l of the cell suspension was mixed with 25 ⁇ l of Bright- GloTM reagent (Bright-GloTM Luciferase Assay System, Promega, Madison, WI) and relative light units per second measured on a Berthold Detection Systems (Oak Ridge, TN) FBI 2 luminometer.
  • Bright- GloTM reagent Bright-GloTM Luciferase Assay System, Promega, Madison, WI
  • FIG. 6B Expression of luciferase is evident from the CMV and 10 kb ovomucoid promoters.
  • the ovomucoid promoter has more activity relative to the CMV promoter in the tubular gland cells (ratio of CMV to ovomucoid is 152) than in the HDl 1 cells (ratio of CMV to ovomucoid is 2221).
  • the CMV promoter region of pAVIJCR-A137.91.1.2 flanked by Nco I sites was replaced with the 1051 bp Nco ⁇ -Nco I fragment from pBS-OVMUP-4.4, thereby inserting the 1 kb ovomucoid promoter in front of the IFN coding sequence and SV40 polyadenylation signal and creating plkb-OM-IFNMM.
  • a 1816 bp Cla I- Sac I fragment of plkb-OM-IFNMM was inserted into the 6245 bp Cla l-Sac I fragment of pBS-OVMUP-4.4, thereby fusing the 4.4 kb ovomucoid fragment with the IFN coding sequence and creating p4.4OM-IFNMM.
  • the 8511 bp BamH I-Sal I fragment of pBS- OVMUP-10 was ligated to the 5148 bp BamH l-Sal I fragment of p4.4OM-IFN, thereby placing the 10 kb ovomucoid promoter in front of the IFN coding sequence, creating plO- OM-EFN.

Abstract

The present invention provides novel isolated nucleic acids that comprise an avian nucleic acid sequence comprising a ovomucoid gene expression control region. The ovomucoid promoter region of the present invention will allow expression of an operably linked heterologous nucleic acid insert in a transfected avian cell such as, for example, an oviduct cell. The isolated avian ovomucoid of the present invention may be operably linked with a selected nucleic acid insert, wherein the nucleic acid insert encodes a polypeptide desired to be expressed in a transfected cell. The recombinant DNA of the present invention may further comprise a polyadenylation signal sequence. The present invention further includes expression vectors comprising an isolated avian ovomucoid gene expression control region of the present invention, and transfected cells and transgenic avians comprising the expression vectors.

Description

Title of the Invention
OVOMUCOID PROMOTER AND METHODS OF USE
This application is a continuation-in-part of application Serial No. 09/998,716 filed
November 30, 2001, which is hereby incorporated by reference in its entirety.
1. Field of the Invention
The present invention relates generally to the identification of an avian ovomucoid gene expression control region, particularly from the chicken. More specifically, the invention relates to recombinant nucleic acids and expression vectors, transfected cells and transgenic animals, especially chickens, that comprise the avian ovomucoid gene expression control region operably linked to a heterologous polypeptide-encoding nucleic acid.
2. Background
The field of transgenics was initially developed to understand the action of a single gene in the context of the whole animal and the phenomena of gene activation, expression, and interaction. This technology has also been used to produce models for various diseases in humans and other animals and is amongst the most powerful tools available for the study of genetics, and the understanding of genetic mechanisms and function. From an economic perspective, the use of transgenic technology for the production of specific proteins or other substances of pharmaceutical interest (Gordon et al, Biotechnology 5: 1183-1187 (1987); Wilmut et al, Theriogenology 33: 113-123 (1990)) offers significant advantages over more conventional methods of protein production by gene expression. Heterologous nucleic acids have been engineered so that an expressed protein may be joined to a protein or peptide that will allow secretion of the transgenic expression product into milk or urine, from which the protein may then be recovered. These procedures have had limited success and may require lactating animals, with the attendant costs of maintaining individual animals or herds of large species, including cows, sheep, or goats. Historically, transgenic animals have been produced almost exclusively by microinjection of the fertilized egg. The pronuclei of fertilized eggs are microinjected in vitro with foreign, i.e., xenogeneic or allogeneic, heterologous DNA or hybrid DNA molecules. The microinjected fertilized eggs are then transferred to the genital tract of a pseudopregnant female (See e.g., Krimpenfort et al, in U.S. Pat. No. 5,175,384). One system that holds potential is the avian reproductive system. The production of an avian egg begins with formation of a large yolk in the ovary of the hen. The unfertilized oocyte or ovum is positioned on top of the yolk sac. After ovulation, the ovum passes into the infundibulum of the oviduct where it is fertilized, if sperm are present, and then moves into the magnum of the oviduct which is lined with tubular gland cells. These cells secrete the egg-white proteins, including ovalbumin, ovomucoid, ovoinhibitor, conalbumin, ovomucin and lysozyme, into the lumen of the magnum where they are deposited onto the avian embryo and yolk. The hen oviduct offers outstanding potential as a protein bioreactor because of the high levels of protein production, the promise of proper folding and post-translation modification of the target protein, the ease of product recovery, and the shorter developmental period of chickens compared to other potential animal species. As a result, efforts have been made to create transgenic chickens expressing heterologous proteins in the oviduct. Chicken oviduct cells, when stimulated by steroid hormones during egg-laying, secrete three principal polypeptides, ovalbumin, ovomucoid and lysozyme (Tsai et al, Biochemistry 17: 5773-5779 (1978)). The mRNA transcript encoding ovalbumin constitutes about 50% of the total mRNA of these cells. Ovomucoid and lysozyme mRNAs contribute about 6.6% and 3.4% respectively of the total mRNA of the steroid stimulated cells. (Hynes et al, Cell 11:923 932 (1977)).
Detailed restriction enzyme analysis of fragments of chicken genomic DNA have shown that the ovomucoid-encoding sequence includes seven intronic sequences (Lindenmaier et al, Nuc. Acid Res. 7;1221-1232 (1979); Catterall et al, Nature 278: 323-327 (1979); Lai et al, Cell 18:829-842 (1979)). Short stretches of the 5' flanking region of the ovomucoid gene have been sequenced (Lai et al, Cell 18: 829-842 (1979); Genbank Accession No. J00897), but extending only 579 bases upstream of the recognized transcription start site. The 5' flanking region of the ovomucoid gene has been isolated (Catterall et al, Nature 278: 323-327 (1979); Lai et al, Cell 18: 829-842 (1979)), but not generally characterized beyond low-resolution restriction site mapping. Scott et al. in Biochemistry 26: 6831-6840 (1987) identified a CRl-like region within the 10 kb chicken genomic DNA located between the ovoinhibitor-encoding region and the downstream ovomucoid gene. The ovoinhibitor-encoding cDNA and the attached 3 '-untranslated region, which extends into the 10 kb ovoinhibitor-ovomucoid region, were also sequenced (Scott et al. J. Biol. Chem. 262: 5899-5907 (1987)). The chicken ovomucoid gene, therefore, is highly expressed in the tubular glands of the mature hen oviduct and represents a suitable candidate for an efficient promoter for heterologous protein production in transgenic animals, especially animals. The regulatory region of the ovomucoid locus extends over a nucleic acid region of about 10 kb of DNA 5' upstream of the transcription start site, and comprises at least one recognized element, the CR1.
3. Summary of the Invention The present invention relates to nucleic acids comprising an avian ovomucoid gene expression control region, which is useful for the expression of nucleotide sequences encoding a polypeptide of interest in a transfected avian cell such as, for example, an oviduct cell. In a preferred embodiment the polypeptide is heterologous, i.e., not the ovomucoid protein product, and more preferably, is a mammalian, most preferably, a human polypeptide. One aspect of the present invention provides a nucleic acid isolated from a region immediately 5' upstream of a transcription start site of the chicken (or other avian) ovomucoid gene locus. The nucleic acid comprises an avian nucleic acid sequence comprising an ovomucoid gene expression control region comprising at least one avian CR1 repeat element, and a proximal ovomucoid promoter. Interspersed between these constituent elements may be stretches of nucleic acid that may serve at least to organize the gene regulatory elements in an ordered array relative to a polypeptide-encoding region. In one embodiment of the present invention the ovomucoid gene expression control region is isolated from a chicken. In a specific embodiment, the ovomucoid gene expression control region has a nucleotide sequence of SEQ ID NO:26. In other embodiments, the ovomucoid gene expression control region is at least 75%, at least 95 %, or at least 99% identical to SEQ ID NO:26 and directs expression of a polypeptide encoding nucleotide sequence in an avian oviduct cell.
The avian ovomucoid gene expression control region of the present invention is useful for directing tissue-specific expression of a polypeptide-encoding nucleic acid. The avian ovomucoid gene expression control region may be operably linked with a selected nucleic acid insert, wherein the nucleic acid insert encodes a polypeptide, preferably heterologous, desired to be expressed in a transfected cell. The nucleic acid insert may be placed in frame with a nucleotide sequence encoding a signal peptide. Translation initiation may start with the signal peptide and continue through the nucleic acid insert, thereby producing an expressed polypeptide having the desired amino acid sequence. The recombinant DNA of the present invention may further comprise a polyadenylation signal sequence that will allow the transcript directed by the ovomucoid gene expression control region of the invention to proceed beyond the nucleic acid insert encoding a heterologous polypeptide (i.e., not the ovomucoid protein that is expressed from the endogenous gene containing the ovomucoid gene expression control region) and allow the transcript to further comprise a 3 ' untranslated region and a polyadenylated tail. Any functional polyadenylation signal sequence may be linked to the 3' end of the nucleic acid insert including the SV40 polyadenylation signal sequence, bovine growth hormone adenylation sequence or the like. Optionally, the nucleic acid of the invention may also comprise gene expression control elements, e.g. promoters, enhancers, IRES's, from other than an ovomucoid gene and may even be from a non-avian gene.
The sequence of the expressed nucleic acid insert may be optimized for codon usage by a host cell. This may be determined from the codon usage of at least one, and preferably more than one, protein expressed in a chicken cell. For example, the codon usage may be determined from the nucleic acid sequences encoding the proteins ovalbumin, ovomucoid, ovomucin and ovotransferrin of chicken.
Yet another aspect of the present invention is expression vectors suitable for delivery to a recipient cell for expression of heterologous protein coding sequences in the vector therein. The expression vector of the present invention may comprise an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a non- ovomucoid polypeptide, and optionally, a polyadenylation signal sequence. The expression vector may further comprise a bacterial plasmid sequence, a viral nucleic acid sequence, or fragments or variants thereof that may allow for replication of the vector in a suitable host. As also contemplated in the present invention the nucleic acid is a YAC, BAC, bacteriophage-derived artificial chromosome (BBPAC), cosmid or PI derived artificial chromosome (PAC).
The present invention further relates to nucleic acid vectors and transgenes inserted therein that incorporate multiple polypeptide-encoding regions, wherein a first polypeptide- encoding region is operatively linked to a transcription promoter and a second polypeptide- encoding region is operatively linked to an Internal Ribosome Entry Sequence (IRES). For example, the vector may contain coding sequences for two different heterologous proteins (e.g., the heavy and light chains of an immunoglobulin).
Such nucleic acid constructs, when inserted into the genome of a bird and expressed therein, will generate individual polypeptides that may be post-translationally modified, for example, glycosylated or, in certain embodiments, be present as complexes, such as heterodimers with each other.
Another aspect of the present invention is a method of expressing a heterologous polypeptide in a eukaryotic cell by transfecting the cell with a recombinant DNA comprising an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding the heterologous polypeptide and, optionally, a polyadenylation signal sequence, and culturing the transfected cell in a medium suitable for expression of the heterologous polypeptide under the control of the avian ovomucoid gene expression control region. In preferred embodiments the polypeptide is a cytokine, growth factor, enzyme, structural protein, and more preferably, an immunoglobulin, or subunit thereof. Other preferred embodiments the polypeptide is a mammalian, preferably a human polypeptide or derived from a human or mammalian polypeptide.
Also within the scope of the present invention are recombinant cells, tissues and animals, in particular avians and avian eggs, most preferably chicken, containing recombinant nucleic acid molecules according to the present invention and described above. In preferred embodiments, the level of expression of the heterologous protein is greater than 5 μg, 10 μg, 50 μg, 100 μg, 250 μg, 500 μg, or 750 μg, more preferably greater than 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, 50 mg, 100 mg, 200 mg, 500 mg, 700 mg, 1 gram, 2 grams, 3 grams, 4 grams or 5 grams in an egg (preferably the egg white) produced by the transgenic avian of the invention. In one embodiment of the present invention, the transformed cell is a chicken oviduct cell and the nucleic acid comprises the chicken ovomucoid gene expression control region, a nucleic acid insert encoding a heterologous polypeptide of interest, e.g. human interferon α2, which optionally is codon optimized for expression in an avian cell, and an SV40 polyadenylation sequence.
3.1 Definitions
The term "animal" is used herein to include all vertebrate animals, including humans. It also includes an individual animal in all stages of development, including embryonic and fetal stages.
The term "avian" as used herein refers to any species, subspecies or race of organism of the taxonomic class ava, such as, but not limited to, such organisms as chicken, turkey, duck, goose, quail, pheasants, parrots, finches, hawks, crows and ratites including ostrich, emu and cassowary. The term includes the various known strains of Gallus gallus, or chickens, (for example, White Leghorn, Brown Leghorn, Barred-Rock, Sussex, New Hampshire, Rhode Island, Ausstralorp, Minorca, Amrox, California Gray, Italian Partidge-colored), as well as strains of turkeys, pheasants, quails, duck, ostriches and other poultry commonly bred in commercial quantities. The term "nucleic acid" as used herein refers to any natural and synthetic linear and sequential arrays of nucleotides and nucleosides, for example cDNA, genomic DNA, mRNA, tRNA, oligonucleotides, oligonucleosides and derivatives thereof. Representative examples of the nucleic acids of the present invention include bacterial plasmid vectors including expression, cloning, cosmid and transformation vectors such as, but not limited to, pBR322, animal viral vectors such as, but not limited to, modified adenovirus, influenza virus, polio virus, pox virus, retrovirus, and the like, vectors derived from bacteriophage nucleic acid, e.g., plasmids and cosmids, artificial chromosomes, such as but not limited to, Yeast Artificial Chromosomes (YACs) and Bacterial Artificial Chromosomes (BACs), and synthetic oligonucleotides like chemically synthesized DNA or RNA. The term "nucleic acid" further includes modified or derivatised nucleotides and nucleosides such as, but not limited to, halogenated nucleotides such as, but not only, 5-bromouracil, and derivatised nucleotides such as biotin-labeled nucleotides.
The term "isolated nucleic acid" as used herein refers to a nucleic acid that has been removed from other components of the cell containing the nucleic acid or from other components of chemical/synthetic reaction used to generate the nucleic acid. In specific embodiments, the nucleic acid is 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100% pure. The "isolated nucleic acid" does not include nucleic acids that are members of a library, e.g. cDNA or genomic library, unless identified and separated from the other members of the library. The techniques used to isolate and characterize the nucleic acids and proteins of the present invention are well known to those of skill in the art and standard molecular biology and biochemical manuals may be consulted to select suitable protocols without undue experimentation. See, for example, Sambrook et al, 2001, Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Press; the content of which is herein incorporated by reference in its entirety. By the use of the term "enriched" in reference to nucleic acid it is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction of the total DNA or RNA present in the cells or solution of interest than in normal or diseased cells or in the cells from which the sequence was taken. Enriched does not imply that there are no other DNA or RNA sequences present, just that the relative amount of the sequence of interest has been significantly increased, for example, by 1 fold, 2 fold, 5 fold, 10 fold, 50 fold, 100 fold, 500 fold, 1000 fold, 10,000 fold, 100,000 fold, or 1,000,000 fold. The other DNA may, for example, be derived from a yeast or bacterial genome, or a cloning vector, such as a plasmid or a viral vector. It is advantageous for some purposes that a nucleotide sequence is in purified form.
The term "purified" in reference to nucleic acid represents that the sequence has increased purity relative to the natural environment, preferably, 50%, 60%, 70%, 80%, 90%, 95%, or 99% pure.
The terms "polynucleotide" and "nucleic acid sequence" are used interchangeably herein and include, but are not limited to, coding sequences (polynucleotide(s) or nucleic acid sequence(s) which are transcribed and translated into polypeptide in vitro or in vivo when placed under the control of appropriate regulatory or control sequences); control sequences (e.g., translational start and stop codons, promoter sequences, ribosome binding sites, polyadenylation signals, transcription factor binding sites, transcription termination sequences, upstream and downstream regulatory domains, enhancers, silencers, and the like); and regulatory sequences (DNA sequences to which a transcription factor(s) binds and alters the activity of a gene's promoter either positively (induction) or negatively (repression)). No limitation as to length or to synthetic origin are suggested by the terms described herein. As used herein the terms "polypeptide" and "protein" refer to a polymer of amino acids of three or more amino acids in a serial array, linked through peptide bonds. The term "polypeptide" includes proteins, protein fragments, protein analogues, oligopeptides and the like. The term "polypeptides" contemplates polypeptides as defined above that are encoded by nucleic acids, produced through recombinant technology (isolated from an appropriate source such as a bird), or synthesized. The term "polypeptides" further contemplates polypeptides as defined above that include chemically modified amino acids or amino acids covalently or noncovalently linked to labeling ligands.
The term "fragment" as used herein to refers to an at least 10, 20, 50, 75, 100, 150, 200, 250, 300, 500, 1000, 2000 or 5000 nucleotide long portion of a nucleic acid (e.g., cDNA) that has been constructed artificially (e.g., by chemical synthesis) or by cleaving a natural product into multiple pieces, using restriction endonucleases or mechanical shearing, or enzymatically, for example, by PCR or any other polymerizing technique known in the art, or expressed in a host cell by recombinant nucleic acid technology known to one of skill in the art. The term "fragment" as used herein may also refer to an at least 5, 10, 20, 30, 40, 50, 75, 100, 150, 200, 250, 300, 400, 500, 1000, 2000, or 5000 amino acid portion of a polypeptide, which portion is cleaved from a naturally occurring polypeptide by proteolytic cleavage by at least one protease, or is a portion of the naturally occurring polypeptide synthesized by chemical methods or using recombinant DNA technology (e.g., expressed from a portion of the nucleotide sequence encoding the naturally occurring polypeptide) known to one of skill in the art.
The term "gene" or "genes" as used herein refers to nucleic acid sequences (including both RNA or DNA) that encode genetic information for the synthesis of a whole RNA, a whole protein, or any portion of such whole RNA or whole protein. Genes that are not naturally part of a particular organism's genome are referred to as "foreign genes," "heterologous genes" or "exogenous genes" and genes that are naturally a part of a particular organism's genome are referred to as "endogenous genes". The term "gene product" refers to RNAs or proteins that are encoded by the gene. "Foreign gene products" are RNA or proteins encoded by "foreign genes" and "endogenous gene products" are RNA or proteins encoded by endogenous genes. "Heterologous gene products" are RNAs or proteins encoded by "foreign, heterologous or exogenous genes" and are, therefore, not naturally expressed in the cell.
The term "expressed" or "expression" as used herein refers to the transcription from a gene to give an RNA nucleic acid molecule at least complementary in part to a region of one of the two nucleic acid strands of the gene. The term "expressed" or "expression" as used herein also refers to the translation from said RNA nucleic acid molecule to give a protein, a polypeptide or a portion thereof.
As used herein, the term "locus" or "loci" refers to the site of a gene on a chromosome. Pairs of genes control hereditary traits, each in the same position on a pair of chromosomes. These gene pairs, or alleles, may both be dominant or both be recessive in expression of that trait. In either case, the individual is said to be homozygous for the trait controlled by that gene pair. If the gene pair (alleles) consists of one dominant and one recessive trait, the individual is heterozygous for the trait controlled by the gene pair. Natural variation in genes or nucleic acid molecules caused by, for example, recombination events or resulting from mutation, gives rise to allelic variants with similar, but not identical, nucleotide sequences. Such allelic variants typically encode proteins with similar activity to that of the protein encoded by the gene to which they are compared, because natural selection typically selects against variations that alter function. Allelic variants can also comprise alterations in the untranslated regions of the gene as, for example, in the 3' or 5' untranslated regions or can involve alternate splicing of a nascent transcript, resulting in alternative exons being positioned adjacently. The terms "operably linked" or "operatively linked" refer to the configuration of the coding and control sequences so as to perform the desired function. Thus, control sequences operably linked to a coding sequence are capable of effecting the expression of the coding sequence and regulating in which tissues, at what developmental timepoints, or in response to which signals, etc., a gene is expressed. A coding sequence is operably linked to or under the control of transcriptional regulatory regions in a cell when DNA polymerase will bind the promoter sequence and transcribe the coding sequence into mRNA that can be translated into the encoded protein. The control sequences need not be contiguous with the coding sequence, so long as they function to direct the expression thereof. Thus, for example, intervening untranslated yet transcribed sequences, can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered "operably linked" to the coding sequence. Such intervening sequences include but are not limited to enhancer sequences which are not transcribed or are not bound by polymerase. The terms "transcription regulatory sequences" and "gene expression control regions" as used herein refer to nucleotide sequences that are associated with a gene nucleic acid sequence and which regulate the transcriptional expression of the gene. Exemplary transcription regulatory sequences include enhancer elements, hormone response elements, steroid response elements, negative regulatory elements, and the like. The "transcription regulatory sequences" may be isolated and incorporated into a nucleic acid vector to enable regulated transcription in appropriate cells of portions of the vector DNA. The "transcription regulatory sequence" may precede, but is not limited to, the region of a nucleic acid sequence that is in the region 5' of the end of a protein coding sequence that may be transcribed into mRNA. Transcriptional regulatory sequences may also be located within a protein coding region, in regions of a gene that are identified as "intron" regions, or may be in regions of nucleic acid sequence that are in the region of nucleic acid.
The term "promoter' as used herein refers to the DNA sequence that determines the site of transcription initiation by an RNA polymerase. A "promoter-proximal element" may be a regulatory sequence within about 200 base pairs of the transcription start site. A "magnum-specific" promoter, as used herein, is a promoter that is primarily or exclusively active in the tubular gland cells of the avian magnum. Useful promoters also include exogenously inducible promoters. These are promoters that can be "turned on" in response to an exogenously supplied agent or stimulus, which is generally not an endogenous metabolite or cytokine. Examples include an antibiotic-inducible promoter, such as a tetracycline- inducible promoter, a heat-inducible promoter, a light- inducible promoter, or a laser inducible promoter, (e.g., Halloran et al, 2000, Development 127: 1953-1960; Gemer et al, 2000, Int. J. Hyperthermia 16: 171-81; Rang and Will, 2000, Nucleic Acids Res. 28: 1120-5; Hagihara et al, 1999, Cell Transplant 8: 4314; Huang et al, 1999, Mol. Med. 5: 129-37; Forster et al, 1999, Nucleic Acids Res. 27: 708-10; Liu et al, 1998, Biotechniques 24: 624-8, 630-2; the contents of which have been incorporated herein by reference in their entireties).
The term "coding region" as used herein refers to a continuous linear arrangement of nucleotides which may be translated into a protein. A full length coding region is translated into a full length protein; that is, a complete protein as would be translated in its natural state absent any post-translational modifications. A full length coding region may also include any leader protein sequence or any other region of the protein that may be excised naturally from the translated protein.
The term "complementary" as used herein refers to two nucleic acid molecules that can form specific interactions with one another. In the specific interactions, an adenine base within one strand of a nucleic acid can form two hydrogen bonds with thymine within a second nucleic acid strand when the two nucleic acid strands are in opposing polarities. Also in the specific interactions, a guanine base within one strand of a nucleic acid can form three hydrogen bonds with cytosine within a second nucleic acid strand when the two nucleic acid strands are in opposing polarities. Complementary nucleic acids as referred to herein, may further comprise modified bases wherein a modified adenine may form hydrogen bonds with a thymine or modified thymine, and a modified cytosine may form hydrogen bonds with a guanine or a modified guanine.
The term "probe" as used herein, when referring to a nucleic acid, refers to a nucleotide sequence that can be used to hybridize with and thereby identify the presence of a complementary sequence, or a complementary sequence differing from the probe sequence but not to a degree that prevents hybridization under the hybridization stringency conditions used. The probe may be modified with labels such as, but not only, radioactive groups, biotin, and the like that are well known in the art.
The term "capable of hybridizing under stringent conditions" as used herein refers to annealing a first nucleic acid to a second nucleic acid under stringent conditions as defined below. Stringent hybridization conditions typically permit the hybridization of nucleic acid molecules having at least 70% nucleic acid sequence identity with the nucleic acid molecule being used as a probe in the hybridization reaction. For example, the first nucleic acid may be a test sample or probe, and the second nucleic acid may be the sense or antisense strand of a ovomucoid gene expression control region or a fragment thereof. Hybridization of the first and second nucleic acids may be conducted under stringent conditions, e.g., high temperature and/or low salt content that tend to disfavor hybridization of dissimilar nucleotide sequences. Alternatively, hybridization of the first and second nucleic acid maybe conducted under
5 reduced stringency conditions, e.g. low temperature and/or high salt content that tend to favor hybridization of dissimilar nucleotide sequences. Low stringency hybridization conditions may be followed by high stringency conditions or intermediate medium stringency conditions to increase the selectivity of the binding of the first and second nucleic acids. The hybridization conditions may further include reagents such as, but not limited to, dimethyl
10 sulfoxide (DMSO) or formamide to disfavor still further the hybridization of dissimilar nucleotide sequences. A suitable hybridization protocol may, for example, involve hybridization in 6X SSC (wherein IX SSC comprises 0.015 M sodium citrate and 0.15 M sodium chloride), at 65° C in an aqueous solution, followed by washing with IX SSC at 65° C. Formulae to calculate appropriate hybridization and wash conditions to achieve
15 hybridization permitting 30% or less mismatch between two nucleic acid molecules are disclosed, for example, in Meinkoth et al. (1984) Anal. Biochem. 138: 267-284; the content of which is herein incorporated by reference in its entirety. Protocols for hybridization techniques are well known to those of skill in the art and standard molecular biology manuals may be consulted to select a suitable hybridization protocol without undue experimentation.
20 See, for example, Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Press, the contents of which are herein incorporated by reference in their entirety.
Typically, stringent conditions will be those in which the salt concentration is less than about 1.5 M sodium ion, typically about 0.01 to 1.0 M Na ion concentration (or other
25 salts) from about pH 7.0 to about pH 8.3 and the temperature is at least about 30° C for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCl, 1% SDS (sodium dodecyl sulphate) at
30 37° Celsius, and a wash in lx to 2x SSC at 50 to 55° Celsius. Exemplary moderate stringency conditions include hybridization in 40 to 45 % formamide, 1 M NaCl, 1% SDS at 37° Celsius, and a wash in 0.5x to lx SSC at 55 to 60° Celsius. Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° Celsius, and a wash in 0.1 x SSC at 60 to 65° Celsius.
35 The terms "unique nucleic acid region" and "unique protein (polypeptide) region" as used herein refer to sequences present in a nucleic acid or protein (polypeptide) respectively that is not present in any other nucleic acid or protein sequence. The terms "conserved
5 nucleic acid region" as referred to herein is a nucleotide sequence present in two or more nucleic acid sequences, to which a particular nucleic acid sequence can hybridize under low, medium or high stringency conditions. The greater the degree of conservation between the conserved regions of two or more nucleic acid sequences, the higher the hybridization stringency that will allow hybridization between the conserved region and a particular nucleic
10 acid sequence.
The terms "percent sequence identity" or "percent sequence similarity" as used herein refer to the degree of sequence identity between two nucleic acid sequences or two amino acid sequences as determined using the algorithm of Karlin & Attschul (1990) Proc. Natl. Acad. Sci. 87: 2264-2268, modified as in Karlin & Attschul (1993) Proc. Natl. Acad. Sci. 90:
15 5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Attschul et al. (1990) T. Mol. Biol. Q15: 403-410. BLAST nucleotide searches are performed with the NBLAST program, score = 100, wordlength = 12, to obtain nucleotide sequences homologous to a nucleic acid molecule of the invention. BLAST protein searches are performed with the XBLAST program, score = 50, wordlength = 3, to obtain amino acid
20 sequences homologous to a reference polypeptide. To obtain gapped alignments for comparison purposes, Gapped BLAST is utilized as described in Attschul et al. (1997) Nucl. Acids Res. 25: 3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g. XBLAST and NBLAST) are used. Other algorithms, programs and default settings may also be suitable such as, but not only, the 5 GCG-Sequence Analysis Package of the U.K. Human Genome Mapping Project Resource Centre that includes programs for nucleotide or amino acid sequence comparisons.
The term "sense strand" as used herein refers to a single stranded DNA molecule from a genomic DNA that may be transcribed into RNA and translated into the natural polypeptide product of the gene. The term "antisense strand" as used herein refers to the single strand 0 DNA molecule of a genomic DNA that is complementary with the sense strand of the gene. The term "antisense DNA" as used herein refers to a gene sequence DNA that has a nucleotide sequence complementary to the "sense strand" of a gene when read in reverse orientation, i.e., DNA read into RNA in a 3' to 5' direction rather than in the 5' to 3' direction. The term "antisense RNA" is used to mean an RNA nucleotide sequence (for example that 5 encoded by an antisense DNA or synthesized complementary with the antisense DNA). Antisense RNA is capable of hybridizing under stringent conditions with an antisense DNA. The antisense RNA of the invention is useful for regulating expression of a "target gene" either at the transcriptional or translational level. For example, transcription of the subject nucleic acids may produce antisense transcripts that are capable of inhibiting transcription by inhibiting initiation of transcription or by competing for limiting transcription factors; the antisense transcripts may inhibit transport of the "target RNA", or, the antisense transcripts may inhibit translation of "target RNA".
The term "nucleic acid vector" as used herein refers to a natural or synthetic single or double stranded plasmid or viral nucleic acid molecule, or any other nucleic acid molecule, such as but not limited to YACs, BACs, bacteriophage-derived artificial chromosome (BBPAC), cosmid or PI derived artificial chromosome (PAC), that can be transfected or transformed into cells and replicate independently of, or within, the host cell genome. A circular double stranded vector can be linearized by treatment with an appropriate restriction enzyme based on the nucleotide sequence of the vector. A nucleic acid can be inserted into a vector by cutting the vector with restriction enzymes and ligating the pieces together. The nucleic acid molecule can be RNA or DNA.
The term "expression vector" as used herein refers to a nucleic acid vector that comprises the ovomucoid gene expression control region operably linked to a nucleotide sequence coding at least one polypeptide. As used herein, the term "regulatory sequences" includes promoters, enhancers, and other elements that may control gene expression. Standard molecular biology textbooks such as Sambrook et al. eds "Molecular Cloning: A Laboratory Manual" 3rd ed., Cold Spring Harbor Press (2001) maybe consulted to design suitable expression vectors that may further include an origin of replication and selectable gene markers. It should be recognized, however, that the choice of a suitable expression vector and the combination of functional elements therein depends upon multiple factors including the choice of the host cell to be transformed and/or the type of protein to be expressed.
The terms "transformation" and "transfection" as used herein refer to the process of inserting a nucleic acid into a host. Many techniques are well known to those skilled in the art to facilitate transformation or transfection of a nucleic acid into a prokaryotic or eukaryotic organism. These methods involve a variety of techniques, such as treating the cells with high concentrations of salt such as, but not only, a calcium or magnesium salt, an electric field, detergent, or liposome mediated transfection, to render the host cell competent for the uptake of the nucleic acid molecules, and by such methods as sperm-mediated and restriction-mediated integration.
The term "transfecting agent" as used herein refers to a composition of matter added to the genetic material for enhancing the uptake of heterologous DNA segment(s) into a eukaryotic cell, preferably an avian cell. The enhancement is measured relative to the uptake in the absence of the transfecting agent. Examples of transfecting agents include adenovirus-transferrin-polylysine-DNA complexes. These complexes generally augment the uptake of DNA into the cell and reduce its breakdown during its passage through the cytoplasm to the nucleus of the cell. These complexes can be targeted to, e.g., the male germ cells using specific ligands that are recognized by receptors on the cell surface of the germ cell, such as the c-kit ligand or modifications thereof.
Other preferred transfecting agents include but are not limited to lipofectin, lipfectamine, DIMRIE C, Supeffect, and Effectin (Qiagen), unifectin, maxifectin, DOTMA, DOGS (Transfectam; dioctadecylamidoglycylspermine), DOPE (1,2-dioleoyl-sn-glycero- 3-phosphoethanolamine), DOTAP (l,2-dioleoyl-3-trimethylammonium propane), DDAB (dimethyl dioctadecytammonium bromide), DHDEAB (N,N-di-n-hexadecyl-N,N- dihydroxyethyl ammonium bromide), HDEAB (N-n-hexadecylN,N- dihydroxyethylammonium bromide), polybrene, or poly(ethylenimine) (PEI). These non- viral agents have the advantage that they can facilitate stable integration of xenogeneic DNA sequences into the vertebrate genome, without size restrictions commonly associated with virus-derived transfecting agents.
The term "recombinant cell" refers to a cell that has a new combination of nucleic acid segments that are not covalently linked to each other in nature in that particular configuration. A new configuration of nucleic acid segments can be introduced into an organism using a wide array of nucleic acid manipulation techniques available to those skilled in the art. A recombinant cell can be a single eukaryotic cell, such as a mammalian or avian cell (including within a transgenic mammal or avian) or a single prokaryotic cell. The recombinant cell may harbor a vector that is extragenomic. An extragenomic nucleic acid vector does not insert into the cell's genome. A recombinant cell may further harbor a vector or a portion thereof (e.g., the portion containing the regulatory sequences and the coding sequence) that is intragenomic. The term intragenomic defines a nucleic acid construct incorporated within the recombinant cell's genome.
The terms "recombinant nucleic acid" and "recombinant DNA" as used herein refer a combination of at least two nucleic acids that is not naturally found in a eukaryotic or prokaryotic cell in that particular configuration. The nucleic acids may include, but are not limited to, nucleic acid vectors, gene expression regulatory elements, origins of replication, suitable gene sequences that when expressed confer antibiotic resistance, protein-encoding sequences and the like. The term "recombinant polypeptide" is meant to include a polypeptide produced by recombinant DNA techniques such that it is distinct from a naturally occurring polypeptide either in its location, purity or structure. Generally, such a recombinant polypeptide will be present in a cell in an amount different from that normally observed in nature. Pharmaceutical compositions comprising agents that will modulate the regulation of the expression of a polypeptide-encoding nucleic acid operably linked to a ovomucoid gene expression control region can be administered in dosages and by techniques well known to those skilled in the medical or veterinary arts, taking into consideration such factors as the age, sex, weight, species and condition of the recipient animal, and the route of administration. The route of administration can be percutaneous, via mucosal administration (e.g., oral, nasal, anal, vaginal) or via a parenteral route (intradermal, intramuscular, subcutaneous, intravenous, or intraperitoneal). Pharmaceutical compositions can be administered alone, or can be co-administered or sequentially administered with other treatments or therapies. Forms of administration may include suspensions, syrups or elixirs, and preparations for parenteral, subcutaneous, intradermal, intramuscular or intravenous administration (e.g., injectable administration) such as sterile suspensions or emulsions. Pharmaceutical compositions may be administered in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, or the like. The compositions can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, adjuvants, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired. Standard pharmaceutical texts, such as Remmington's Pharmaceutical Science, 17th edition, 1985 may be consulted to prepare suitable preparations, without undue experimentation. Dosages can generally range from a few hundred milligrams to a few grams.
As used herein, a "transgenic animal" is any non-human animal, such as an avian species, including the chicken, in which one or more of the cells of the animal contain a heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into a cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA. In the typical transgenic animal, the transgene causes cells to express a recombinant form of the subject polypeptide, e.g. either agonistic or antagonistic forms, or in which the gene has been disrupted. In certain embodiments, the genome of the animal has been modified such that a heterologous gene expression element is inserted so as to be operably linked to an endogenous coding sequence. The terms "chimeric animal" or "mosaic animal" are used herein to refer to animals in which the recombinant gene is found, or in which the recombinant gene is expressed in some but not all cells of the animal. The term "tissue-specific chimeric animal" indicates that the recombinant gene is present and/or expressed in some tissues but not others. As used herein, the term "transgene" means a nucleic acid sequence (encoding, for example, a human interferon polypeptide) that is partly or entirely heterologous, i.e., foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted (e.g., it is inserted at a location that differs from that of the natural gene or its insertion results in a knockout). A trangene also includes a regulatory sequence designed to be inserted into the genome such that it regulates the expression of an endogenous coding sequence, e.g., to increase expression and or to change the timing and or tissue specificity of expression, etc. (e.g., to effect "gene activation"). The term "cytokine" as used herein refers to any secreted polypeptide that affects the functions of cells and is a molecule that modulates interactions between cells in the immune, inflammatory or hematopoietic responses. A cytokine includes, but is not limited to, monokines and lymphokines regardless of which cells produce them. For instance, a monokine is generally referred to as being produced and secreted by a mononuclear cell, such as a macrophage and/or monocyte. Many other cells however also produce monokines, such as natural killer cells, fibroblasts, basophils, neutrophils, endothelial cells, brain astrocytes, bone marrow stromal cells, epideral keratinocytes and B-lymphocytes. Lymphokines are generally referred to as being produced by lymphocyte cells. Examples of cytokines include, but are not limited to, Interleukin-1 (IL-1), Interleukin-6 (IL-6), Interleukin-8 (IL-8), Tumor Necrosis Factor-alpha (TNF-alpha) and Tumor Necrosis Factor beta (TNF-beta). The term "antibody" as used herein refers to polyclonal and monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof. The term "antibody" refers to a homogeneous molecular entity, or a mixture such as a polyclonal serum product made up of a plurality of different molecular entities, and may further comprise any modified or derivatised variant thereof that retains the ability to specifically bind an epitope. A monoclonal antibody is capable of selectively binding to a target antigen or epitope. Antibodies may include, but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, camelized antibodies, single chain antibodies (scFvs), Fab fragments, F(ab')2 fragments, disulfide-linked Fvs (sdFv) fragments, e.g., as produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, intrabodies, synthetic antibodies, and epitope-binding fragments of any of the above.
The term "immunoglobulin polypeptide" as used herein refers to a polypeptide derived from a constituent polypeptide of an immunoglobulin. An "immunoglobulin polypeptide" may be, but is not limited to, an immunoglobulin (preferably an antibody) heavy or light chain and may include a variable region, a diversity region, joining region and a constant region or any combination, variant or truncated form thereof. The term "immunoglobulin polypeptides" further includes single-chain antibodies comprised of, but not limited to, an immunoglobulin heavy chain variable region, an immunoglobulin light chain variable region and optionally a peptide linker.
The techniques used to isolate and characterize the nucleic acids and proteins of the present invention are well known to those of skill in the art and standard molecular biology and biochemical manuals may be consulted to select suitable protocols without undue experimentation. See, for example, Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Press, the content of which is herein incorporated by reference in its entirety.
This description uses gene nomenclature accepted by the Cucurbit Genetics Cooperative as it appears in the Cucurbit Genetics Cooperative Report 18:85 (1995), herein incorporated by reference in its entirety. Using this gene nomenclature, genes are symbolized by italicized Roman letters. If a mutant gene is recessive to the normal type, then the symbol and name of the mutant gene appear in italicized lower case letters.
Abbreviations Abbreviations used in the present specification include the following: aa, amino acid(s); bp, base pair(s); cDNA, DNA complementary to RNA; nt, nucleotide(s); SSC, sodium chloride-sodium citrate; DMSO, dimethyl sulfoxide.
Additional objects and aspects of the present invention will become more apparent upon review of the detailed description set forth below when taken in conjunction with the accompanying figures, which are briefly described as follows.
4. Brief Description of the Figures
FIG. 1 illustrates an agarose gel analysis of PCR products from PCR amplification of chicken genomic DNA using the primers OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2).
FIG. 2 illustrates the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site, and the positions and orientations of primers used to sequence this region.
FIG. 3 shows the PCR primers SEQ ID NOS: 1 - 25 used to PCR amplify and/or sequence the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site.
FIG. 4 shows the nucleic acid sequence SEQ ID NO: 26 of the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site.
FIG. 5 illustrates the 10 kb ovomucoid promoter linked to the luciferase or human IFNα-2b coding sequences.
FIG. 6 A shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a luciferase gene into HDl 1 cells, a chicken myeloid cell line. FIG. 6B shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a luciferase gene into primary quail tubular gland cells isolated from the magnum portion of the oviduct of a laying quail hen.
FIG. 7 shows the results of transfections of plasmids containing the ovomucoid promoter or CMV promoter linked to a interferon gene into primary quail tubular gland cells isolated from the magnum portion of the oviduct of a laying quail hen.
5. Detailed Description of the Invention
5.1 Chicken ovomucoid gene expression control region nucleic acid sequences
A series of PCR amplifications of template chicken genomic DNA were used to isolate the gene expression control region of the chicken ovomucoid locus. The region of the chicken genome lying between the 3' end of the ovoinhibitor gene and the 5' transcription start site of the ovomucoid gene was PCR amplified using the primers OVINs 2,
5'-TAGGCAGAGCAATAGGACTCTCAACCTCGT-3' (SEQ ID NO: 1) and OVMUa2, 5'-AAGCTTCTGCAGCACTCTGGGAGTTACTCA-3' (SEQ ID NO: 2) as described in detail in Example 1 below and FIG. 1. The approximately 10 kb fragment was blunt-ended and cleaved with the restriction endonuclease Bam HI. The resulting fragments of about 4.7 kb and 5.5 kb were subcloned into the linearized plasmid vector pBluescript KS II (+/-)
(Stratagene, La Jolla, CA). Each insert was sequenced using the primers SEQ ID NOS: 5 - 25 shown in Figs. 2 and 3 and as described in Example 3 below. The compiled nucleic acid sequence (SEQ ID NO: 26) of the approximately 10 kb nucleic acid region that is 5' upstream of the chicken ovomucoid transcription start site is shown in FIG. 4. SEQ ID NO: 26 includes the ovoinhibitor gene 3' untranslated region described by
Scott et al. (J. Biol. Chem. 262: 5899 -5909 (1987)) from bases positions 1-255 as shown in FIG. 4. A CRl-like element (Scott et al, Biochemistry 26: 6831-6840 (1987); Genbank Accession No: M17966) is located at base positions 2761-3024 as shown in FIG. 4. The region of SEQ ID NO: 26 from base positions 9403-9920, as shown in FIG. 4, has been described in Genbank Accession No: J00897 and in Lai et al, Cell 18: 829-842 (1979) and includes a portion of the 5' untranslated region of the ovomucoid gene.
It is contemplated that any nucleic acid sequence encoding a polypeptide may be operably linked to the avian ovomucoid gene expression control region of the invention so as to be expressed in a transfected avian cell. For example, a plasmid construct contacting the cloned ovomucoid promoter region and a desired polypeptide-encoding nucleic acid sequence may be transfected into cultured quail or chicken oviduct cells, which may then be incubated to synthesize a polypeptide detectable with antibodies directed against the desired polypeptide.
The present invention can be used to express, in large yields and at low cost, a wide range of desired proteins including those used as human and animal pharmaceuticals, diagnostics, and livestock feed additives. Proteins such as growth hormones, cytokines, structural proteins and enzymes, including human growth hormone, interferon, lysozyme, and β-casein, are examples of proteins that are desirably expressed in the oviduct and deposited in eggs according to the invention. Other possible proteins to be produced include, but are not limited to, albumin, α-1 antitrypsin, antithrombin m, collagen, factors VIH, IX, X (and the like), fibrinogen, hyaluronic acid, insulin, lactoferrin, protein C, erythropoietin (EPO), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), tissue-type plasminogen activator (tPA), feed additive enzymes, somatotropin, and chymotrypsin Immunoglobulins and genetically engineered antibodies, including immunotoxins that bind to surface antigens on human tumor cells and destroy them, can also be expressed for use as pharmaceuticals or diagnostics. It is contemplated that immunoglobulin polypeptides expressed in avian cells following transfection by the methods of the present invention may include monomeric heavy and light chains, single-chain antibodies or multimeric immunoglobulins comprising variable heavy and light chain regions, i.e., antigen-binding domains, or intact heavy and light immunoglobulin chains.
The chicken ovomucoid gene expression control region of the present invention comprises the nucleotide elements that are positioned 5' upstream of the transcription start site of the native chicken ovomucoid locus and which are necessary for the regulated expression of a downstream polypeptide-encoding nucleic acid. It is contemplated that this region includes those transcription control regions regulatable by hormones including, for example, steroid hormones and the like.
One aspect of the present invention, therefore, provides a novel isolated nucleic acid that comprises the nucleotide sequence SEQ ID NO: 26, shown in FIG. 4, (Genbank Accession No: AF 453747) and derivatives and variants thereof, that is located immediately 5' upstream of the transcription start site of the chicken ovomucoid gene locus.
In one embodiment of the present invention, the isolated nucleic acid may be isolated from an avian selected from the group consisting of a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird. In another embodiment of the present invention, the isolated nucleic acid is obtained from a chicken. In this embodiment, the isolated nucleic acid has the sequence of SEQ ID NO: 26, as shown in FIG. 4, or a variant thereof. SEQ ID NO: 26 was cloned into pBluescript KS π (+/-) vector, as described in Example 2, and named pBS-OVMUP-10. pBS-OVMUP-10 was deposited with American Type Culture Collection (ATCC) as ATCC No. on November 26, 2002 under the conditions set forth in the Budapest Treaty. Another aspect of the invention provides nucleic acids that can hybridize under high, medium or low stringency conditions to an isolated nucleic acid comprising a chicken ovomucoid gene expression control region having all, a derivative of, or a portion of the nucleic acid sequence SEQ ID NO: 26 shown in FIG. 4 and direct expression of a polypeptide coding sequence in an avian oviduct cell. The nucleotide sequence determined from the isolation of the ovomucoid gene expression control region from a chicken (SEQ ID NO: 26) will allow for the generation of probes designed for use in identifying ovomucoid gene expression control regions, or homo logs thereof in other avian species. Fragments of a nucleic acid comprising a portion of the subject ovomucoid gene expression control region are also within the scope of the invention. As used herein, a fragment of the nucleic acid comprising an active portion of a ovomucoid gene expression control region refers to a nucleotide sequence having fewer nucleotides than the nucleotide sequence comprising the entire nucleic acid sequence of the ovomucoid gene expression control region.
A fragment of the ovomucoid gene expression control region may contain one or more of the following elements: the ovoinhibitor gene 3' untranslated region from bases positions 1-255 as shown in FIG. 4, a CRl-like element located at base positions 2761-3024 as shown in FIG. 4, the region from base positions 9403-9920, as shown in FIG. 4 which includes a portion of the 5' untranslated region of the ovomucoid gene. Alternatively, the fragment may be 10, 20, 50, 75, 100, 150, 200, 250, 300, 500, 1000, 2000, 4000, 5000, 6000, 7000, 8000 or 9000 nucleotides in length and be capable of directing expression of an operably linked heterologous gene sequence, particularly in an avian oviduct cell.
In one embodiment of the present invention, the nucleotide sequence of the isolated DNA molecule of the present invention may be used as a probe in nucleic acid hybridization assays for the detection of the ovomucoid gene expression control region. The nucleotide sequence of the present invention may be used in any nucleic acid hybridization assay system known in the art, including, but not limited to, Southern blots (Southern, EM. J. Mol. Biol. 98: 508 (1975)), Northern blots (Thomas et al, Proc. Natl. Acad. Sci. 77: 5201-05 (1980)), and Colony blots (Grunstein et al, Proc. Natl. Acad. Sci. 72: 3961-65 (1975)), which are hereby incorporated by reference in their entireties. Alternatively, the isolated DNA molecules of the present invention can be used in a gene amplification detection procedure such as a polymerase chain reaction (Erlich et al, Science 252: 1643-51 (1991), which is hereby incorporated by reference in its entirety) or in restriction fragment length polymorphism (RFLP) diagnostic techniques, as described in Watson et al, (2d ed. 1992), Recombinant DNA, Scientific American Books, 519-522, 545-547, which is hereby incorporated by reference.
Nucleic acids constructed in accordance with the present invention can be labeled to provide a signal as a means of detection. For example, radioactive elements such as 32P, 3H, and 35S or the like provide sufficient half-life to be useful as radioactive labels. Other materials useful for labeling synthetic nucleotides include fluorescent compounds, enzymes and chemiluminescent moieties. Methods useful in selecting appropriate labels and binding protocols for binding the labels to the synthetic nucleotides are well known to those of skill in the art. Standard immunology manuals such as Promega: Protocol and Applications Guide. 2nd Edition, 1991 (Promega Corp., Madison, WI, the content of which is incorporated herein in its entirety) may be consulted to select an appropriate labeling protocol without undue experimentation.
In another embodiment of the present invention, an isolated nucleic acid molecule of the present invention includes a nucleic acid that is at least about 75%, preferably at least about 80%, more preferably at least about 85%, even more preferably at least about 90%, still more preferably at least about 95%, and even more preferably at least about 99%, identical to a chicken-derived ovomucoid gene expression control region -comprising nucleic acid molecule as depicted in SEQ ID NO: 26 and directs expression of a polypeptide encoding sequence in an avian oviduct cell, when operably linked to the polypeptide encoding sequence.
In another embodiment of the present invention, an isolated nucleic acid molecule of the present invention includes a nucleic acid that hybridizes to SEQ ID NO: 26 or the reverse complement thereof, or the insert in pBS-OVMUP-10, under high, moderate or low stringency hybridization conditions.
In another embodiment of the present invention, an avian ovomucoid gene expression control region gene or nucleic acid molecule can be an allelic variant of SEQ ID NO: 26 or a homolog from a different avian, e.g., quail, duck, etc.
The present invention also contemplates the use of antisense nucleic acid molecules that are designed to be complementary to a coding strand of a nucleic acid (i.e., complementary to an mRNA sequence) or, alternatively, complimentary to a 5' or 3' untranslated region of the mRNA. Another use of synthetic nucleotides is as primers (DNA or RNA) for a polymerase chain reaction (PCR), ligase chain reaction (LCR), or the like. Synthesized oligonucleotides can be produced in variable lengths. The number of bases synthesized will depend upon a variety of factors, including the desired use for the probes or primers. Additionally, sense or anti-sense nucleic acids or oligonucleotides can be chemically synthesized using modified nucleotides to increase the biological stability of the molecule or of the binding complex formed between the anti-sense and sense nucleic acids. For example, acridine substituted nucleotides can be synthesized. Protocols for designing isolated nucleotides, nucleotide probes, and/or nucleotide primers are well-known to those of ordinary skill, and can be purchased commercially from a variety of sources (e.g., Sigma Genosys, The Woodlands, TX or The Great American Gene Co., Ramona, CA).
The nucleic acid sequence of a chicken ovomucoid gene expression control region nucleic acid molecule (SEQ ID NO: 26) of the present invention allows one skilled in the art to, for example, (a) make copies of those nucleic acid molecules by procedures such as, but not limited to, insertion into a cell for replication by the cell, by chemical synthesis or by procedures such as PCR or LCR, (b) obtain nucleic acid molecules which include at least a portion of such nucleic acid molecules, including full-length genes, full-length coding regions, regulatory control sequences, truncated coding regions and the like, (c) obtain ovomucoid gene expression control region nucleic acid homologs in other avian species such as, but not limited to, turkey, duck, goose, quail, pheasant, parrot, finch, ratites including ostrich, emu and cassowary and, (d) to obtain isolated nucleic acids capable of hybridizing to an avian ovomucoid gene expression control region nucleic acid and be used to detect the presence of nucleic acid-related sequences by complementation between the probe and the target nucleic acid.
Such nucleic acid homologs can be obtained in a variety of ways including by screening appropriate expression libraries with antibodies of the present invention, using traditional cloning techniques to screen appropriate libraries, amplifying appropriate libraries or DNA using oligonucleotide primers of the present invention in a polymerase chain reaction or other amplification method, and screening public and/or private databases containing genetic sequences using nucleic acid molecules of the present invention to identify targets. Examples of preferred libraries to screen, or from which to amplify nucleic acid molecules, include but are not limited to mammalian BAC libraries, genomic DNA libraries, and cDNA libraries. Similarly, preferred sequence databases useful for screening to identify sequences in other species homologous to chicken ovomucoid gene expression control region include, but are not limited to, GenBank and the mammalian Gene Index database of The Institute of Genomics Research (TIGR).
Codon-optimized proteins Another aspect of the present invention is a recombinant DNA molecule comprising the novel isolated avian ovomucoid gene expression control region of the present invention operably linked to a selected polypeptide-encoding nucleic acid insert, and which may express the nucleic acid insert when transfected to a suitable host cell, preferably an avian cell. The nucleic acid insert may be placed in frame with a signal peptide sequence, whereby translation initiation from the transcript may start with the signal peptide and continue through the nucleic acid insert, thereby producing an expressed polypeptide having the desired amino acid sequence. It is anticipated that the recombinant DNA may further comprise a polyadenylation signal sequence that will allow the transcript directed by the novel ovomucoid gene expression control region to proceed beyond the nucleic acid insert encoding a polypeptide and allow the transcript to further comprise a 3' untranslated region and a polyadenylated tail. Any functional polyadenylation signal sequence may be linked to the 3' end of the nucleic acid insert including the S V40 polyadenylation signal sequence, bovine growth hormone adenylation sequence or the like, or derivatives thereof.
Another aspect of the present invention is to provide nucleic acid sequences of a protein optimized for expression in avian cells, and derivatives and fragments thereof.
One embodiment of the present invention is a recombinant DNA molecule comprising the isolated avian ovomucoid gene expression control region of the present invention, operably linked to a nucleic acid insert encoding a polypeptide, and a polyadenylation signal sequence optionally operably linked thereto. It is contemplated that when the recombinant DNA is to be delivered to a recipient cell for expression therein, the sequence of the nucleic acid sequence may be modified so that the codons are optimized for the codon usage of the recipient species. When a heterologous nucleic acid is to be delivered to a recipient cell for expression therein, the sequence of the nucleic acid sequence may be modified so that the codons are optimized for the codon usage of the recipient species. For example, if the heterologous nucleic acid is transfected into a recipient chicken cell, the sequence of the expressed nucleic acid insert is optimized for chicken codon usage. This may be determined from the codon usage of at least one, and preferably more than one, protein expressed in a chicken cell. For example, the codon usage may be determined from the nucleic acid sequences encoding the proteins ovalbumin, lysozyme, ovomucin and ovotransferrin of chicken. Briefly, the DNA sequence for the target protein may be optimized using the BACKTRANSLATE® program of the Wisconsin Package, version 9.1 (Genetics Computer Group, Inc., Madison, WI) with a codon usage table compiled from the chicken (Gallus gallus) ovalbumin, lysozyme, ovomucoid, and ovotransferrin proteins. The template and primer oligonucleotides are then amplified, by any means known in the art, including but not limited to PCR with Pfu polymerase (STRATAGENE®, La Jolla CA).
5 In one exemplary embodiment of a heterologous nucleic acid for use by the methods of the present invention, a nucleic acid insert encoding the human interferon α2b polypeptide optimized for codon-usage by the chicken is used. Optimization of the sequence for codon usage is useful in elevating the level of translation in avian eggs.
It is contemplated to be within the scope of the present invention for any nucleic acid
10 encoding a polypeptide to be optimized for expression in avian cells. It is further contemplated that the codon usage may be optimized for a particular avian species used as a source of the host cells. In one embodiment of the present invention, the heterologous polypeptide is encoded using the codon-usage of a chicken.
In yet another embodiment of the present invention, the recombinant DNA comprises
15 the isolated avian ovomucoid gene expression control region operably linked to a nucleic acid encoding a human interferon α2b and the SV40 polyadenylation sequence.
The protein of the present invention may be produced in purified form by any known conventional technique. In a preferred embodiment, the protein is purified from chicken eggs, preferably egg whites. For example, chicken cells may be homogenized and
20 centrifuged. The supernatant is then subjected to sequential ammonium sulfate precipitation and heat treatment. The fraction containing the protein of the present invention is subjected to gel filtration in an appropriately sized dextran or polyacrylamide column to separate the proteins. If necessary, the protein fraction may be further purified by HPLC.
25 Multimeric proteins
The invention, in preferred embodiments, provides methods for producing multimeric proteins, preferably immunoglobulins, such as antibodies, and antigen binding fragments thereof.
In one embodiment of the present invention, the multimeric protein is an
30 immunoglobulin, wherein the first and second heterologous polypeptides are an immunoglobulin heavy and light chains respectively. Illustrative examples of this and other aspects and embodiments of the present invention for the production of heterologous multimeric polypeptides in avian cells are fully disclosed in U.S. Patent Application No. 09/877,374, filed June 8, 2001, by Rapp, published as US-2002-0108132-A1 on August 8,
35 2002, and U.S. Patent Application No. 10/251,364, filed September 18, 2002, by Rapp, both of which are incorporated herein by reference in their entirety. In one embodiment of the present invention, therefore, the multimeric protein is an immunoglobulin wherein the first and second heterologous polypeptides are an immunoglobulin heavy and light chain respectively. Accordingly, the invention provides immunoglobulin and other multimeric proteins that have been produced by transgenic avians of the invention.
In the various embodiments of this aspect of the present invention, an immunoglobulin polypeptide encoded by the transcriptional unit of at least one expression vector may be an immunoglobulin heavy chain polypeptide comprising a variable region or a variant thereof, and may further comprise a D region, a J region, a C region, or a combination thereof. An immunoglobulin polypeptide encoded by the transcriptional unit of an expression vector may also be an immunoglobulin light chain polypeptide comprising a variable region or a variant thereof, and may further comprise a J region and a C region. It is also contemplated to be within the scope of the present invention for the immunoglobulin regions to be derived from the same animal species, or a mixture of species including, but not only, human, mouse, rat, rabbit and chicken. In preferred embodiments, the antibodies are human or humanized.
In other embodiments of the present invention, the immunoglobulin polypeptide encoded by the transcriptional unit of at least one expression vector comprises an immunoglobulin heavy chain variable region, an immunoglobulin light chain variable region, and a linker peptide thereby forming a single-chain antibody capable of selectively binding an antigen.
Another aspect of the present invention provides a method for the production in an avian of an heterologous protein capable of forming an antibody suitable for selectively binding an antigen comprising the step of producing a transgenic avian incorporating at least one transgene, wherein the transgene encodes at least one heterologous polypeptide selected from an immunoglobulin heavy chain variable region, an immunoglobulin heavy chain comprising a variable region and a constant region, an immunoglobulin light chain variable region, an immunoglobulin light chain comprising a variable region and a constant region, and a single-chain antibody comprising two peptide-linked immunoglobulin variable regions. In an embodiment of this method of the present invention, the isolated heterologous protein is an antibody capable of selectively binding to an antigen. In one embodiment, the antibody may be generated by combining at least one immunoglobulin heavy chain variable region and at least one immunoglobulin light chain variable region, preferably cross-linked by at least one di-sulfide bridge. The combination of the two variable regions will generate a binding site capable of binding an antigen using methods for antibody reconstitution that are well known in the art.
It is, however, contemplated to be within the scope of the present invention for immunoglobulin heavy and light chains, or variants or derivatives thereof, to be expressed in separate transgenic avians, and therefore isolated from separate media including serum or eggs, each isolate comprising a single species of immunoglobulin polypeptide. The method may further comprise the step of combining a plurality of isolated heterologous immunoglobulin polypeptides, thereby producing an antibody capable of selectively binding to an antigen. In this embodiment, two individual transgenic avians maybe generated wherein one transgenic produces serum or eggs having an immunoglobulin heavy chain variable region, or a polypeptide comprising such, expressed therein. A second transgenic animal, having a second transgene, produces serum or eggs having an immunoglobulin light chain variable region, or a polypeptide comprising such, expressed therein. The polypeptides may be isolated from their respective sera and eggs and combined in vitro to generate a binding site capable of binding an antigen.
Examples of therapeutic antibodies that can be used in methods of the invention include but are not limited to HERCEPTIN® (Trastuzumab) (Genentech, CA) which is a humanized anti-HER2 monoclonal antibody for the treatment of patients with metastatic breast cancer; REOPRO® (abciximab) (Centocor) which is an anti-glycoprotein flb/πia receptor on the platelets for the prevention of clot formation; ZENAPAX® (daclizumab) (Roche Pharmaceuticals, Switzerland) which is an immunosuppressive, humanized anti- CD25 monoclonal antibody for the prevention of acute renal allograft rejection; PANOREX™ which is a niurine anti-17-IA cell surface antigen IgG2a antibody (Glaxo Wellcome/Centocor); BEC2 which is a murine anti-idiotype (GD3 epitope) IgG antibody (ImClone System); 1MC-C225 which is a chimeric anti-EGFR IgG antibody (ImClone System); VITAXTN™ which is a humanized anti-αVβ3 integrin antibody (Applied Molecular Evolution/Medlmmune); Campath 1H/LDP-03 which is a humanized anti CD52 IgGl antibody (Leukosite); Smart Ml 95 which is a humanized anti-CD33 IgG antibody (Protein Design Lab/Kanebo); RITUXAN™ which is a chimeric anti-CD20 IgGl antibody (IDEC Pharm/Genentech, Roche/Zettyaku); LYMPHOCIDE™ which is a humanized anti-CD22 IgG antibody (Immunomedics); ICM3 is a humanized anti-ICAM3 antibody (ICOS Pharm); IDEC-114 is a primatied anti-CD80 antibody (IDEC Pharm/Mitsubishi); ZEVALIN™ is a radiolabelled murine anti-CD20 antibody (IDEC/Schering AG); IDEC-131 is a humanized anti-CD40L antibody (IDEC/Eisai); IDEC-151 is a primatized anti-CD4 antibody (IDEC); IDEC- 152 is a primatized anti-CD23 antibody (IDEC/Seikagaku); SMART anti-CD3 is a humanized anti-CD3 IgG (Protein Design Lab); 5G1.1 is a humanized anti-complement factor 5 (C5) antibody (Alexion Pharm); D2E7 is a humanized anti-TNF-α antibody (CAT/BASF); CDP870 is a humanized anti-TNF-α Fab fragment (Celltech); IDEC- 151 is a primatized anti-CD4 IgGl antibody (IDEC Pharm/SmithKline Beecham); MDX-CD4 is a human anti-CD4 IgG antibody (Medarex/Eisai/Genmab); CDP571 is a humanized anti-TNF- α IgG4 antibody (Celltech); LDP-02 is a humanized anti-α4β7 antibody (LeukoSite/Genentech); OrthoClone OKT4A is a humanized anti-CD4 IgG antibody (Ortho Biotech); ANTOVA™ is a humanized anti-CD40L IgG antibody (Biogen); ANTEGREN™ is a humanized anti-VLA-4 IgG antibody (Elan); and CAT- 152 is a human anti-TGF-β2 antibody (Cambridge Ab Tech).
Recombinant nucleic acids, and expression thereof, under the control of an avian ovomucoid promoter
Another potentially useful application of the novel isolated ovomucoid gene expression control region of the present invention is the possibility of increasing the amount of a heterologous protein present in a bird, (especially the chicken) by gene transfer. In most instances, a heterologous polypeptide-encoding nucleic acid insert transferred into the recipient animal host will be operably linked with the ovomucoid gene expression control region to allow the cell to initiate and continue production of the genetic product protein. A recombinant DNA molecule of the present invention can be transferred into the extra-chromosomal or genomic DNA of the host.
The recombinant DNA nucleic acid molecules of the present invention can be delivered to cells using conventional recombinant DNA technology. The recombinant DNA molecule may be inserted into a cell to which the polypeptide-encoding nucleic acid insert of the recombinant DNA molecule is heterologous (i.e. not normally present). Alternatively, as described more fully below, the recombinant DNA molecule may be introduced into cells which normally contain the polypeptide-encoding nucleic acid insert of the recombinant DNA molecule, for example, to correct a deficiency in the expression of a polypeptide, or where over-expression of the polypeptide is desired.
For expression in heterologous systems, the heterologous DNA molecule is inserted into the expression system or vector of the present invention in proper sense orientation and correct reading frame. The vector contains the necessary elements for the transcription and translation of the inserted protein-coding sequences, including the novel isolated ovomucoid gene expression control region.
U.S. Patent No. 4,237,224 to Cohen & Boyer, which is hereby incorporated by reference in its entirety, describes the production of expression systems in the form of recombinant plasmids using restriction enzyme cleavage and ligation with DNA ligase. These recombinant plasmids are then introduced to a cell by means of transformation and replicated in cultures, including eukaryotic cells grown in tissue culture.
One aspect of the present invention, therefore, is an expression vector suitable for delivery to a recipient cell for replication OR expression of a polypeptide-encoding nucleic acid of the vector therein. It is contemplated to be within the scope of the present invention for the expression vector to comprise an isolated avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a polypeptide, and optionally a polyadenylation signal sequence. The expression vector of the present invention may further comprise a bacterial plasmid sequence, a viral nucleic acid sequence, or fragments or variants thereof that may allow for replication of the vector in a suitable host.
The novel isolated avian ovomucoid gene expression control region of the present invention (SEQ ID NO: 26) and a polypeptide-encoding nucleic acid sequence operably linked thereto and optionally a polyadenylation signal sequence may be introduced into viruses such as vaccinia virus. Methods for making a viral recombinant vector useful for expressing a protein under the control of the ovomucoid promoter are analogous to the methods disclosed in U.S. Patent Nos. 4,603,112; 4,769,330; 5,174,993; 5,505,941; 5,338,683; 5,494,807; 4,722,848; Paoletti, E. Proc. Natl. Acad. Sci. 93: 11349-11353 (1996); Moss Proc. Natl. Acad. Sci. 93: 11341-11348 (1996); Roizman Proc. Natl. Acad. Sci. 93: 11307-11302 (1996); Frolov et al. Proc. Natl. Acad. Sci. 93: 11371-11377 (1996); Grunhaus et al. Seminars in Virology 3: 237-252 (1993) and U.S. Patent Nos. 5,591,639; 5,589,466; and 5,580,859 relating to DNA expression vectors, inter alia; the contents of which are incorporated herein by reference in their entireties.
Recombinant viruses can also be generated by transfection of plasmids into cells infected with virus. Suitable vectors include, but are not limited to, viral vectors such as lambda vector system λgtl 1, λgt WES.tB, Charon 4, and plasmid vectors such as pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pKC37, pKClOl, SV 40, pBluescript II SK +/- or KS +/- (see "Stratagene Cloning Systems" Catalog (1993) from Stratagene, La Jolla, Calif, which is hereby incorporated by reference), pQE, pIH821, pGEX, pET series (see Studier, F. W. et. al. (1990) "Use of T7 RNA Polymerase to Direct Expression of Cloned Genes" Gene Expression Technology, vol. 185, which is hereby incorporated by reference in its entirety) and any derivatives thereof, cosmid vectors and, in preferred embodiments, artificial chromosomes, such as, but not limited to, YACs, BACs, BBPACs or PACs. Such artificial chromosomes are useful in that a large nucleic acid insert can be propagated and introduced into the avian cell. Recombinant molecules can be introduced into cells via transformation, particularly transduction, conjugation, mobilization, or electroporation. The DNA sequences are cloned into the vector using standard cloning procedures in the art, as described by Sambrook et al. Molecular Cloning: A Laboratory Manual, 3rd ed., Cold Spring Harbor Laboratory, Cold Springs Harbor, N.Y. (2001), which is hereby incorporated by reference in its entirety.
The vectors of the invention comprise one or more nucleotide sequences encoding a heterologous protein desired to be expressed in the transgenic avian, as well as regulatory elements such as promoters, enhancers, Matrix Attachment Regions, IRES's and other translation control elements, transcriptional termination elements, polyadenylation sequences, etc. In particular embodiments, the vector of the invention contains at least two nucleotide sequences coding for heterologous proteins, for example, but not limited to, the heavy and light chains of an immunoglobulin.
The present invention further relates to nucleic acid vectors and transgenes inserted therein, having the avian ovomucoid gene expression control region of the invention, that incorporate multiple polypeptide-encoding regions, wherein a first polypeptide-encoding region is operatively linked to a transcription promoter and a second polypeptide-encoding region is operatively linked to an IRES. For example, the vector may contain coding sequences for two different heterologous proteins (e.g., the heavy and light chains of an immunoglobulin). Such nucleic acid constructs, when inserted into the genome of a bird and expressed therein, will generate individual polypeptides that may be post-translationally modified, for example, glycosylated or, in certain embodiments, form complexes, such as heterodimers with each other in the white of the avian egg. Alternatively, the expressed polypeptides may be isolated from an avian egg and combined in vitro, or expressed in a non-reproductive tissue such as serum. In other embodiments, for example, but not limited to, when expression of both heavy and light chains of an antibody is desired, two separate constructs, each containing a coding sequence for one of the heterologous proteins operably linked to the ovomucoid gene expression control region of the invention are introduced into the avian cell. Alternatively, two transgenic avians each containing one of the two heterologous proteins (e.g., one transgenic avian having a transgene encoding the light chain of an antibody and a second transgenic avian having a transgene encoding the heavy chain of the antibody) can be bred to obtain an avian containing both transgenes in its germline and expressing both transgene encoded proteins, preferably in eggs. Once the ovomucoid gene expression control region of the present invention has been cloned into a vector system, it is ready to be incorporated into a host cell. Such incorporation can be carried out by the various forms of transformation noted above, depending upon the vector/host cell system. Suitable host cells include, but are not limited to, bacteria, virus, yeast, mammalian or avian cells, and the like. Alternatively, it is contemplated that the incorporation of the DNA of the present invention into a recipient cell may be by any suitable method such as, but not limited to, viral transfer, electroporation, gene gun insertion, sperm mediated transfer to an ovum, microinjection, cytoplasmic injection, pronuclear injection and the like.
Another aspect of the present invention, therefore, is a method of expressing a heterologous polypeptide in a eukaryotic cell by transfecting the cell with a recombinant DNA comprising an avian ovomucoid gene expression control region operably linked to a nucleic acid insert encoding a polypeptide and, optionally, a polyadenylation signal sequence, and culturing the transfected cell in a medium suitable for expression of the heterologous polypeptide under the control of the avian ovomucoid gene expression control region. In preferred embodiments, the ovomucoid gene expression control region directs a level of expression of the heterologous protein in avian eggs that is greater than 5 μg, 10 μg, 50 μg, 100 μg, 250 μg, 500 μg, or 750 μg, more preferably greater than 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, 50 mg, 100 mg, 200 mg, 500 mg, 700 mg, 1 gram, 2 grams, 3 grams, 4 grams or 5 grams per egg. Such levels of expression can be obtained using the expression control regions of the invention.
In one embodiment of the method of the present invention, the recipient eukaryotic cell is derived from an avian. In one embodiment, the avian is a chicken.
Yet another aspect of the present invention is a eukaryotic cell transformed with an expression vector according to the present invention and described above. In one embodiment of the present invention, the transformed cell is a chicken oviduct cell and the nucleic acid insert comprises the chicken ovomucoid gene expression control region, a nucleic acid insert encoding a human interferon α2d with codons optimized for expression in an avian cell, and an SV40 polyadenylation sequence.
It is contemplated that the transfected cell according to the present invention may be transiently transfected, whereby the transfected recombinant DNA or expression vector may not be integrated into the genomic nucleic acid. It is further contemplated that the transfected recombinant DNA or expression vector may be stably integrated into the genomic DNA of the recipient cell, thereby replicating with the cell so that each daughter cell receives a copy of the transfected nucleic acid. It is still further contemplated for the scope of the present invention to include a transgenic animal producing a heterologous protein expressed from a transfected nucleic acid according to the present invention.
In one embodiment of the present invention, the transgenic animal is an avian selected from a turkey, duck, goose, quail, pheasant, ratite, an ornamental bird or a feral bird. In another embodiment, the avian is a chicken and the heterologous protein produced under the transcriptional control of the isolated avian ovomucoid gene expression control region according to the present invention is produced in the white of an egg.
Viral vector cell transformation An exemplary approach for the in vivo introduction of a polypeptide-encoding nucleic acid operably linked to the subject novel isolated ovomucoid gene expression control region into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA, encoding the gene product. Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells that have taken up viral vector nucleic acid.
Retrovirus vectors and adeno-associated virus vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host. Recombinant retrovirus can be constructed in the part of the retro viral coding sequence (gag, pol, env) that has been replaced by nucleic acid comprising a ovomucoid gene expression control region, thereby rendering the retrovirus replication defective. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel et al. (1989) (eds.) Greene Publishing Associates, Sections 9.10-9.14 and other standard laboratory manuals. Examples of suitable retroviruses include pLJ, pZIP, pWE and pEM which are all well known to those skilled in the art. Examples of suitable packaging virus lines for preparing both ecotropic and amphotropic retroviral systems include psiCrip, psiCre, psi2 and psiAm.
Furthermore, it is possible to limit the infection spectrum of retroviruses and consequently of retroviral-based vectors, by modifying the viral packaging proteins on the surface of the viral particle (see, for example PCT publications WO93/25234, WO94/06920, and WO94/11524). For instance, strategies for the modification of the infection spectrum of retroviral vectors include coupling antibodies specific for cell surface antigens to the viral env protein (Roux et al., Proc. Natl. Acad. Sci. 86: 9079-9083 (1989); Julan et al, J. Gen. Virol. 73: 3251-3255 (1992); and Goud et al, Virology 163: 251-254 (1983)) or coupling cell surface ligands to the viral env proteins (Neda et al, J. Biol. Chem. 266: 14143-14146 (1991)), all of which are incorporated herein by reference in their entireties. Coupling can be in the form of the chemical cross-linking with a protein or other moiety (e.g. lactose to convert the env protein to an asialoglycoprotein), as well as by generating fusion proteins (e.g. single-chain antibody/env fusion proteins). This technique, while useful to limit or otherwise direct the infection to certain tissue types, can also be used to convert an ecotropic vector into an amphotropic vector.
Another viral gene delivery system useful in the present invention utilizes adenovirus-derived vectors. The genome of an adenovirus can be manipulated such that it encodes a gene product of interest, but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle (see, for example, Berkner et al, BioTechniques 6: 616 (1988); Rosenfeld et al, Science 252: 43 1434 (1991); and Rosenfeld et al, Cell 68: 143-155 (1992)), all of which are incorporated herein by reference in their entireties. Suitable adeno viral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are well known to those skilled in the art. The virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity. Additionally, introduced adeno viral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA). Most replication-defective adenoviral vectors currently in use and therefore favored by the present invention are deleted for all or parts of the viral El and E3 genes but retain as much as 80% of the adenoviral genetic material (see, e.g., Jones et al, Cell 16:683 (1979); Berkner et al, supra; and Graham et al, in Methods in Molecular Biology, E. J. Murray, (1991) Ed. (Humana, Clifton, N.J.) vol. 7. pp. 109-127), all of which are incorporated herein by reference in their entireties. Expression of an inserted gene such as, for example, encoding the human interferon α2b, can be under control of the exogenously added ovomucoid gene expression control region sequences. Yet another viral vector system useful for delivery of, for example, the subject avian ovomucoid gene expression control region operably linked to a nucleic acid encoding a polypeptide, is the adeno-associated virus (AAN). Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate. Space for exogenous DΝA is limited to
5 about 4.5 kb. An AAV vector such as that described in Tratschin et al, Mol. Cell. Biol. 5:3251-3260 (1985) can be used to introduce DΝA into cells. A variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et al., Proc. Νatl. Acad. Sci. 81:6466-6470 (1984); Tratschin et al, Mol. Cell. Biol. 4:2072-2081 (1985); Wondisford et al, Mol. Endocrinol. 2:32-39 (1988); Tratschin et al, J. Virol.
10 51 :611-619 (1984); and Flotte et αl., J. Biol. Chem. 268:3781-3790 (1993)), all of which are incorporated herein by reference in their entireties.
Non-virαl expression vectors
Most non-viral methods of gene transfer rely on normal mechanisms used by
15 eukaryotic cells for the uptake and intracellular transport of macromolecules. In preferred embodiments, non- viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject ovomucoid gene expression control region and operably linked polypeptide-encoding nucleic acid by the targeted cell. Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and 0 artificial viral envelopes.
In a representative embodiment, a nucleic acid comprising the novel isolated ovomucoid gene expression control region of the present invention can be entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et αl., NO 5 Shinkei Geka 20:547-551 (1992); PCT publication WO91/06309; Japanese patent application 1047381 ; and European patent publication EP-A-43075), all of which are incorporated herein by reference in their entireties.
In similar fashion, the gene delivery system comprises an antibody or cell surface ligand that is cross-linked with a gene binding agent such as polylysine (see, for example, 0 PCT publications WO93/04701, WO92/22635, WO92/20316, WO92/19749, and
WO92/06180), all of which are incorporated herein by reference in their entireties. It will also be appreciated that effective delivery of the subject nucleic acid constructs via receptor-mediated endocytosis can be improved using agents which enhance escape of gene from the endosomal structures. For instance, whole adenovirus or fusogenic peptides of the 5 influenza HA gene product can be used as part of the delivery system to induce efficient disruption of DNA-containing endosomes (Mulligan et al, Science 260: 926 (1993); Wagner et al, Proc. Natl. Acad. Sci. 89:7934 (1992); and Christiano et al, Proc. Natl. Acad. Sci. 90:2122 (1993)), all of which are incorporated herein by reference in their entireties. It is further contemplated that a recombinant DNA molecule comprising the novel isolated ovomucoid gene expression control region of the present invention may be delivered to a recipient host cell by other non- viral methods including by gene gun, microinjection, sperm-mediated transfer as described in PCT/US02/30156, filed September 23, 2002 and incorporated herein by reference in its entirely, nuclear transfer, or the like.
Transgenic animals
Another aspect of the present invention concerns transgenic animals, such as chickens that contain a transgene comprising the novel isolated ovomucoid gene expression control region of the present invention and which preferably (though optionally) express a heterologous gene in one or more cells in the animal. Suitable methods for the generation of transgenic avians having heterologous DNA incorporated therein, for example, cytoplasmic injection and pronuclear injection, are described, for example, in U.S. Patent Application No: 10/251,364, entitled "Methods of Generating Transgenic Avians Using Microinjection into the Cytoplasm of an Avian Egg or Embryo", by Rapp et al., filed September 18, 2002, and incorporated herein by reference in its entirety.
In various embodiments of the present invention, the expression of the transgene may be restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences acting on the ovomucoid gene expression control region of the present invention and which control gene expression in the desired pattern. Tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial patterns. Moreover, temporal patterns of expression can be provided by, for example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
One embodiment of the present invention, therefore, is a transgenic avian having a heterologous polynucleotide sequence comprising a nucleic acid insert encoding the heterologous polypeptide and operably linked to the novel isolated avian ovomucoid gene expression control region. In an embodiment of the present invention, the transgenic avian is selected from a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird. In another embodiment of the present invention, the transgenic avian is a chicken. In still another embodiment of the transgenic avian of the present invention, the transgenic avian includes an avian ovomucoid gene expression control region comprising the nucleic acid sequence in SEQ ID NO: 26, or a degenerate variant thereof. In yet another embodiment of the transgenic avian of the present invention, the transgenic avian further comprises a polyadenylation signal sequence.
In still yet another embodiment of the transgenic avian of the present invention, the polyadenylation signal sequence is derived from the SV40 virus.
In another embodiment of the transgenic avian of the present invention, the nucleic acid insert encoding a polypeptide has a codon complement optimized for protein expression in an avian.
In another embodiment of the transgenic avian of the present invention, the transgenic avian produces the heterologous polypeptide in the serum or an egg white. In another embodiment of the transgenic avian of the present invention, the transgenic avian produces the heterologous polypeptide in an egg white.
The present invention is further illustrated by the following examples, which are provided by way of illustration and should not be construed as limiting. The contents of all references, published patents and patents cited throughout the present application are hereby incorporated by reference in their entireties.
Example 1 : PCR amplification of Ovomucoid promoter
Sense primer OVINs2, 5'-TAGGCAGAGCAATAGGACTCTCAACCTCGT-3' (SEQ ID NO: 1) and the antisense primer, OVMUa2, 5'-AAGCTTCTGCAGCACTCTGGGAGTTACTCA-3' (SEQ ID NO: 2) were designed according to the sequences of chick ovoinhibitor exon 16 (Genbank Accession No: M16141) and a fragment of the chick ovomucoid promoter region (Genbank Accession No: J00897) respectively. The template DNA for PCR amplification of the ovomucoid promoter region was prepared from white leghorn chick blood. A series of different PCR conditions were carried out to optimize synthesis of the approximately 10.0 kb product, the results of which are shown in FIG. 2. In these tests, the template DNA concentrations were 500 ng, 100 ng, 50 ng, or 10 ng. Two sets of primers, OVINsl (SEQ ID NO: 3) and OVMUal (SEQ ID NO: 4), or OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) shown in FIG. 3, three Mg^ concentrations (1.0 mM, 1.5 mM and 2.0 mM) and annealing temperatures from 50° C to 70° C were used. The results of the tests were as shown in FIG. 2. As shown in lanes 1 through 8, test reactions having 500 ng DNA template, the OVI sl (SEQ ID NO: 3) and OVMUal (SEQ ID NO: 4) primers, 60 mM Tris-SO4, pH9.1, 18 mM (NH4)2SO4, 1.0 mM Mg2*, and annealing temperatures between 50°C to 58°C gave no specific DNA product. Also, as shown in lanes 17 through 24 of FIG. 2, in test reactions having 100 ng DNA template, the OVINsl and OVMUal primers, 60 mM Tris-SO4, pH9.1, 18 mM (NH4)2SO4, 1.0 mM Mg2*, and annealing temperatures between 50°C to 58°C, no specific bands were seen. However, as shown in lanes 9 through 16 of FIG. 2, test reactions having 500 ng DNA template, the OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) primers, 60 mM Tris-SO4, pH9.1, 18 mM
(NH4)2SO4, 2 mM Mg2+ and annealing temperatures between 60°C to 68°C have the band of the desired length of approximately 10 kb. As shown in lanes 25 through 32, reaction conditions containing 100 ng DNA template, the OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) primers, 60 mM Tris-SO4, pH9.1, 18 mM (NH4)2SO4, 2mM Mg2+ and annealing temperatures between about 60 °C to about 68 °C gave an increased yield of the desired product.
An approximately 10 kb product was, therefore, detected when the following conditions were used: the optimum DNA template concentration was between about 50 ng to 500 ng; the primers were OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2); the Mg2+ concentration was 2 mM; the annealing temperature was at or between about 60 °C to about 68°C. Each 50 μl PCR reaction consisted of 50 ng or 100 ng of template DNA, 0.1 μg each primer, 5 μl buffer B (from Elongase Enzyme Mix kit, Invitrogen Corp., Carlsbad, CA), 1 ml of 10 μM dNTP solution, and distilled deionized water. The PCR protocol was one cycle at 94°C for 30 sees; thirty cycles at 94°C for 30 sees, 60 °C for 30 sees and 68 °C for 10 mins. One cycle was performed at 68 °C for 10 mins, 35 °C for 30 mins with a final hold at 4°C. The PCR products were examined by 0.65% agarose gel analysis.
Example 2: Cloning of PCR products.
The PCR products were purified by standard methods. Briefly, PCI (phenol: chloroform: isoamyl alcohol, 24:25:1) and chloroform extraction were performed once. The DNA was precipitated by adding 3M sodium acetate pH5.2 to a final concentration of 0.3M together with 2.5 volumes of 100% ethanol. The DNA pellet was dried and dissolved in distilled deionized water and then sequenced on a ABI3700 automatic sequencer (Applied Biosystems, Foster City, CA) using the primers OVINs2 (SEQ ID NO: 1) and OVMUa2 (SEQ ID NO: 2) to confirm the identity of each PCR product. After confirmation of the identities, the approximately 10 kb PCR product was treated with T4 polynucleotide kinase to add a phosphate to the 5' end. Mung bean nuclease removed any overhanging adenines from the ends of the PCR products, thereby producing a blunt end. The PCR product was purified by PCI and chloroform extraction and precipitated by standard methods. This 10 kb product was then cleaved with Bam HI to give two fragments, of about 4.7 and about 5.5 kb respectively.
The vector plasmid pBluescript II KS (+/-) was cut by Bam HI and Eco RV and treated with calf intestinal alkaline phosphatase. DNA fragments to be ligated into the vector were analyzed by agarose gel electrophoresis and purified from agarose gel slices using a NucleoTrap Nucleic Acid Purification Kit (BD Biosciences Clontech, Palo Alto, CA). Fragments of 4.7 kb and 5.5 kb were inserted into the Bam HVEco RV-treated pBluescript to give the constructs pBS-OVMUP4.7 and pBS-OVMUP5.5 respectively.
Positive clones were screened by Xba VXho I digestion. Clone pBS-OVMUP4.7, gave fragments of about 4.7 kb and 2.96 kb. Clone pBS-OVMUP5.5 gave fragments of about 5.5 kb and 2.96 kb. Apparent positive clones having the 4.7 kb insert were further confirmed by Xba VHind HI digestion that gave three fragments of 0.5 kb, 4.2 kb and 2.9 kb. The apparent positive clones with an insert of about 5.5 kb insert were further confirmed by Xba VKpn I digestion that gave three fragments of 2 kb, 3.5 kb and 2.96 kb. A construct, pBS-OVMUP-10, containing the entire 10 kb PCR product cloned into the pBluescript KS II (+/-) vector was made by taking a 4.7 kb Bam Hl/Xho I fragment from the pBS-OVMUP4.7 plasmid and inserting it into the Bam ΗUXba I cleaved sites of pBS-OVMUP5.5. The Xho I and Xba I cut ends were blunt-ended by treating the digested fragments with Klenow enzyme and dNTPs at 25 °C for 15 mins before the digestion with Bam HI.
Example 3: Sequencing
The plasmids pBS-OVMUP4.7 and pBS-OVMUP5.5 were sequenced from both ends of each insert as shown in FIG. 1. The initial primers were T7 and T3 having the nucleic acid sequences 5'-TAATACGACTCACTATAGGG-3' (SEQ ID NO: 5) and
5'-ATTAACCCTCACTAAAGGGA-3* (SEQ ID NO: 6) respectively. Subsequent primers (SEQ ID NOS: 7 - 25), as shown in FIG. 3, were designed according to the sequence results as they became available. The approximately 10 kb sequence was edited and assembled by the ContigExpress software of the Vector NTI Suite, version 6.0 (InforMax, Inc.). The region of the approximately 10 kb PCR product described in Example 1 above that encompassed the Bam HI junction was sequenced using the primers OVMUa9 (SEQ ID NO 27) and OVINs9 (SEQ ID NO 28) (shown in FIG. 3).
Each sequence chromatogram was visually checked for sequence accuracy and to
5 locate base ambiguities. Regions containing ambiguous bases were re-sequenced with the same primer or, if still ambiguous, with a new primer designed to sequence the complementary strand. Sequencing of the original 10 kb PCR fragment using the primers OVMUa9 (SEQ ID NO 27) and OVINs9 (SEQ ID NO 28) showed that the subcloned inserts of the plasmids pBS-OVMUP4.7 and pBS-OVMUP5.5 included all of the nucleic acid
10 sequence of the parent fragment and no intervening Bam HI - Bam HI fragments were included in the final sequence SEQ ID NO: 26. The sequence (SEQ DD NO: 26) of the region lying between the 3' end of the ovoinhibitor gene and the transcription start site of the ovomucoid-encoding region is shown in FIG. 4.
15 Example 4: Expression in Transfected Cultured Avian Myeloid and Oviduct Cells of luciferase Regulated by the 10 kb ovomucoid Promoter Construction of plO-OM-luc
To facilitate insertion of coding sequences behind the ovomucoid promoter and in frame with the second ATG of the ovomucoid coding sequence, the Nco I site which overlaps 20 the second ATG was changed to a Pci I site as depicted below. On the top is the wild type ovomucoid sequence at the start site of translation. On the bottom, the second Nco I site was changed to a Pci I site.
Nco I Nco I
25 MetAlaMet
CTCACCATGGCCATGGC (SEQ ID NO: 32) GAGTGGTACCGGTACCG (SEQ ID NO: 33)
Nco I Pci I
MetAspMet
30 CTCACCAATTGGGGAACCAATTGGCGA ( SEQ ID NO : 34 )
GAGTGGTACCGGTACCG (SEQ ID NO: 35)
The Pci I site in the Bluescript backbone of pBS-OVMUP-10 was destroyed by cutting with Pci I, filling in the ends with Klenow polymerase and religating, creating pOM- 35 10-alpha. The proximal promoter region was PCR amplified with primers OM-5 (SEQ ID NO.:29) and OM-6 (SEQ ID NO.:30) and template pBS-ONMUP-10. The resulting PCR product (SEQ ID ΝO.:31) was cut with Not I and TthU 1 1 and cloned into the 12059 bp Not
\-Tth\ 11 1 fragment of pOM-10-alpha, thereby creating pOM-10-Pci. The 1964 Nco I-Sl- treated Kpn I frament of gWiz-luciferase (Gene Therapy Systems, Inc., San Diego, California) was cloned into the 12824 Pci l-Sma I fragment of pOM-10-Pci, creating plO-OM-luc.
Primer sequences
OM-5 5'-GCGCGGCCGCCCGGGACATGTCCATGGTGAGAGTACTGCCC-3' (SEQ
ID NO: 29) OM-6 5'-GGCCCGGGATTCGCTTAACTGTGACTAGG-3' (SEQ ID NO:30)
PCR product
GCGCGGCCGCCCGGGACATGTCCATGGTGAGAGTACTGCCCGGCTCTGCAGGCGGCTGCCGG TGCTCTGCTCCTGAGATGGTCCCCCCGAGGCTGCCTGCAAATATATACAAACGTGGCGTCCG AACTGTTGGACTGGAACACGGAGCAGCCAGCTGAATCTGTCAGCGGCACAATGAGGCTGGTA ATATTTATTGAGGTCCTGACCTCCAGGTAATGGTCTGCGTCTCCCAGGCAATTGATTTTGGC TGGACACTTGGTTAATAGCTTGAGACAAGTGTCACATGCTCTCAGTGGTCAAAACCAAACAA ACAGACTTTTGGACCAAAAAAAAAAAAAACCTCTTAAGGACTCTGGTAGAACCCTAAATAGC ACAGAATGCTGAGGGGAGTAAGGGACAGGTCCTTCATTCGTCTCTGCATCCACATCTCCCAG CAGGAAGCAGCTAAGGCTCAGCACCATCGTGCCTGCAGCTCTGCTTTCCATGCAGTTCTGCA T CTTGGATATTCACCTCTAGGTAAAAGCACAGGCCAGGGAGGCTTTGTCACCAGCAGAACT GACCAACCACTGCCAGGTGAAGCTGGCAGCACCGTATCTAACCTATGAAGTTAATGGTATTT AGCACTAGCTTGATAAAAGGAAGGGTTTCTTGGCGGTTTCACTGCTTAAGTATAGAAGAGCT TGGTAGAAGACTTGAAAGCAAGGTAAATGCTGTCAAATACCACTAAAAATGTCACTTGAACC TTATCAGCAGGGAGCACTTATTTACAGACCTAGTCACAGTTAAGCGAATTCCCGGGCC(SEQ IDNO:31)
The 1st and 2nd ATGs of the ovomucoid sequence are shown underlined. Note that the ovomucoid coding sequence is in reverse. The underlined, bold A is not in the wildtype sequence but was incorporated into pOM-10-Pci due to a error in the oligo OM-5.
Expression of luciferase
For expression in avian cells of non-magnum origin, HDl 1 cells, a chicken myeloid cell line was used. Cells were cultured as described in Beug, H., et al. (Chicken hematopoietic cells transformed by seven strains of defective avian leukemia viruses display three distinct phenotypes of differentiation. (1979) Cell, 18: 375-90, in which these cells were referred to as HBCI cells), herein incorporated by reference in its entirety. Plasmid DNA was transfected into HDl 1 cells with Lipofectamine 2000 (Invitrogen Corporation, Carlsbad, California) according to the manufacturer's instructions.
48 hours post-transfection, the cells were harvested and pelleted. The supernatant was removed and 20 μl of 10 mM Tris, pH 7.8, 1 mM EDTA (TE) was added. The cells were frozen at -80°C and thawed. 5 μl of the cell suspension was mixed with 25 μl of Bright- Glo™ reagent (Bright-Glo™ Luciferase Assay System, Promega, Madison, WI) and relative light units per second measured on a Berthold Detection Systems (Oak Ridge, TN) FBI 2 luminometer. Results are depicted in FIG. 6 A. HDl 1 cells are permissive for the CMN promoter and should be able to only weakly activate the ovomucoid promoter. Some expression of the luciferase gene linked to the 10 kb ovomucoid is evident.
For expression in avian oviduct cells, primary tubular gland cells were isolated as follows. The oviduct of a Japanese quail (Coturnix coturnixjaponica) was removed and the magnum portion minced and enzymatically dissociated with 0.8 mg/ml collagenase (Sigma Chemical Co., St. Louis, MO) and 1.0 mg/ml dispase (Roche Molecular Biochemicals, Indianapolis, IN) by shaking and titurating for 30 minutes at 37°C. The cell suspension was then filtered through sterile surgical gauze, washed three times with F-12 medium (Life Technologies, Grand Island, NY) by centrifugation at 200 x g, and resuspended in OPTEVΪEM™ (Life Technologies) such that the OD600 was approximately 2. 800 μl of the cell suspension was plated in each well of a 6-well dish. For each transfection, 4.0 μl of
DMRJJE-C liposomes (Life Technologies) and 2.0 μg of plasmid DNA was preincubated for
15 minutes at room temperature in 200 μl of OPTIMEM™, and then added to the oviduct cells. Cells with DNA/liposomes were incubated for about 5 hours at 37°C in 5% CO2. Next,
2.0 ml of DMEM (Life Technologres), supplemented with 15% fetal bovine serum (FBS)
(Atlanta Biologicals, Atlanta, GA), 2X penicillin/streptomycin (Life Technologies), 50 ng/ml insulin (Sigma), 10"7 M α-estradiol (Sigma), and 10'6 M corticosterone (Sigma) were added to each well, and incubation continued for about 40 hours. Medium was then harvested and centrifuged at 110 x g for 5 minutes.
For quantitation, the cells were scraped into the media with a rubber policeman. One milliliter was transferred to an eppendorf tube and the cells pelleted. The supernatant was removed and 20 μl of 10 mM Tris, ph 7.8, 1 mM EDTA (TE) was added. The cells were frozen at -80°C and thawed. 5 μl of the cell suspension was mixed with 25 μl of Bright- Glo™ reagent (Bright-Glo™ Luciferase Assay System, Promega, Madison, WI) and relative light units per second measured on a Berthold Detection Systems (Oak Ridge, TN) FBI 2 luminometer.
The results are depicted in FIG. 6B. Expression of luciferase is evident from the CMV and 10 kb ovomucoid promoters. The ovomucoid promoter has more activity relative to the CMV promoter in the tubular gland cells (ratio of CMV to ovomucoid is 152) than in the HDl 1 cells (ratio of CMV to ovomucoid is 2221).
Example 5: Expression in Transfected Cultured Avian Oviduct Cells of Human Interferon α2b Regulated by the 10 kb ovomucoid Promoter Construction of plO-OM-IFN
The CMV promoter region of pAVIJCR-A137.91.1.2 flanked by Nco I sites (pCMV- human IFN-alpha-2b-MagMax) was replaced with the 1051 bp Nco Ϊ-Nco I fragment from pBS-OVMUP-4.4, thereby inserting the 1 kb ovomucoid promoter in front of the IFN coding sequence and SV40 polyadenylation signal and creating plkb-OM-IFNMM. A 1816 bp Cla I- Sac I fragment of plkb-OM-IFNMM was inserted into the 6245 bp Cla l-Sac I fragment of pBS-OVMUP-4.4, thereby fusing the 4.4 kb ovomucoid fragment with the IFN coding sequence and creating p4.4OM-IFNMM. The 8511 bp BamH I-Sal I fragment of pBS- OVMUP-10 was ligated to the 5148 bp BamH l-Sal I fragment of p4.4OM-IFN, thereby placing the 10 kb ovomucoid promoter in front of the IFN coding sequence, creating plO- OM-EFN.
Expression of interferon Quail primary tubular gland cells were isolated and treated as described in Example 4.
100 μl of supernatants were analyzed by ELISA (PBL Biomedical Laboratories, Flanders, NJ) for human interferon α2b content. The results are depicted in FIG. 7. Expression of interferon is evident from the CMV and 10 kb ovomucoid promoters.
All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.
The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.
Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention is to be limited only by the terms of the appended claims along with the full scope of equivalents to which such claims are entitled.
- 43.1 -
International Application No: PCT/ /
MICROORGANISMS
Optional Sheet in connection with the microorganism referred to on pages Q , lines 34 and 35 of the description '
A. IDENTIFICATION OF DEPOSIT '
Further deposits are identified on an additional sheet
Name of depositary institution ' American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Blvd. Manassas, VA 201 10-2209 US
Date of deposit ' November 26, 2002 Accession Number ' _
B. ADDITIONAL INDICATIONS ' (leave blank if not applicable) This information is continued on a separate attached sheet
C. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE ' ι ,h.,„ ,c„m m no, di d,,=d 1,
D. SEPARATE FURNISHING OF INDICATIONS ' (leave blank if not applicable)
The indications listed below will be submitted to the International Bureau later ' {Specify the general nature of the indications e g , Accession Number of Deposit' )
E. D This sheet was received with the International application when filed (to be checked by the receiving Office)
(Authorized Officer)
D The date of receipt (from the applicant) by the International Bureau "
(Authorized Officer) Form PCT/RO/134 (January 1981 )

Claims

What is Claimed Is:
1. An isolated nucleic acid comprising an avian ovomucoid gene expression control 5 region comprising the nucleic acid sequence of SEQ ID NO: 26.
2. The nucleic acid of Claim 1, wherein the avian is selected from the group consisting of a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird.
10
3. The nucleic acid of Claim 1, wherein the avian is a chicken.
4. An isolated nucleic acid comprising a sequence at least 75% identical to SEQ ID NO: 26 and directs expression of an operably linked nucleotide sequence in an avian
15 oviduct cell.
5. The nucleic acid of Claim 4 comprising a sequence at least 95% identical to SEQ ID NO: 26.
20 6. The nucleic acid of Claim 4 comprising a sequence at least 99% identical to SEQ ID NO: 26.
7. A nucleic acid comprising an avian ovomucoid gene expression control region operably linked to a nucleotide sequence encoding a heterologous polypeptide.
25
8. The nucleic acid of Claim 7, wherein the avian is selected from the group consisting of a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird.
30 9. The nucleic acid of Claim 7, wherein the avian is a chicken.
10. The nucleic acid of Claim 7, wherein the ovomucoid gene expression control region comprises the nucleic acid sequence in SEQ ID NO: 26.
35 11. The nucleic acid of Claim 7, further comprising a polyadenylation signal sequence.
- 44 -
12. The nucleic acid of Claim 11, wherein the polyadenylation signal sequence is derived from the SV40 virus.
5 13. The nucleic acid of Claim 7, wherein the nucleotide sequence has a codon complement optimized for protein expression in an avian.
14. The nucleic acid of Claim 13, wherein said polypeptide is interferon α2b polypeptide.
10 15. The nucleic acid of Claim 7, wherein the nucleic acid insert encodes an immunoglobulin subunit.
16. The nucleic acid of Claim 7 that comprises an origin of replication selected from a bacterial origin of replication or a viral origin of replication.
15
17. The nucleic acid of Claim 16 which is a plasmid.
18. The nucleic acid of Claim 16 which is an eukaryotic viral vector.
20 19. The nucleic acid of claim 7 which is an expression vector that integrates into a host cell.
20. A method of expressing a heterologous polypeptide in a host cell, said method comprising culturing a eukaryotic cell transfected with a nucleic acid molecule 25 comprising an avian ovomucoid gene expression control region operably linked to a nucleotide sequence encoding the heterologous polypeptide, or progeny of said eukaryotic cell, in a medium suitable for expression of said heterologous polypeptide, thereby expressing said heterologous polypeptide.
30 21. The method of Claim 20, wherein the eukaryotic cell is derived from an avian.
22. The method of Claim 21, wherein the eukaryotic cell is derived from a chicken.
23. A eukaryotic cell transformed with the expression vector according to Claim 19, or a 35 progeny of the cell, wherein the cell or the progeny thereof expresses a heterologous
- 45 - polypeptide.
24. The eukaryotic cell of Claim 23, wherein the cell is an avian cell. 5
25. The eukaryotic cell of Claim 23, wherein the cell is an chicken cell.
26. The eukaryotic cell of Claim 23, wherein the cell is an oviduct cell of a chicken.
10 27. The eukaryotic cell of Claim 23, wherein the cell is an oviduct cell of a quail.
28. The eukaryotic cell of Claim 23, wherein the cell is a cultured cell.
29. The eukaryotic cell of Claim 23, wherein the nucleotide sequence encoding said
15 heterologous polypeptide has a codon complement optimized for protein expression in an avian.
30. The eukaryotic cell of Claim 29, wherein the nucleotide sequence encodes an interferon α-2b polypeptide.
20
31. The eukaryotic cell of claim 23, wherein the nucleotide sequence encodes an immunoglobulin or a subunit thereof.
32. A transgenic avian comprising a nucleic acid comprising an avian ovomucoid gene 5 expression control region operably linked to a nucleotide sequence encoding a heterologous polypeptide.
33. The transgenic avian of Claim 32, wherein the avian is a chicken, a turkey, a duck, a goose, a quail, a pheasant, a ratite, an ornamental bird or a feral bird.
30
34. The transgenic avian of Claim 32, wherein the avian is a chicken.
35. The transgenic avian of Claim 32, wherein the ovomucoid gene expression control region comprises the nucleic acid sequence of SEQ ID NO: 26. 5
- 46 -
36. The transgenic avian of Claim 32, wherein the nucleic acid comprises a polyadenylation signal sequence.
5 37. The transgenic avian of Claim 36, wherein the polyadenylation signal sequence is derived from the SV40 virus.
38. The transgenic avian of Claim 32, wherein the nucleotide sequence has a codon complement optimized for protein expression in an avian.
10
39. The transgenic avian of Claim 32, wherein the nucleic acid insert encodes an interferon α2b polypeptide.
40. The transgenic avian of Claim 32, wherein the transgenic avian produces the 15 heterologous polypeptide in the serum or an egg white.
41. The transgenic avian of Claim 40, wherein the transgenic avian produces the heterologous polypeptide in an egg white.
20 42. The transgenic avian of claim 41, wherein the nucleotide sequence encodes an immunoglobulin or subunit thereof.
25
30
35
- 47 -
PCT/US2002/038413 2001-11-30 2002-12-02 Ovomucoid promoter and methods of use WO2003048364A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
JP2003549541A JP2005536984A (en) 2001-11-30 2002-12-02 Ovomucoid promoter and method of use
CA2467648A CA2467648C (en) 2001-11-30 2002-12-02 Ovomucoid promoter and methods of use
US10/496,731 US7375258B2 (en) 2001-11-30 2002-12-02 Transgenic avians with an ovomucoid gene expression control region linked to a nucleotide sequence encoding a heterologous polypeptide
AU2002351192A AU2002351192B2 (en) 2001-11-30 2002-12-02 Ovomucoid promoter and methods of use
EP02786838A EP1461430A4 (en) 2001-11-30 2002-12-02 Ovomucoid promoter and methods of use
US10/856,218 US7294507B2 (en) 2001-11-30 2004-05-28 Ovomucoid promoters and methods of use
US11/047,184 US7335761B2 (en) 2001-11-30 2005-01-31 Avian gene expression controlling regions
US11/649,543 US7507873B2 (en) 2001-11-30 2007-01-04 Transgenic avians containing recombinant ovomucoid promoters
US12/313,064 US7812215B2 (en) 2001-11-30 2008-11-17 Methods and protein production using ovomucoid promoters
US12/799,605 US20100333219A1 (en) 2001-11-30 2010-04-28 Methods of protein production using ovomucoid regulatory regions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/998,716 2001-11-30
US09/998,716 US6875588B2 (en) 2001-11-30 2001-11-30 Ovomucoid promoter and methods of use

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US09/998,716 Continuation-In-Part US6875588B2 (en) 2001-11-30 2001-11-30 Ovomucoid promoter and methods of use
US10/856,218 Continuation-In-Part US7294507B2 (en) 2001-11-30 2004-05-28 Ovomucoid promoters and methods of use

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10496731 A-371-Of-International 2002-12-02
US10/856,218 Continuation-In-Part US7294507B2 (en) 2001-11-30 2004-05-28 Ovomucoid promoters and methods of use

Publications (2)

Publication Number Publication Date
WO2003048364A2 true WO2003048364A2 (en) 2003-06-12
WO2003048364A3 WO2003048364A3 (en) 2003-09-12

Family

ID=25545501

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/038413 WO2003048364A2 (en) 2001-11-30 2002-12-02 Ovomucoid promoter and methods of use

Country Status (6)

Country Link
US (2) US6875588B2 (en)
EP (1) EP1461430A4 (en)
JP (1) JP2005536984A (en)
AU (1) AU2002351192B2 (en)
CA (1) CA2467648C (en)
WO (1) WO2003048364A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005040215A2 (en) * 2003-06-06 2005-05-06 Avigenics, Inc. Ovomucoid promoters and mehtods of use
US7294507B2 (en) 2001-11-30 2007-11-13 Avigenics, Inc. Ovomucoid promoters and methods of use
US7335761B2 (en) 2001-11-30 2008-02-26 Avigenics, Inc. Avian gene expression controlling regions
US8071364B2 (en) 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
KR102159050B1 (en) * 2019-07-12 2020-09-24 대한민국 Recombinant promoter comprising the promoter region of the ovomucoid gene and the promoter region of the ovalbumin gene
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040019923A1 (en) * 1997-10-16 2004-01-29 Ivarie Robert D. Exogenous proteins expressed in avians and their eggs
ATE489461T1 (en) * 1997-10-16 2010-12-15 Univ Georgia TRANSGENIC BIRDS AND PROTEIN PRODUCTION
DK1395669T3 (en) * 2001-01-26 2009-11-16 Selexis Sa Matrix binding regions and methods for using them
AU2002341746A1 (en) 2001-09-18 2003-04-01 Avigenics, Inc. Production of a transgenic avian by cytoplasmic injection
US7550650B2 (en) * 2001-09-18 2009-06-23 Synageva Biopharma Corp. Production of a transgenic avian by cytoplasmic injection
US20100333219A1 (en) * 2001-11-30 2010-12-30 Synageva Biopharma Corp. Methods of protein production using ovomucoid regulatory regions
US20040172667A1 (en) * 2002-06-26 2004-09-02 Cooper Richard K. Administration of transposon-based vectors to reproductive organs
US7527966B2 (en) * 2002-06-26 2009-05-05 Transgenrx, Inc. Gene regulation in transgenic animals using a transposon-based vector
US20040255345A1 (en) * 2003-03-07 2004-12-16 Rapp Jeffrey C. Production of transgenic avians
US20050273873A1 (en) * 2003-03-07 2005-12-08 Avigenics, Inc. Genomic modification
US20050034186A1 (en) * 2003-03-07 2005-02-10 Harvey Alex J. Site specific nucleic acid integration
US20050198700A1 (en) * 2003-03-07 2005-09-08 Avigenics, Inc. Genomic modification
US7381712B2 (en) * 2003-05-09 2008-06-03 Avigenics, Inc. In vivo transfection in avians
US20080222744A1 (en) * 2003-05-09 2008-09-11 Avigenics, Inc. In vivo transfection in avians
IN2014DN02483A (en) 2003-10-24 2015-05-15 Selexis Sa
US20060174364A1 (en) * 2004-03-01 2006-08-03 Avigenics, Inc. Artificial chromosomes and transchromosomic avians
US20060123504A1 (en) * 2004-12-07 2006-06-08 Avigenics, Inc. Methods of producing polyclonal antibodies
GB0419957D0 (en) * 2004-09-09 2004-10-13 Viragen Scotland Ltd Screening method
WO2006042214A1 (en) * 2004-10-08 2006-04-20 Avigenics, Inc. Modified integrase and methods of use
US20080064862A1 (en) * 2004-12-29 2008-03-13 Avigenics, Inc. Transgene expression in a avians
WO2010036976A2 (en) * 2008-09-25 2010-04-01 Transgenrx, Inc. Novel vectors for production of antibodies
US9157097B2 (en) * 2008-09-25 2015-10-13 Proteovec Holding, L.L.C. Vectors for production of growth hormone
US20100081789A1 (en) * 2008-09-25 2010-04-01 Cooper Richard K Novel Vectors for Production of Interferon
WO2010118360A1 (en) * 2009-04-09 2010-10-14 The Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Production of proteins using transposon-based vectors
CA2790245C (en) * 2010-02-19 2018-10-09 Universite De Liege A polynucleotide for use in treatment of influenza a virus induced diseases, encoding modified mx protein, said modified mx protein, and a transgenic animal expressing gene encoding modified mx protein
US8846603B2 (en) 2010-03-12 2014-09-30 Synageva Biopharma Corp. NPP1 fusion proteins
WO2012125182A1 (en) 2011-03-11 2012-09-20 Synageva Biopharma Corp Npp1 fusion proteins
US9510571B2 (en) 2011-09-02 2016-12-06 Kaneka Corporation Transgenic bird expressing foreign gene using endoplasmic reticulum chaperone promoter
US20140255383A1 (en) 2011-10-12 2014-09-11 Synageva Biopharma Corp. Recombinant human naglu protein and uses thereof
EP3200816A1 (en) 2014-09-29 2017-08-09 Alexion Pharmaceuticals, Inc. Methods of treating mucopolysaccharidosis iiib (mpsiiib)
US11518797B2 (en) 2014-11-11 2022-12-06 Clara Foods Co. Methods and compositions for egg white protein production
WO2019060993A1 (en) 2017-09-28 2019-04-04 The Governors Of The University Of Alberta Retinoic acid-inducible gene i promoter and compositions and methods relating to same
AU2020309602A1 (en) 2019-07-11 2022-02-24 Clara Foods Co. Protein compositions and consumable products thereof
US10927360B1 (en) 2019-08-07 2021-02-23 Clara Foods Co. Compositions comprising digestive enzymes

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4237224A (en) 1974-11-04 1980-12-02 Board Of Trustees Of The Leland Stanford Jr. University Process for producing biologically functional molecular chimeras
US5505941A (en) 1981-12-24 1996-04-09 Health Research, Inc. Recombinant avipox virus and method to induce an immune response
US5338683A (en) 1981-12-24 1994-08-16 Health Research Incorporated Vaccinia virus containing DNA sequences encoding herpesvirus glycoproteins
US5174993A (en) 1981-12-24 1992-12-29 Health Research Inc. Recombinant avipox virus and immunological use thereof
US4603112A (en) 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US4769330A (en) 1981-12-24 1988-09-06 Health Research, Incorporated Modified vaccinia virus and methods for making and using the same
US4722848A (en) 1982-12-08 1988-02-02 Health Research, Incorporated Method for immunizing animals with synthetically modified vaccinia virus
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
AU660629B2 (en) 1990-10-01 1995-07-06 University Of Connecticut, The Targeting viruses and cells for selective internalization by cells
EP0575491B1 (en) 1991-03-07 2003-08-13 Virogenetics Corporation Genetically engineered vaccine strain
WO1992019749A1 (en) 1991-05-03 1992-11-12 The Board Of Regents Acting For And On Behalf Of The University Of Michigan Targeted delivery of genes encoding cell surface receptors
ES2154637T3 (en) 1991-05-14 2001-04-16 Univ Connecticut CONTRIBUTION OF DIRECT GENES THAT CODIFY IMMUNOGENIC PROTEINS.
ATE195656T1 (en) 1991-06-05 2000-09-15 Univ Connecticut TARGETED RELEASE OF GENES ENCODING SECRETORY PROTEINS
CA2116107A1 (en) 1991-09-05 1993-03-18 George Y. Wu Targeted delivery of poly-or oligonucleotides to cells
EP0650370A4 (en) 1992-06-08 1995-11-22 Univ California Methods and compositions for targeting specific tissue.
DE69333115D1 (en) 1992-09-22 2003-08-28 Biofocus Discovery Ltd RECOMBINANT VIRUSES PRESENTING A NON-VIRAL POLYPEPTIDE ON THEIR OUTER SURFACE
CA2148934C (en) 1992-11-09 2005-08-02 French W. Anderson Targetable vector particles
US6825396B2 (en) 1996-06-12 2004-11-30 Board Of Trustees Operating Michigan State University Methods for tissue specific synthesis of protein in eggs of transgenic hens
ATE489461T1 (en) 1997-10-16 2010-12-15 Univ Georgia TRANSGENIC BIRDS AND PROTEIN PRODUCTION

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GERLINGER ET AL.: 'Multiple initiation and polyadenylation sites for the chicken ovomucoid transcription unit' JOURNAL OF MOLECULAR BIOLOGY vol. 162, 1982, pages 345 - 364, XP002963243 *
See also references of EP1461430A2 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7294507B2 (en) 2001-11-30 2007-11-13 Avigenics, Inc. Ovomucoid promoters and methods of use
US7335761B2 (en) 2001-11-30 2008-02-26 Avigenics, Inc. Avian gene expression controlling regions
US7507873B2 (en) 2001-11-30 2009-03-24 Avigenics, Inc. Transgenic avians containing recombinant ovomucoid promoters
US7812215B2 (en) 2001-11-30 2010-10-12 Synageva Biopharma Corp. Methods and protein production using ovomucoid promoters
WO2005040215A2 (en) * 2003-06-06 2005-05-06 Avigenics, Inc. Ovomucoid promoters and mehtods of use
WO2005040215A3 (en) * 2003-06-06 2005-06-30 Avigenics Inc Ovomucoid promoters and mehtods of use
US8071364B2 (en) 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
US11344608B2 (en) 2014-11-12 2022-05-31 Ucl Business Ltd Factor IX gene therapy
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
US11517631B2 (en) 2018-08-20 2022-12-06 Ucl Business Ltd Factor IX encoding nucleotides
KR102159050B1 (en) * 2019-07-12 2020-09-24 대한민국 Recombinant promoter comprising the promoter region of the ovomucoid gene and the promoter region of the ovalbumin gene

Also Published As

Publication number Publication date
US20050050581A1 (en) 2005-03-03
CA2467648C (en) 2012-06-12
EP1461430A2 (en) 2004-09-29
AU2002351192B2 (en) 2006-12-14
AU2002351192A1 (en) 2003-06-17
JP2005536984A (en) 2005-12-08
WO2003048364A3 (en) 2003-09-12
US20030126628A1 (en) 2003-07-03
US6875588B2 (en) 2005-04-05
EP1461430A4 (en) 2005-04-13
US7375258B2 (en) 2008-05-20
CA2467648A1 (en) 2003-06-12

Similar Documents

Publication Publication Date Title
AU2002351192B2 (en) Ovomucoid promoter and methods of use
US7199279B2 (en) Recombinant promoters in avian cells
US7541512B2 (en) Avians containing a lysozyme promoter transgene
US7507873B2 (en) Transgenic avians containing recombinant ovomucoid promoters
AU2007345347B2 (en) Transgene expression in avians
US7294507B2 (en) Ovomucoid promoters and methods of use
US7176300B2 (en) Avian lysozyme promoter
US20090210951A1 (en) Avian transgenesis using an ovalbumin nucleotide sequence
US20080124792A1 (en) Hybrid promoters
WO2003024199A2 (en) Production of transgenic avians using sperm-mediated transfection
WO2003024200A2 (en) Production of transgenic birds using stage x primordial germ cells
NZ578390A (en) Transgene expression in avians using oviduct specific promoters and SIN vectors
WO2005040215A2 (en) Ovomucoid promoters and mehtods of use
AU2007201063B2 (en) Ovomucoid promoter and methods of use
US20100333219A1 (en) Methods of protein production using ovomucoid regulatory regions

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 NO NZ OM PH PL PT RO RU SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ 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 IE IT LU MC NL PT 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
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2002351192

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2467648

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 10496731

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2003549541

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2002786838

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002786838

Country of ref document: EP