WO1993009236A1 - Myogenic vector systems - Google Patents

Myogenic vector systems Download PDF

Info

Publication number
WO1993009236A1
WO1993009236A1 PCT/US1992/009353 US9209353W WO9309236A1 WO 1993009236 A1 WO1993009236 A1 WO 1993009236A1 US 9209353 W US9209353 W US 9209353W WO 9309236 A1 WO9309236 A1 WO 9309236A1
Authority
WO
WIPO (PCT)
Prior art keywords
mvs
promoter
nucleic acid
myogenic
human
Prior art date
Application number
PCT/US1992/009353
Other languages
French (fr)
Inventor
Robert J. Schwartz
Franco J. Demayo
Bert W. O'malley
Original Assignee
Baylor College Of Medicine
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Baylor College Of Medicine filed Critical Baylor College Of Medicine
Priority to AU31246/93A priority Critical patent/AU660751B2/en
Priority to JP5508631A priority patent/JPH07500967A/en
Priority to EP92925044A priority patent/EP0635060A4/en
Publication of WO1993009236A1 publication Critical patent/WO1993009236A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4707Muscular dystrophy
    • C07K14/4708Duchenne dystrophy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/001Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
    • C12N2830/002Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/90Vector systems having a special element relevant for transcription from vertebrates avian

Definitions

  • the present invention relates generally to expression vectors for use in expressing polypeptides in myogenic cells. More particularly it relates to vectors containing the skeletal alpha actin gene promoter and the corresponding 3' transcribed but untranslated region of the gene and the 10 contiguous noncoding DNA containing the gene's natural transcriptional termination region.
  • Actin is a contractile protein found in most cell types. Actin proteins are represented by similar but nonidentical isoforms which in warm blooded
  • Actin is expressed in a developmentally timed dependent and tissue specific manner, in which adult muscle tissues express exclusively the skeletal alpha actin gene as the predominant actin isoform.
  • the nucleic acid sequence of skeletal alpha actin gene has been characterized in chicken, rat, mouse and human. Fornwald et
  • RNA transcripts are initiated from a promoter region start site at the messenger RNA cap and continue through the protein coding region to the noncoding region involved with transcriptional termination. Post transcriptional processing of the RNA transcripts removes the noncoding introns to form a continuous coding sequence. Further, there is progressive trimming of the RNA transcript to the polyadenylation signals, found in the noncoding 3' untranslated portion of the processed messenger RNA.
  • the levels of messenger RNA biosynthesis and accumulation is determined by parts of the unexpressed portion of the gene. For example the promoter, which is part of the unexpressed portion of the gene, is involved in determining gene expression. It regulates when and how the expressed gene is transcribed.
  • RNA transcripts may relate to the intrinsic stability of that RNA in a particular cell type. This intrinsic stability is dependent on sequences within the mRNA that provide stabilization. Thus, the ability to express a particular gene product is a balance between transcription rate and the stability of the mRNA transcript.
  • Expression of the actin gene family is tissue specific. The skeletal alpha actin protein is expressed primarily in cardiac and skeletal muscle tissues. Transient transfection experiments indicated that regions within the 200 bp promoter region are sufficient for tissue restricted expression in primary myoblasts. Bergsma, et al, Mol. Cell. Biol., Vol.,6 pp. 2462-2475 (1986) and Chow and Schwartz Mol. Cell Biol., Vol.10 pp. 528-538 (1990).
  • the promoter region harbors conserved cis-acting elements to accurately initiate skeletal alpha actin transcripts from the bacterial reporter gene chloramphenicol acetyltransferase (CAT) in differentiating myoblasts.
  • CAT chloramphenicol acetyltransferase
  • Transgenic mice with an integrated skeletal alpha actin promoter showed preferential expression of the CAT gene in myogenic tissues Petropulous et al. Mol. Cell. Biol., Vol. 9 pp. 3785-3792 (1989). It is also known that CAT activity is detectable as early as 10 days in the mouse embryo when the embryonic heart is first being formed. Further, CAT activity can be induced in newborn skeletal muscle.
  • the skeletal actin promoter can switch transcriptional activity in mammalian systems, to provide muscle restricted expression.
  • the CAT messenger RNA could not be detected by standard RNA blotting techniques.
  • the skeletal actin promoter is not the cause for low levels of messenger RNA accumulation.
  • the actin promoter is by itself insufficient to drive the expression of other gene products to accumulate at levels which are comparable to the intact skeletal alpha actin mRNA.
  • the rate of metabolic breakdown of mRNA molecules is an important factor in the regulation of gene expression.
  • the rates of decay of individual mRNA species can affect strongly the steady state levels of these species in the cytoplasm. Consequently, the extent of expression of a given gene, as measured by the rate of synthesis of the corresponding protein, will be dependent to a large extent on the degree of stability of the mRNA derived from this gene.
  • Messenger RNA from skeletal muscle was previously shown to be distributed into two populations with regard to its stability.
  • An object of the present invention is a myogenic vector system capable of expressing any specific nucleic acid sequence in myogenic tissue.
  • An additional object of the present invention is a regulatable myogenic vector system.
  • a further object of the present invention is a method for treating muscle atrophy in aging humans.
  • Another method of the present invention is a method for treating muscle atrophy induced by spinal cord injuries or muscular disease.
  • a further object of the present invention is a method of preventing or treating atherosclerotic cardiovascular disease.
  • An additional method of the present invention is the introduction of a myogenic vector system for gene replacement.
  • a further object of the invention is a method for vaccine production in human or animals.
  • a myogenic vector system for the expression of a nucleic acid sequence in myogenic tissue comprising a promoter; a cassette whose 5' end is attached to the 3' end of the promoter, the cassette containing a nucleic acid sequence which is to be expressed; a myogenic specific 3' untranslated region (3' UTR) and a non ⁇ coding region (NCR) contiguous to the 3' end of the 3' UTR, the NCR containing a transcriptional termination signal, wherein the 5' end of the 3' UTR is attached to the 3' end of the cassette.
  • MVS myogenic vector system
  • Specific embodiments of the MVS of the present invention can further include a leader sequence following the promoter region. Additional embodiments of the MVS include a first intron, an initiation ATG and Ncol cloning site inserted between the promoter leader region area and the cassette area. Specific embodiments of the cassette include an EcoRl site at the 3' end. The 3' UTR can have an EcoRV site at its 5' end.
  • any promoter will work although in the preferred embodiment a myogenic promoter selected from a group consisting of skeletal alpha actin gene promoter, first myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter/enhancer, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR.
  • a myogenic promoter selected from a group consisting of skeletal alpha actin gene promoter, first myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter/enhancer, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR.
  • the skeletal alpha actin promoter is used.
  • the 3' UTR and NCR are selected from any myogenic specific genes.
  • the skeletal alpha actin gene 3' UTR and NCR regions are used.
  • Specific embodiment also include the addition of regulatory promoter elements to regulate the expression of any specific nucleic acid sequence in myogenic tissue.
  • Vitamin D is used to regulate expression. treatment of prevention of atherosclerotic cardiovascular disease, gene replacement, and vaccine production.
  • Figure 1 is a schematic drawing of the chicken skeletal alpha actin gene which includes location of unique restriction sites.
  • Figure 2 illustrates the transcriptional domain of the avian skeletal alpha actin gene and the contiguous noncoding region where transcription terminates (OD»mm/kb Insert Relative GAPDH).
  • Figure 3 is a schematic representation of a myogenic vector system.
  • Figure 4 is a schematic diagram of skeletal alpha actin/insulin like growth factor-I hybrid genes
  • Figure 5 illustrates the increased activity of the myogenic IGF-I hybrid vectors containing the skeletal alpha actin 3' untranslated region and contiguous noncoding region by the accumulation of IGF-I RNA in stably transfected C 2 C 12 myoblasts.
  • Figure 6 shows the accumulation of IGF-I RNA in transgenic mouse lines generated with SK202IGF-I-3'SVa myogenic expression vector.
  • Figure 7 shows the accumulation of IGF-I RNA in transgenic mouse lines generated with SK202IGF-I-3'SK myogenic expression vector.
  • Figure 8 is a schematic representative of a regulatable MVS using a Vitamin D receptor.
  • Figure 9 is a schematic representative of a regulatable MVS using a chimeric receptor.
  • SUBSTITUTE SHEET Figure 10 illustrates the stability of IGF-I mRNAs with different 3' UTRS.
  • the drawings are not necessarily at this scale. Certain features of the invention may be exaggerated in scale or shown in schematic form in the interest of clarity and conciseness.
  • promoter refers to a recognition site on a DNA strand to which the RNA polymerase binds.
  • the promoter usually is a DNA fragment of about 100 to 200 bp in the 5' flanking DNA upstream of the cap site or the transcriptional initiation start site.
  • the promoter forms an initiation complex with RNA polymerase to initiate and drive transcriptional activity.
  • the complex can be modified by activating sequences termed “enhancers” or inhibiting sequences termed “silencers.”
  • enhancers activating sequences termed "enhancers” or inhibiting sequences termed "silencers.”
  • Usually specific regulatory sequences or elements are embedded adjacent to or within the protein coding regions of DNA. The elements, located adjacent to the gene, are termed cis-acting elements.
  • trans-acting factors The presence of trans ⁇ acting factors and cis-acting elements have been shown to contribute to the timing and developmental expression pattern of a gene. Cis acting elements
  • SUBSTITUTE SHEET are usually thought of as those that regulate transcription and are found within promoter regions and other upstream DNA flanking sequences.
  • leader refers to a DNA sequence at the 5' end of a structural gene which is transcribed along with the gene.
  • the leader usually results in the protein having an N-terminal peptide extension sometimes called a pro-sequence.
  • this signal sequence which largely hydrophobic, directs the protein into endoplasmic reticulum from which it is discharged to the appropriate destination.
  • intron refers to a section of DNA occurring in the middle of a gene which does not code for an amino acid in the gene product. The precursor RNA of the intron is excised and is therefore not transcribed into mRNA nor translated into protein.
  • cassette refers to the sequence of the present invention which contains the nucleic acid sequence which is to be expressed.
  • the cassette is similar in concept to a cassette tape. Each cassette will have its own sequence. Thus by interchanging the cassette the vector will express a different sequence. Because of the restrictions sites at the 5' and 3' ends, the cassette can be easily inserted, removed or replaced with another cassette.
  • 3' untranslated region refers to the sequence at the 3' end of a structural gene which is usually transcribed with the gene. This 3' UTR region usually contains the poly A sequence. Although the 3' UTR is transcribed from the DNA it is excised before translation into the protein. In the present invention it is preferred to have a myogenic specific 3' UTR. This allows for specific stability in the myogenic tissues.
  • NCR Non-Coding Region
  • the 3' UTR and NCR are a key aspect of the present invention because they provide a higher level of mRNA accumulation through increased mRNA stability in myogenic cells rather than non-myogenic cells. Thus, this increased stability of mRNA leads to the increased levels of protein production.
  • the 3' untranslated region of the chicken skeletal alpha actin gene which starts at nucleotide 2060 and extends to 2331.
  • the complete 3' untranslated region and contiguous noncoding DNA extends an additional 2.0
  • This 2.3 Kb fragment can be linked immediately following the natural translation termination codon to a copy DNA sequence coding for a polypeptide desired to be expressed.
  • muscle tissue can be in vivo tissue, in vitro tissue or in vitro tissue cultures of cells capable of differentiating into muscle tissue.
  • Myogenic cells include skeletal, heart and smooth muscle cells. These vectors are transfected into myogenic cells in culture, and are injected into intact muscle tissue.
  • vectors containing the construction are injected into mammalian oocytes and may be stably incorporated into the genome to generate transgenic animals, in which the vector expresses polypeptides in myogenic cells.
  • restriction site refers to a sequence specific cleavage site of restriction endonucleases.
  • vector refers to some means by which DNA fragments can be introduced into a host organism or host tissue. There are various types of vectors including plasmid, bacteriophages and cosmids.
  • the term "effective amount” means sufficient MVS is injected into myogenic tissue or culture to produce the adequate levels of the polypeptide.
  • This actual level will depend on the use of the MVS. The levels will be different in treatment, vaccine production, or vaccination.
  • a myogenic vector system for the expression of a nucleic acid sequence in myogenic tissue, comprising a promotor; a cassette whose 5' end is attached to the 3' end of the promoter; a myogenic specific 3' UTR and a NCR contiguous to the 3' end of the 3' UTR.
  • This basic system can be enhanced in a variety of ways including the addition of a leader sequence between the promoter and cassette.
  • the MVS for expression of a nucleic acid sequence in myogenic tissue comprises a functional unit which expresses the nucleic acid sequence.
  • the functional unit is comprised of elements all sequentially linked together 5' to 3'.
  • the elements in the order of linkage include a promoter, a 5' mRNA leader sequence, a first intron and initiation ATG and Ncol cloning site, a cassette which has a EcoRI site at its 3' end, a myogenic specific 3' UTR in which the 5' end of the 3' UTR has an EcoRV site, and a NCR which is contiguous to the 3' end of the 3' UTR.
  • promoters can be used in the MVS. Some examples include skeletal alpha actin gene promoter, first myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR. In the preferred embodiment, a myogenic specific promoter such as the skeletal alpha actin gene promoter is used.
  • the 3' UTR and NCR can be selected from any group of myogenic specific genes. Examples of genes in this group are the skeletal alpha actin gene, fast myosin light chain 1 gene, myosin heavy chain gene, tropinin T gene, acetyl choline receptor subunit genes and muscle creatinine kinase gene.
  • the 3' UTR and NCR are from the skeletal alpha actin gene.
  • Alternate embodiments of the present invention include the addition of a regulator system for regulating the expression of nucleic acid sequence. Any of a variety of regulator systems can be used. In the preferred embodiment, two different regulatory systems have been used.
  • a regulated MVS for the expression of a specific nucleic acid sequence in myogenic tissue, is comprised of a first functional unit and a second functional unit.
  • the first functional unit and second functional unit can be in the same vector or in two separate vectors.
  • the first functional unit is composed of the following elements all sequentially linked 5' to 3': a myogenic specific promoter, a nucleic acid sequence coding for a receptor, a myogenic specific 3 ' UTR and a myogenic specific NCR.
  • the second functional unit is composed of the following elements sequentially linked 5' to 3' : a response element corresponding to the receptor, a thymidine kinase promoter, a cassette containing the specific nucleic acid sequence of interest, a myogenic specific 3 * UTR and a contiguous myogenic specific NCR. In this regulatable MVS, it is preferable for the first functional unit to continuously express the receptor.
  • a receptor which is not found in high levels in myogenic tissue, when the agent which is specific to the receptor is introduced into the system.
  • the receptor forms an interaction with the response element and the specific agent. This binding interaction causes the thymidine kinase promoter to express the specific nucleic acid sequence.
  • the activity of the MVS is regulated.
  • the response element and receptor are usually complimentary and can be selected from a variety of receptor groups.
  • any vitamin, steroid, thyroid, orphan, hormone, retinoic acid and thyroxine can be used.
  • the Vitamin D receptor and Vitamin D response element are used.
  • ingestion of Vitamin D for instance by drinking milk, causes the level of Vitamin D in the blood. This binds to the receptor being generated in the myogenic tissue and the complex binds to the receptor element causing the expression of the gene of interest.
  • the MVS can be regulated by dietary intake.
  • FIG. 9 An alternate method of regulating the MVS is shown in Figure 9.
  • at least one of the serum response elements in the alpha actin promoter region is made into a receptor binding site. Then a chimeric transfactor is constructed in which the normal DNA binding domain of the serum response factor is replaced with the DNA binding domain of the receptor. The transactivation domain of the serum response factor is not changed.
  • an agent or ligand specific to the receptor it binds to the receptor in the binding domain allowing the serum response transactivation factor to activate transcription.
  • the regulation can be controlled by controlling the amount of the agent.
  • the DNA binding domain which is substituted into the normal serum response factor is usually selected from the following families of receptors binding domains: the vitamin, steroid, thyroid, orphan, hormone, retinoic acid and thyroxine.
  • the Vitamin D receptor is used.
  • the cassette can contain the nucleic acid sequence of interest and include any nucleic acid sequence which is to be expresses in the myogenic tissue or tissue culture. These nucleic acid sequence can call for any variety of polypeptides.
  • the polypeptide can be any desired polypeptide including but not limited to known proteins.
  • the sequence could code for a hormone, a growth factor, an enzyme, an apolipoprotein, tumor suppressor, tumor antigen, a viral protein, a clotting factor and any proteins associated with the AIDS virus, any other viral proteins, including viral surface coat proteins, bacterial surface proteins, parasitic cell surface proteins, viral reverse transcriptase and any gene which needs to be replaced by gene replacement.
  • the cassette included the nucleic acid for the insulin like growth factor I, or the insulin like factor II or the insulin growth binding protein.
  • This specific embodiment can be used to treat muscle atrophy in aging humans, muscle atrophy induced by spinal cord injuries or neuromuscular diseases. A specific example of the latter case would be amyotrophic lateral sclerosis.
  • Another specific embodiment is a MVS where the cassette contains the nucleic acid sequence coding for growth hormone releasing factor. This MVS can be used for treating muscle atrophy in aging humans.
  • Another embodiment of the present invention includes an MVS where the cassette includes the nucleic acid sequence for apolipoprotein A-I.
  • This MVS can be used for prevention or treatment of atherosclerotic cardiovascular disease.
  • the MVS cassette contains a sequence coding for a viral, bacterial or parasitic protein
  • the MVS can be used to make a vaccine.
  • the procedure for making a vaccine humans or animals comprising the step of injecting an effective amount of the MVS into skeletal muscle or into tissue culture.
  • the cassette contains a nucleic acid sequence which codes for a polypeptides capable of eliciting an antibody response.
  • the vaccine can be generated in tissue culture, or in vivo in humans or animals. Examples of nucleic acid sequences which can elicit an antibody response include those for the viral proteins, bacterial proteins, and parasitic proteins. A specific case example is the AIDS proteins.
  • Another specific embodiment of the present invention is a method of treating growth disease which comprises the step of injecting an effective amount of an MVS into skeletal muscle wherein the nucleic acid sequence in the cassette contains the growth hormone sequence.
  • Another application of the present invention is a method for gene replacement.
  • an effective amount of the MVS is injected into skeletal muscle and the cassette contains any sequence which codes for a defective gene.
  • the genes for glycogen phosphorylase, alpha- 1- antitrypsin and dystrophin can be inserted into the cassette. Individuals with the corresponding diseases of glycogen storage disease, alpha-1-antitrypsin deficiency or pulmonary emphysemia and Duchenne's Muscular Dystrophy would thus produce a sufficient amount of the normal polypeptide.
  • the expressed protein may systemically spread throughout the animal via the circulatory blood system as well as remain in the injected musculature.
  • the polypeptide may be harvested and purified as desired.
  • the coating includes a DNA initiation complex and histones.
  • the initiation complex comprises a serum response factor (SRF), a transcription initiation factor (TIF) and a transregulatory factor (TRF).
  • SRF serum response factor
  • TIF transcription initiation factor
  • TRF transregulatory factor
  • the SRF is attached to the serum response element within the promoter region of the MVS.
  • TIF and the TRF then interact with the SRF and the TATA box within the promoter to form a stable DNA complex.
  • the histones bind nonspecifically to the remaining DNA in the MVS.
  • RNA transcripts with radioactive tagged nucleotides. Labeled RNA hybridized to dotted DNA probes showed that transcription terminates approximately 1 kb downstream of the skeletal alpha actin gene's poly A addition site. This is within a 800 bp Pvu II fragment between +2800 and +3600 nucleotides from the start of transcription.
  • the 3' untranslated region (3' UTR) and the contiguous noncoding region (NCR) can be isolated by restriction endonucleases digestion of the 6.2 Kb actin gene with blunt cutter Nae I, which cuts 30 bp upstream of the translation termination codon TAA. Hind III releases the 3' most portion of the actin gene from the vector pBR322 ( Figure 3). The 3' UTR and NCR were used to prepare DNA constructs.
  • the skeletal alpha actin promoter and DNA flanking sequences (at least 411 nucleotides from the mRNA cap site) and DNA sequences extending through the skeletal 5' noncoding leader, first intron and up to the initiation of translation ATG, converted to a Nco I cloning site at +196, was liberated from a M13 double stranded DNA by Xba
  • the SK733 NcoI vector contains approximately 411 nucleotides of the skeletal alpha actin promoter, the natural cap site, 5' untranslated leader and the first intron.
  • An NcoI site was engineered to create a unique insertion cloning site for the cassette containing the IGF-I cDNA, in which the initiation ATG was also converted to an NcoI site.
  • the SK733IGF-I construction utilizes its own poly A site.
  • Example 3 Activity of MVS Constructs To determine the efficacy of actin promoter/gene IGF-I hybrid genes in mouse myogenic cells the MVS was studied using these genes in the background of mammalian C 2 C 12 myoblasts by making a population of stable transfected C 2 C 12 myoblasts. The altered IGF-I expression levels were directly evaluated in these stablemyoblast cell lines. Each IGF-I construction ( Figure 4) was co-transfected with the drug selectable vector EMSV-Hygromycin into mouse C JJ C- J , cells. After two weeks of selection, a population of stable myoblasts was selected. A population of C 2 C 12 myoblasts stably transfected only with EMSV-Hygromycin served as the controls.
  • SK733IGF-I-SK1 and SK733IGF-I-SK2 fused at least two-to-three days earlier than C 2 C 12 or EMSV-Hygromycin control myoblasts.
  • Figure 5 shows the steady state accumulation of IGF-I mRNA in C 2 C 12 myoblasts.
  • Equal amounts of total cellular RNA was isolated from stably transfected C 2 C 12 myoblasts grown in growth media (G) or differentiation media (D) .
  • the RNA was electrophoretically separated on denaturing agarose gels, transferred onto nylon filters and probed with uniformly ⁇ P labeled full length human IGF-I cDNA under standard hybridization techniques.
  • the intensity of the autoradiographic signal on X-ray film provides a relative measure of mRNA accumulation, an overall index of combined transcriptional activity and mRNA stability of the myogenic expression vector system.
  • the IGF-I mRNA in vector, SK202IGF-I-3'SVa did not accumulate in myotubes above myoblast levels. This is a typical expression activity.
  • the SK733IGF-I vector contains the IGF-I 3' untranslated region.
  • the IGF-I mRNA from this vector accumulated in myotubes but at levels substantially lower than SK202IGF-I-SK or SK733IGF-I-SK2.
  • These later two vectors contain the skeletal actin 3' UTR and contiguous noncoding regions. Since, the primary difference in these vectors is the 3' UTR, the increased stabilization of the mRNA transcripts due to the skeletal 3' UTR accounts for about a 100 fold difference in mRNA content.
  • SK733IGF-I-SK2 is the most effective construction to express IGF-I in muscle cells. IGF-I was assayed by both radioimmunoassays of tissue culture media and by immunoperoxidase staining of cells. We have found increased levels of IGF-I during the fusion of several of our muscle cultures. The comparison of levels from different MVS are shown in Table 1. In control cultures, the level of IGF-I was in the range of 0.2-0.5 ng/ml. In comparison, vector SK733IGF-I-SK2 has levels of IGF-I at least one hundred times greater.
  • Transcripts containing the natural human IGF-I 3' UTR were found to turn over rapidly with a half life of less than 1 hour. These results are consistent with the short half life of growth factors. IGF-I transcripts containing SV40 3' UTR and poly A addition signals displayed a half life of about 4 hours. In contrast, IGF-I transcripts containing the skeletal alpha actin 3' UTR showed a high level of stability with a half life greater than 18 hours. These results support the claim that the high level of IGF-I mRNA that accumulates in the
  • SK733IGF-I-SK2 transfected C2C12 myoblasts is due in part to the differential stabilization of mRNA which contains the contiguous skeletal alpha actin 3' UTR.
  • immunoperoxidase staining of myogenic cultures revealed the increased production of immunological reactive IGF-I in stable transfected myoblasts but not in the control EMSV-Hygromycin transfected myoblasts or in perfus ⁇ on C 2 C 12 cells.
  • Antibodies against the A and D regions were used at dilutions of 1:1000. All of the transfected lines including SK202IGF-I were positively immunoperoxidase stained.
  • mice carrying SK202IGF-I-3'SVa or SK202IGF-I-SK were generated by standard oocyte injection (Brinster, et al, Proc. Natl. Acad. Sci.
  • Transgenics were identified by polymerase chain reaction or Southern genomic DNA blotting analysis from tail cut DNA. Transgenics were tested for muscle specific expression of the transferred IGF-I vector by RNA blotting of total RNA isolated from several tissues as shown in Figures 6 and 7. Independent transgenic mouse lines 5484, 5496, 5832, 5834 were generated with SK202IGF-I-3'SVa, containing the SV40 3' intron and poly A addition sequence. Mice from these strains were found to have weak expression primarily in heart tissue, but very low levels were found in skeletal muscle and non-myogenic tissues such as the kidney and brain ( Figure 6).
  • Muscle tissue homogenates were analyzed by radioimmunoassay to determine the relative levels of IGF-I production. Homogenates were acid chromatographed to separate IGF-I from IGF binding proteins after acid chromatography fractions containing IGF-I were pooled, lyophilized, reconstituted and assayed by the protocol of Liu, F, Powell, D.R., Styne, D.M. and Hintz, R.L. (191) J. Clin. Endocrin. Metab. 72:905-911. The IGF-I antiserum was provided by the National Hormone and Pituitary Program.
  • IGF-I 100 ng/lOOug of muscle protein
  • Example 6 Expression of MVS into Intact Muscle
  • Intact plasmid DNA in a sterile 20% sucrose solution (wt/vol) can be injected into mature avian or mammalian muscle. Following a single injection the vector DNA is stable for at least 30 days as a non-integrated extrachromosomal circular DNA in muscle nuclei and, is transcriptionally active.
  • Wolf et al. Science, 247: 1465-1468 (1990). However, greater than 99% of the injected DNA is degraded in muscle under the Wolff protocol (Wolff, et al, BioTechniques Vol. 11:4374-485, 1991). This protocol can be improved by increasing the uptake of plasmid DNA into muscle and reducing vector degradation.
  • the procedure of the present invention uses MVS DNA coated with the relevant transcriptional regulatory factors, the human serum response factor and other human associated nuclear proteins, such as histones, and transcription initiation factors to enhance uptake and stability.
  • the regulatory proteins protect the DNA against muscle nucleases and facilitate the uptake of the protein coated DNA into myogenic nuclei.
  • the MVS forms a protein/DNA complex by the sequence specific binding of the serum response factor with the inner core CC(A/T) 6 GG of the serum response element and by the addition of histones.
  • the interaction with the inner core of the promoter facilitates myogenic cell type restricted expression of the skeletal alpha actin gene.
  • the serum response factor, transcription initiation factor, transregulatory factor and histones are added to the MVS by an in vitro binding reaction to form a reconstituted protein/DNA complex.
  • Myogenic vector SK733IGF-I-SK2 was injected at about 100 ⁇ g/100 ⁇ l into the gastrocnemius muscle of hypophysectomized BALB/C mice. The entire muscle was removed between 1 to 4 weeks following injection and assayed for IGF-I mRNA content in comparison to the uninjected contralateral limb. As compared to controls, tube injected limbs should increase levels of IGF-I mRNA due to the expression of the MVS.
  • IGF-I levels in comparison to controls injected with only the plasmid vector pSK-3'SK, as assayed by IGF-I radioimmunoassay.
  • Two animals injected with the SK7331IGF-I-SK2 vector doubled IGF-I levels as shown in Table III.
  • Example 7 Treatment with Growth Hormone
  • Growth hormone is produced and secreted from the anterior pituitary and promotes linear growth in prepuberty children.
  • the secretion of growth hormone is regulated by a stimulatory (growth hormone releasing hormone) and an inhibitory (somatostatin) hypothalamic hormone.
  • Growth hormone acts on the liver and other tissues to stimulate the production of insulin like growth factor I.
  • This factor is responsible for the growth promoting effects of growth hormone. Further, this factor serves as an indicator of overall growth hormone secretion. Serum IGF-I concentration increases in response to endogenous and exogenous administered growth hormone. These concentrations are low in growth hormone deficiency.
  • the injection of a MVS cont_-ining the sequence for IGF-I can be used to treat growth disorders.
  • the injection of the MVS is a long-term inexpensive way to increase systemic blood concentration of IGF-I in patients with growth hormone deficiency.
  • Example 8 Treatment of Muscle Atrophy Due To Age Growth hormone levels decline with increasing age. The levels in healthy men and women above age of 55 are approximately one third lower than the levels in men and women 18 to 33. The decline in growth hormone and IGI-I production correlate with the decrease in muscle mass, termed senile muscle atrophy, and increase in adiposity that occur in healthy human subjects. Administering growth hormone three times a week to healthy 61 to 81 year old men who had serum levels below those of healthy younger men increased the Serum IGF-I levels to within the range found in young healthy adults. This increase level led to increased muscle mass and strength and reduced body fat.
  • the convenient cloning sites in the Myogenic Vector System are used to construct MVS vectors containing human growth hormone cDNA sequence and/or the human growth hormone releasing (secretory) hormone.
  • the expression of an MVS driven growth hormone gene following intramuscular injections is another way to increase IGF-I serum levels.
  • the MVS expression of the growth factor releasing hormone (GHRH) might be more advantageous than the expression of either IGF-I or the growth hormone vectors transcripts.
  • GHRH Since GHRH is reduced in the elderly it appears to be responsible for the lack of GH secretion rather than the anterior pituitary capability of synthesizing growth hormone, thus the increased expression of GHRH from muscle would increase GHRH levels in systemic blood system and can allow for the natural diurnal secretion pattern of GH from the anterior pituitary. In this way, GHRH could act as the natural secretogogue allowing for elevated secretion or release of GH from the hypothalamus of the elderly.
  • Insulin-like growth factors are one of the key factors that potentiate muscle development and muscle growth. Myoblasts naturally secrete IGF-
  • I/IGF-II as well as its cognate binding proteins during the onset of fusion.
  • IGF genes In terminally differentiated muscle, signals propagated from passive stretch induced hypertrophy induce the expression of IGF genes. Many of the actions of IGFs on muscle result from interactions with the IGF-I receptor.
  • the receptor is a ligand activated tyrosine specific protein kinase.
  • Insulin-like growth factors are also known neurotrophic agents which maintain neuronal muscular synapses, neuron integrity, and neuronal cell life under neurodegenerative conditions. It has been possible to increase muscle growth under otherwise stringent growth conditions in myogenic cultures and transgenic mice by injecting a MVS containing the IGF-I sequence. The direct injections into adult muscle allows direct transfer of growth promoting gene constructions into muscle and thus provides postnatal gene therapy.
  • the MVS driven genes are relatively insensitive to the innerva tion state of the muscle, they provide a direct and rather broad application for remedying certain kinds of human muscle atrophies caused by spinal cord injuries and neuromuscular diseases. These diseases include the spinal cord muscular atrophies and aymtrophic lateral sclerosis (ALS).
  • ALS aymtrophic lateral sclerosis
  • the product of the MVS acts as a neurotrophic agent secreted from injected muscle and as a hypertrophic agent to maintain muscle integrity.
  • Example 10 Atherosclerotic Cardiovascular Diseases Atherosclerotic cardiovascular disease is a major cause of mortality in the United States and the world.
  • the atherosclerotic plaque the basic underlying lesion in atherosclerosis, contains cholesterol esters that are derived from circulating lipids. these circulating lipids are essential to the development of atherosclerosis.
  • the plasma concentration of high density Hpoprotein (HDL) is inversely related to the propensity for developing atherosclerosis. In the nascent state, HDL is secreted in the form of discoidal particles. These particles consist of a bilayer of phosphlipids onto which the apolipoproteins (apoA-I, ApoII and E) are embedded. HDL captures cholesterol esters by the action of an enzyme, lecithin-cholesterol acyltransferase. HDL is secreted from the liver, the small intestine and possibly other tissues.
  • the APO A-I cDNA is 878 bp and encodes 267 amino acids, including the 24 amino acid propropeptides. Increasing the circulating levels of HDL can influence or reverse cholesterol transport, and thus reduce the propensity for forming atherosclerotic plaques.
  • the insertion of the human APO A-I coding sequences into the MVS serves as an expression vector for enhanced APO A-I expression following injection of plasmid DNA into skeletal muscle.
  • the MVS APO A-I hybrid gene is effective for long term expression, biosynthesis and secretion of HDL in an ectopic site, and thus increases the content of total secretable HDL in blood plasma.
  • Example 11 Vitamin D Regulatable Myogenic Vector System The first tier of myogenic vectors designed with the skeletal actin promoters and 3' untranslated regions provide high levels of unregulated transcriptional activity. Under certain circumstances, it is desirable to control the vector's transcriptional activity and to switch gene transcription on and off by the systemic introduction of a simple inducer or ligand. It is also important that the regulation of the myogenic vector be controlled by natural inducer products that are neither considered toxic to humans nor are immunogenic. Two different Vitamin D regulatory systems are shown in Figures 8 and 9.
  • VDR Vitamin D receptor
  • SEQ. ID. No. 2 The target, SEQ. ID. No. 2, is constructed to contain synthesized multimers of the Vitamin D regulatory element (VDRE). This target is linked to a minimal Herpes Simplex Virus (HSV) thymidine kinase promoter. Transcriptional activity emanating from the TK promoter is regulated by the presence of VDR and coactivated by the ligand, Vitamin D.
  • VDRE Vitamin D regulatory element
  • HSV Herpes Simplex Virus
  • any polypeptide sequence cloned in tandem to the HSV promoter, as a cDNA, is driven from the target vector when Vitamin D is introduced into the muscle cells.
  • the hybrid actin VDR gene and the target vector are linked on the same plasmid or coinjected on separate plasmids. Premeasured levels of
  • Vitamin D are administered by drinking a glass of milk or taking a Vitamin D pill. The levels are used to activate transcription of the target vector. Taking the ligand on every other day, will oscillate the promoter activity. Removal of the ligand, Vitamin D, from the diet down regulates or represses transcription from the target vector.
  • the MVS system is well suited for directing the expression of an exogenous protein epitope in muscle, and thus, for generating vaccines in humans and animals.
  • Targeted sequences are inserted into the cassette of a
  • MVS for expression of protein epitopes for mediating protective immunization.
  • the constant regions of the AIDS viral proteins are included in the constant regions of the AIDS viral proteins.
  • GP 120, GP 160 and GP 41 and for cell mediated immunity GP 24 the surface proteins of Hepatitis A, B, C, and C viruses, respiratory viruses including influenza, and the gastrointestinal rotoviruses are used.
  • the MVS is then injected into the human or animal.
  • MOLECULE TYPE DNA (genomic)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Magnetic Resonance Imaging Apparatus (AREA)
  • Eye Examination Apparatus (AREA)

Abstract

The present invention includes a Myogenic vector system (MVS) for the expression of a nucleic acid sequence in myogenic tissue. The MVS contains a promoter, a cassette with the sequence of interest, a 3' untranslated region (3' UTR) and contiguous noncoding region (NCR). Further enhancements can include the addition of a leader sequence, intron sequence, initiation codon and specific restriction endonucleases. To facilitate uptake and myogenic expression, the MVS can be coated with histones and a DNA initiation complex composed of a serum response factor, transcription initiation factor and a transregulatory factor attached to the promoter by interaction with the serum response element and TATA box. The MVS can be used for a variety of purposes including gene replacement, vaccine production, vaccination, treatment of disease and muscle atrophy. When the MVS is injected into a myogenic system it will produce the polypeptide from the sequence of interest.

Description

MYOGENIC VECTOR SYSTEMS
x' This invention was partially supported by a grant from the United
_
States government under HL-38401 awarded by the National Institute of Health. The Government has certain rights in the invention.
5 Field of the Invention
The present invention relates generally to expression vectors for use in expressing polypeptides in myogenic cells. More particularly it relates to vectors containing the skeletal alpha actin gene promoter and the corresponding 3' transcribed but untranslated region of the gene and the 10 contiguous noncoding DNA containing the gene's natural transcriptional termination region.
Background of the Invention
Actin is a contractile protein found in most cell types. Actin proteins are represented by similar but nonidentical isoforms which in warm blooded
15 vertebrates are encoded by multigene families. Actin is expressed in a developmentally timed dependent and tissue specific manner, in which adult muscle tissues express exclusively the skeletal alpha actin gene as the predominant actin isoform. The nucleic acid sequence of skeletal alpha actin gene has been characterized in chicken, rat, mouse and human. Fornwald et
20 al., Nucleic Acids Res., Vol. 10, pp. 3861-3876 (1982); French et al., Gene, Vol.
88, pp. 173-180 (1990); Zak et al, Nature, Vol. 298, pp. 857-859 (1982); Hu et al, Mol. Cell. Biol., Vol. 6, pp. 15-25 (1986) and Minty and Kedes, Mol. Cell. Biol., Vol. 6, pp. 2125-2136 (1986).
Once a gene has been characterized and sequenced it can be mapped
•*.
25 to determine its protein coding and noncoding portions. RNA transcripts are initiated from a promoter region start site at the messenger RNA cap and continue through the protein coding region to the noncoding region involved with transcriptional termination. Post transcriptional processing of the RNA transcripts removes the noncoding introns to form a continuous coding sequence. Further, there is progressive trimming of the RNA transcript to the polyadenylation signals, found in the noncoding 3' untranslated portion of the processed messenger RNA. The levels of messenger RNA biosynthesis and accumulation is determined by parts of the unexpressed portion of the gene. For example the promoter, which is part of the unexpressed portion of the gene, is involved in determining gene expression. It regulates when and how the expressed gene is transcribed. The accumulation of RNA transcripts, however, may relate to the intrinsic stability of that RNA in a particular cell type. This intrinsic stability is dependent on sequences within the mRNA that provide stabilization. Thus, the ability to express a particular gene product is a balance between transcription rate and the stability of the mRNA transcript. Expression of the actin gene family is tissue specific. The skeletal alpha actin protein is expressed primarily in cardiac and skeletal muscle tissues. Transient transfection experiments indicated that regions within the 200 bp promoter region are sufficient for tissue restricted expression in primary myoblasts. Bergsma, et al, Mol. Cell. Biol., Vol.,6 pp. 2462-2475 (1986) and Chow and Schwartz Mol. Cell Biol., Vol.10 pp. 528-538 (1990).
Further, the promoter region harbors conserved cis-acting elements to accurately initiate skeletal alpha actin transcripts from the bacterial reporter gene chloramphenicol acetyltransferase (CAT) in differentiating myoblasts. Grichnik et al. Nucleic Acids Res., Vol. 14, pp. 1683-1701 (1986). Transgenic mice with an integrated skeletal alpha actin promoter showed preferential expression of the CAT gene in myogenic tissues Petropulous et al. Mol. Cell. Biol., Vol. 9 pp. 3785-3792 (1989). It is also known that CAT activity is detectable as early as 10 days in the mouse embryo when the embryonic heart is first being formed. Further, CAT activity can be induced in newborn skeletal muscle. Thus, the skeletal actin promoter can switch transcriptional activity in mammalian systems, to provide muscle restricted expression. However, when measurements were made to compare the levels of CAT messenger RNA to that of the endogenous skeletal alpha actin messenger RNA (which in the adult muscle was found at levels over thousand fold higher than the embryo), the CAT messenger RNA could not be detected by standard RNA blotting techniques. These experimental results showed that even though small amounts CAT protein accumulated in skeletal muscle, the CAT messenger RNA never accumulated to the levels of RNA transcripts from the endogenous skeletal actin gene.
Current trends for optimizing gene transcription vectors have been directed towards increasing the transcriptional activity of the vector system. For example, Observations in the human Beta globin gene system indicated that expression in transgenic mice was never as high as the expression of the endogenous mouse Beta globin gene. Grosveld and co-workers Cell, Vol. 51, pp 975-985 (1987) It was determined that the sites surrounding the globin locus contain a number of DNAse hypersensitive sites that are termed the dominant control region. This region appears to act as tissue specific enhancers. When some of these sites were cloned into a minilocus gene it provided erythroid tissue specific expression in transgenic animals. Regulatory sequences similar to the dominant control regions have been described for human CD2. Greaves et al. Cell, Vol. 56, pp. 979-986 (1988). It is known that the avian skeletal actin promoter is as active as the SV40 promoter which is a standard for high levels of expression. Bergsma, et al,
(1986). Hence, the skeletal actin promoter is not the cause for low levels of messenger RNA accumulation. Thus, the actin promoter is by itself insufficient to drive the expression of other gene products to accumulate at levels which are comparable to the intact skeletal alpha actin mRNA. The rate of metabolic breakdown of mRNA molecules is an important factor in the regulation of gene expression. The rates of decay of individual mRNA species can affect strongly the steady state levels of these species in the cytoplasm. Consequently, the extent of expression of a given gene, as measured by the rate of synthesis of the corresponding protein, will be dependent to a large extent on the degree of stability of the mRNA derived from this gene. Messenger RNA from skeletal muscle was previously shown to be distributed into two populations with regard to its stability. Medford et al. J. Biol. Chem., Vol. 258, pp. 11063-11073 (1983). One mRNA population had a half life of less than 4 hours and the other population had a half-life of 17-to over 54 hours.
Comparison of the untranslated regions in vertebrate skeletal alpha, cardiac alpha, and beta actin mRNA has revealed regions of high sequence homology within the 3 ' untranslated portion of each of these actin isoformic mRNA and that this homology is greater among the alpha-cardiac and skeletal actin isoforms than between alpha striated actin and beta actin isoform mRNA. Mayer et al., Nuc Acids Res., Vol 12. pp. 1087-1100 (1984); Ponte et al., Nucl. Acids Res., Vol. 12 pp. 1687-1696 (1984); Chang et al., Nucl. Acids Res., Vol. 13 pp. 1223-1237 (1985). In comparison, other vertebrate genes, such as those encoding insulin and prolactin share common coding regions but usually contain divergent 3' untranslated regions. The preservation of the 3' untranslated regions of the skeletal alpha actin gene in animal species ranging from birds to humans suggests that they have important biological roles. In the present invention, the incorporation of the myogenic specifics 3' untranslated region into recombinant DNA vectors for expressingpolypeptides in muscle tissue was found to be highly advantageous, since it enhanced the polypeptide mRNA content in muscle by increasing stability. Siimmary of the Invention
An object of the present invention is a myogenic vector system capable of expressing any specific nucleic acid sequence in myogenic tissue.
An additional object of the present invention is a regulatable myogenic vector system. A further object of the present invention is a method for treating muscle atrophy in aging humans.
Another method of the present invention is a method for treating muscle atrophy induced by spinal cord injuries or muscular disease.
A further object of the present invention is a method of preventing or treating atherosclerotic cardiovascular disease.
An additional method of the present invention is the introduction of a myogenic vector system for gene replacement. A further object of the invention is a method for vaccine production in human or animals.
Another object of the present invention is a method of treating growth disorders. Thus, in accomplishing the foregoing objects, there is provided in accordance with one aspect of the present invention a myogenic vector system (MVS) for the expression of a nucleic acid sequence in myogenic tissue comprising a promoter; a cassette whose 5' end is attached to the 3' end of the promoter, the cassette containing a nucleic acid sequence which is to be expressed; a myogenic specific 3' untranslated region (3' UTR) and a non¬ coding region (NCR) contiguous to the 3' end of the 3' UTR, the NCR containing a transcriptional termination signal, wherein the 5' end of the 3' UTR is attached to the 3' end of the cassette.
Specific embodiments of the MVS of the present invention can further include a leader sequence following the promoter region. Additional embodiments of the MVS include a first intron, an initiation ATG and Ncol cloning site inserted between the promoter leader region area and the cassette area. Specific embodiments of the cassette include an EcoRl site at the 3' end. The 3' UTR can have an EcoRV site at its 5' end. Any promoter will work although in the preferred embodiment a myogenic promoter selected from a group consisting of skeletal alpha actin gene promoter, first myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter/enhancer, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR. In the preferred embodiment the skeletal alpha actin promoter is used.
Similarly, the 3' UTR and NCR are selected from any myogenic specific genes. In the preferred embodiment the skeletal alpha actin gene 3' UTR and NCR regions are used.
Specific embodiment also include the addition of regulatory promoter elements to regulate the expression of any specific nucleic acid sequence in myogenic tissue. In the preferred embodiment, Vitamin D is used to regulate expression. treatment of prevention of atherosclerotic cardiovascular disease, gene replacement, and vaccine production.
Other and further objects features and advantages will be apparent from the following description of the presently preferred embodiments of the invention, which are given for the purpose of disclosure, when taken in conjunction with the accompanying drawings.
Brief Description of the Drawings
Figure 1 is a schematic drawing of the chicken skeletal alpha actin gene which includes location of unique restriction sites. Figure 2 illustrates the transcriptional domain of the avian skeletal alpha actin gene and the contiguous noncoding region where transcription terminates (OD»mm/kb Insert Relative GAPDH).
Figure 3 is a schematic representation of a myogenic vector system. Figure 4 is a schematic diagram of skeletal alpha actin/insulin like growth factor-I hybrid genes
Figure 5 illustrates the increased activity of the myogenic IGF-I hybrid vectors containing the skeletal alpha actin 3' untranslated region and contiguous noncoding region by the accumulation of IGF-I RNA in stably transfected C2C12 myoblasts. Figure 6 shows the accumulation of IGF-I RNA in transgenic mouse lines generated with SK202IGF-I-3'SVa myogenic expression vector.
Figure 7 shows the accumulation of IGF-I RNA in transgenic mouse lines generated with SK202IGF-I-3'SK myogenic expression vector.
Figure 8 is a schematic representative of a regulatable MVS using a Vitamin D receptor.
Figure 9 is a schematic representative of a regulatable MVS using a chimeric receptor.
-6-
SUBSTITUTE SHEET Figure 10 illustrates the stability of IGF-I mRNAs with different 3' UTRS. The drawings are not necessarily at this scale. Certain features of the invention may be exaggerated in scale or shown in schematic form in the interest of clarity and conciseness.
DETAILED DESCRIPTION OF THE INVENTION It will be readily apparent to one skilled in the art that various substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention.
The term "promoter" as used herein refers to a recognition site on a DNA strand to which the RNA polymerase binds. The promoter usually is a DNA fragment of about 100 to 200 bp in the 5' flanking DNA upstream of the cap site or the transcriptional initiation start site. The promoter forms an initiation complex with RNA polymerase to initiate and drive transcriptional activity. The complex can be modified by activating sequences termed "enhancers" or inhibiting sequences termed "silencers." Usually specific regulatory sequences or elements are embedded adjacent to or within the protein coding regions of DNA. The elements, located adjacent to the gene, are termed cis-acting elements. These signals are recognized by other diffusible biomolecules in trans to potentiate the transcriptional activity. These biomolecules are termed trans-acting factors. The presence of trans¬ acting factors and cis-acting elements have been shown to contribute to the timing and developmental expression pattern of a gene. Cis acting elements
-7-
SUBSTITUTE SHEET are usually thought of as those that regulate transcription and are found within promoter regions and other upstream DNA flanking sequences.
The term "leader" as used herein refers to a DNA sequence at the 5' end of a structural gene which is transcribed along with the gene. The leader usually results in the protein having an N-terminal peptide extension sometimes called a pro-sequence. For proteins destined for either secretion to the extracellular medium or a membrane, this signal sequence, which largely hydrophobic, directs the protein into endoplasmic reticulum from which it is discharged to the appropriate destination. The term "intron" as used herein refers to a section of DNA occurring in the middle of a gene which does not code for an amino acid in the gene product. The precursor RNA of the intron is excised and is therefore not transcribed into mRNA nor translated into protein.
The term "cassette" refers to the sequence of the present invention which contains the nucleic acid sequence which is to be expressed. The cassette is similar in concept to a cassette tape. Each cassette will have its own sequence. Thus by interchanging the cassette the vector will express a different sequence. Because of the restrictions sites at the 5' and 3' ends, the cassette can be easily inserted, removed or replaced with another cassette.
The term "3' untranslated region" or "3' UTR" refers to the sequence at the 3' end of a structural gene which is usually transcribed with the gene. This 3' UTR region usually contains the poly A sequence. Although the 3' UTR is transcribed from the DNA it is excised before translation into the protein. In the present invention it is preferred to have a myogenic specific 3' UTR. This allows for specific stability in the myogenic tissues.
The term "Non-Coding Region" or "NCR" refers to the region which is contiguous to the 3' UTR region of the structural gene. The NCR region contains a transcriptional termination signal.
The 3' UTR and NCR are a key aspect of the present invention because they provide a higher level of mRNA accumulation through increased mRNA stability in myogenic cells rather than non-myogenic cells. Thus, this increased stability of mRNA leads to the increased levels of protein production.
The 3' untranslated region of the chicken skeletal alpha actin gene which starts at nucleotide 2060 and extends to 2331. The complete 3' untranslated region and contiguous noncoding DNA extends an additional 2.0
Kb. This 2.3 Kb fragment can be linked immediately following the natural translation termination codon to a copy DNA sequence coding for a polypeptide desired to be expressed.
The term "myogenic" or "myogenic specific" refers to muscle tissue. The muscle tissue can be in vivo tissue, in vitro tissue or in vitro tissue cultures of cells capable of differentiating into muscle tissue. Myogenic cells include skeletal, heart and smooth muscle cells. These vectors are transfected into myogenic cells in culture, and are injected into intact muscle tissue.
These vectors containing the construction are injected into mammalian oocytes and may be stably incorporated into the genome to generate transgenic animals, in which the vector expresses polypeptides in myogenic cells.
The term "restriction site" refers to a sequence specific cleavage site of restriction endonucleases. The term "vector" refers to some means by which DNA fragments can be introduced into a host organism or host tissue. There are various types of vectors including plasmid, bacteriophages and cosmids.
The term "effective amount" means sufficient MVS is injected into myogenic tissue or culture to produce the adequate levels of the polypeptide. One skilled in the art recognizes that this actual level will depend on the use of the MVS. The levels will be different in treatment, vaccine production, or vaccination.
In one embodiment of the present invention there is a myogenic vector system (MVS) for the expression of a nucleic acid sequence in myogenic tissue, comprising a promotor; a cassette whose 5' end is attached to the 3' end of the promoter; a myogenic specific 3' UTR and a NCR contiguous to the 3' end of the 3' UTR. This basic system can be enhanced in a variety of ways including the addition of a leader sequence between the promoter and cassette.
In a preferred embodiment the MVS for expression of a nucleic acid sequence in myogenic tissue comprises a functional unit which expresses the nucleic acid sequence. The functional unit is comprised of elements all sequentially linked together 5' to 3'. The elements in the order of linkage include a promoter, a 5' mRNA leader sequence, a first intron and initiation ATG and Ncol cloning site, a cassette which has a EcoRI site at its 3' end, a myogenic specific 3' UTR in which the 5' end of the 3' UTR has an EcoRV site, and a NCR which is contiguous to the 3' end of the 3' UTR.
A variety of promoters can be used in the MVS. Some examples include skeletal alpha actin gene promoter, first myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR. In the preferred embodiment, a myogenic specific promoter such as the skeletal alpha actin gene promoter is used.
The 3' UTR and NCR can be selected from any group of myogenic specific genes. Examples of genes in this group are the skeletal alpha actin gene, fast myosin light chain 1 gene, myosin heavy chain gene, tropinin T gene, acetyl choline receptor subunit genes and muscle creatinine kinase gene.
In the preferred embodiment, the 3' UTR and NCR are from the skeletal alpha actin gene.
Alternate embodiments of the present invention include the addition of a regulator system for regulating the expression of nucleic acid sequence. Any of a variety of regulator systems can be used. In the preferred embodiment, two different regulatory systems have been used.
One embodiment of a regulated MVS (see Figure 7) for the expression of a specific nucleic acid sequence in myogenic tissue, is comprised of a first functional unit and a second functional unit. The first functional unit and second functional unit can be in the same vector or in two separate vectors.
In either case both functional units must be introduced into the myogenic tissue. The first functional unit is composed of the following elements all sequentially linked 5' to 3': a myogenic specific promoter, a nucleic acid sequence coding for a receptor, a myogenic specific 3 ' UTR and a myogenic specific NCR. The second functional unit is composed of the following elements sequentially linked 5' to 3' : a response element corresponding to the receptor, a thymidine kinase promoter, a cassette containing the specific nucleic acid sequence of interest, a myogenic specific 3* UTR and a contiguous myogenic specific NCR. In this regulatable MVS, it is preferable for the first functional unit to continuously express the receptor. It is preferable to use a receptor which is not found in high levels in myogenic tissue, when the agent which is specific to the receptor is introduced into the system. The receptor forms an interaction with the response element and the specific agent. This binding interaction causes the thymidine kinase promoter to express the specific nucleic acid sequence. By regulating the amount of agent which is present, The activity of the MVS is regulated.
In this regulatable system, the response element and receptor are usually complimentary and can be selected from a variety of receptor groups. For example, any vitamin, steroid, thyroid, orphan, hormone, retinoic acid and thyroxine can be used. In the preferred embodiment, the Vitamin D receptor and Vitamin D response element are used. In this case, ingestion of Vitamin D, for instance by drinking milk, causes the level of Vitamin D in the blood. This binds to the receptor being generated in the myogenic tissue and the complex binds to the receptor element causing the expression of the gene of interest. Thus, the MVS can be regulated by dietary intake.
An alternate method of regulating the MVS is shown in Figure 9. In this embodiment, at least one of the serum response elements in the alpha actin promoter region is made into a receptor binding site. Then a chimeric transfactor is constructed in which the normal DNA binding domain of the serum response factor is replaced with the DNA binding domain of the receptor. The transactivation domain of the serum response factor is not changed. Thus, when an agent or ligand specific to the receptor is present it binds to the receptor in the binding domain allowing the serum response transactivation factor to activate transcription. Thus, the regulation can be controlled by controlling the amount of the agent. The DNA binding domain which is substituted into the normal serum response factor is usually selected from the following families of receptors binding domains: the vitamin, steroid, thyroid, orphan, hormone, retinoic acid and thyroxine. In the preferred embodiment, the Vitamin D receptor is used.
The cassette can contain the nucleic acid sequence of interest and include any nucleic acid sequence which is to be expresses in the myogenic tissue or tissue culture. These nucleic acid sequence can call for any variety of polypeptides. The polypeptide can be any desired polypeptide including but not limited to known proteins. For example, the sequence could code for a hormone, a growth factor, an enzyme, an apolipoprotein, tumor suppressor, tumor antigen, a viral protein, a clotting factor and any proteins associated with the AIDS virus, any other viral proteins, including viral surface coat proteins, bacterial surface proteins, parasitic cell surface proteins, viral reverse transcriptase and any gene which needs to be replaced by gene replacement.
In specific embodiments of the MVS, the cassette included the nucleic acid for the insulin like growth factor I, or the insulin like factor II or the insulin growth binding protein. This specific embodiment can be used to treat muscle atrophy in aging humans, muscle atrophy induced by spinal cord injuries or neuromuscular diseases. A specific example of the latter case would be amyotrophic lateral sclerosis. Another specific embodiment is a MVS where the cassette contains the nucleic acid sequence coding for growth hormone releasing factor. This MVS can be used for treating muscle atrophy in aging humans.
Another embodiment of the present invention includes an MVS where the cassette includes the nucleic acid sequence for apolipoprotein A-I. This MVS can be used for prevention or treatment of atherosclerotic cardiovascular disease.
In those instances where the MVS cassette contains a sequence coding for a viral, bacterial or parasitic protein, the MVS can be used to make a vaccine. In this the procedure for making a vaccine humans or animals, comprising the step of injecting an effective amount of the MVS into skeletal muscle or into tissue culture. In this MVS vector the cassette contains a nucleic acid sequence which codes for a polypeptides capable of eliciting an antibody response. This procedure would be a safe and effective method for generating vaccines. The vaccine can be generated in tissue culture, or in vivo in humans or animals. Examples of nucleic acid sequences which can elicit an antibody response include those for the viral proteins, bacterial proteins, and parasitic proteins. A specific case example is the AIDS proteins.
Another specific embodiment of the present invention is a method of treating growth disease which comprises the step of injecting an effective amount of an MVS into skeletal muscle wherein the nucleic acid sequence in the cassette contains the growth hormone sequence. Another application of the present invention is a method for gene replacement. In this embodiment, an effective amount of the MVS is injected into skeletal muscle and the cassette contains any sequence which codes for a defective gene. For example, the genes for glycogen phosphorylase, alpha- 1- antitrypsin and dystrophin can be inserted into the cassette. Individuals with the corresponding diseases of glycogen storage disease, alpha-1-antitrypsin deficiency or pulmonary emphysemia and Duchenne's Muscular Dystrophy would thus produce a sufficient amount of the normal polypeptide. The expressed protein may systemically spread throughout the animal via the circulatory blood system as well as remain in the injected musculature. The polypeptide may be harvested and purified as desired.
Any of the MVS's discussed herein can be further modified to enhance uptake by the cell. This enhancement comprises adding a coating. The coating includes a DNA initiation complex and histones. The initiation complex comprises a serum response factor (SRF), a transcription initiation factor (TIF) and a transregulatory factor (TRF). The SRF is attached to the serum response element within the promoter region of the MVS. The TIF and the TRF then interact with the SRF and the TATA box within the promoter to form a stable DNA complex. The histones bind nonspecifically to the remaining DNA in the MVS.
The following examples are offered by way of illustration and are not intended to limit the invention in any manner.
Example 1
Isolation of the Chicken Skeletal Alpha Actin Gene The 25 Kb EcoRI fragment of chicken genomic DNA isolated from a lambda Charon 4A vector, contains the 6.2 Kb skeletal alpha actin gene on a single Hind III site of pBR322 is shown in Figure 1. Chang et al. Mol. Cell. Biol. Vol 4:2498-2508 (1984). Nuclear transcription runoffs were used to map the transcriptional domain of the skeletal alpha actin gene (Figure 2). DNA probes which encompassed portions of the 5' noncoding, promoter, coding, and the contiguous 3' noncoding regions were cloned into M13 vectors which provided sense and antisense probes. Nuclei isolated from fibroblasts, myoblasts and day 19 embryonic muscle cells were used in in vitro transcription assays to extend RNA transcripts with radioactive tagged nucleotides. Labeled RNA hybridized to dotted DNA probes showed that transcription terminates approximately 1 kb downstream of the skeletal alpha actin gene's poly A addition site. This is within a 800 bp Pvu II fragment between +2800 and +3600 nucleotides from the start of transcription.
The 3' untranslated region (3' UTR) and the contiguous noncoding region (NCR) can be isolated by restriction endonucleases digestion of the 6.2 Kb actin gene with blunt cutter Nae I, which cuts 30 bp upstream of the translation termination codon TAA. Hind III releases the 3' most portion of the actin gene from the vector pBR322 (Figure 3). The 3' UTR and NCR were used to prepare DNA constructs. The skeletal alpha actin promoter and DNA flanking sequences (at least 411 nucleotides from the mRNA cap site) and DNA sequences extending through the skeletal 5' noncoding leader, first intron and up to the initiation of translation ATG, converted to a Nco I cloning site at +196, was liberated from a M13 double stranded DNA by Xba
I and Ncol digestion, Eϋenow filled in and then linked into the Xbal and blunt Smal sites of pBluescript II KS (Stratagene). The NCoI site is regenerated by this cloning step. The 3' UTR and NCR on the 2.3 kb Nael/Hindlll fragment were directionally cloned into a blunt EcoRV site and the adjacent Hindlll site of the pBluescript II KS vector cassette. The EcoRV and Nael sites are destroyed. The restored Nco I site was used to insert cDNA sequences encoding polypeptides. Another cloning vector was constructed by inserting the skeletal alpha actin promoter from -411 to -11 adjacent to the 3' UTR and NCR. This myogenic vector eliminates the first intron and the skeletal actin 5' leader sequence. These two vectors were used in preparing DNA constructs to test the efficacy of the 3' UTR and NCR.
Example 2
Construction of a MVS Containing' the Human IGF-I Constructions containing the skeletal actin promoter were linked to the human IGF-I cDNA by standard recombinant DNA techniques. Maniatis, Fritsch and Sambrook, Molecular Cloning. A laboratory manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1982. Examples of a generalized MVS structure is shown in Figure 3. Specific construction IGF-I are shown in Figure 4. The construction was made so that the SV40 poly A addition site and the small t-intron were linked to the 3' untranslated region of the IGF-I cDNA. The SV40 sequences were added to increase the stability of nuclear IGF-I RNA transcripts. Since the SV40-t-intron might not be entirely suitable in the expression of IGF-I in muscle cells, five other vectors were made. The SK733 NcoI vector contains approximately 411 nucleotides of the skeletal alpha actin promoter, the natural cap site, 5' untranslated leader and the first intron. An NcoI site was engineered to create a unique insertion cloning site for the cassette containing the IGF-I cDNA, in which the initiation ATG was also converted to an NcoI site. The SK733IGF-I construction utilizes its own poly A site. An Nael/Hindlll fragment which incorporated the skeletal alpha actin 3' untranslated region, poly A addition site, and terminating sequences was linked to SK202, SK733 NcoI, IGF-I and to SK733IGF-I which the IGF-I poly A site was deleted and replaced by that of skeletal alpha actin. In this way IGF-I RNA transcripts containing the skeletal alpha actin 3' untranslated region are stabilized and accumulate in skeletal muscle cells. In addition, by providing contiguous 3' noncoding DNA, IGF-I is buffered against outside genomic sequences and is thus more protected from position effects, when integrated into the genome. In addition, by providing natural terminating sequences, the additional regulatory sequences that mark the transcriptional domain of skeletal alpha actin improve tissue specificity, developmental timing and transcriptional activity.
Example 3 Activity of MVS Constructs To determine the efficacy of actin promoter/gene IGF-I hybrid genes in mouse myogenic cells the MVS was studied using these genes in the background of mammalian C2C12 myoblasts by making a population of stable transfected C2C12 myoblasts. The altered IGF-I expression levels were directly evaluated in these stablemyoblast cell lines. Each IGF-I construction (Figure 4) was co-transfected with the drug selectable vector EMSV-Hygromycin into mouse CJJC-J, cells. After two weeks of selection, a population of stable myoblasts was selected. A population of C2C12 myoblasts stably transfected only with EMSV-Hygromycin served as the controls. Visual inspection of the transfected myoblast revealed several insights into the role of IGF-I on muscle cell differentiation that would not be obvious in transgenic mice. In general all of the myogenic cell lines containing IGF-I genes caused myoblasts in growth media (10% fetal calf serum) to replicate more extensively than controls. Changing culture medium to 2% horse serum initiates the differentiation process. In the process, control C2C12 myoblasts fuse to form multinucleated myotubes over a period of four days. At the same cell density per culture dish, myoblasts containing SK733IGF-I, SK202IGF-I-SK,
SK733IGF-I-SK1 and SK733IGF-I-SK2 fused at least two-to-three days earlier than C2C12 or EMSV-Hygromycin control myoblasts.
Figure 5 shows the steady state accumulation of IGF-I mRNA in C2C12 myoblasts. Equal amounts of total cellular RNA was isolated from stably transfected C2C12 myoblasts grown in growth media (G) or differentiation media (D) . The RNA was electrophoretically separated on denaturing agarose gels, transferred onto nylon filters and probed with uniformly ^P labeled full length human IGF-I cDNA under standard hybridization techniques. The intensity of the autoradiographic signal on X-ray film provides a relative measure of mRNA accumulation, an overall index of combined transcriptional activity and mRNA stability of the myogenic expression vector system. The IGF-I mRNA in vector, SK202IGF-I-3'SVa did not accumulate in myotubes above myoblast levels. This is a typical expression activity. The SK733IGF-I vector contains the IGF-I 3' untranslated region. The IGF-I mRNA from this vector accumulated in myotubes but at levels substantially lower than SK202IGF-I-SK or SK733IGF-I-SK2. These later two vectors contain the skeletal actin 3' UTR and contiguous noncoding regions. Since, the primary difference in these vectors is the 3' UTR, the increased stabilization of the mRNA transcripts due to the skeletal 3' UTR accounts for about a 100 fold difference in mRNA content.
Example 4 Measurement of Secreted Levels of IGF-I from MVS
Differentiated myotube cultures were grown in minimal media (DMEM and 0.05% bovine serum albumin RIA grade) in order to measure the amount of IGF-I synthesized and secreted into the media. SK733IGF-I-SK2 is the most effective construction to express IGF-I in muscle cells. IGF-I was assayed by both radioimmunoassays of tissue culture media and by immunoperoxidase staining of cells. We have found increased levels of IGF-I during the fusion of several of our muscle cultures. The comparison of levels from different MVS are shown in Table 1. In control cultures, the level of IGF-I was in the range of 0.2-0.5 ng/ml. In comparison, vector SK733IGF-I-SK2 has levels of IGF-I at least one hundred times greater.
The role of the skeletal alpha actin 3' UTR on mRNA stability was examined in the stable population of transfected C2C12 myogenic cells. A transcription blocker, actinomycin D (8 ug/ml) was added to differentiated myogenic cultures at the myotube stage and timed samples were removed for RNA blotting analysis to determine relative amounts of IGF-I mRNA.
Transcripts containing the natural human IGF-I 3' UTR were found to turn over rapidly with a half life of less than 1 hour. These results are consistent with the short half life of growth factors. IGF-I transcripts containing SV40 3' UTR and poly A addition signals displayed a half life of about 4 hours. In contrast, IGF-I transcripts containing the skeletal alpha actin 3' UTR showed a high level of stability with a half life greater than 18 hours. These results support the claim that the high level of IGF-I mRNA that accumulates in the
SK733IGF-I-SK2 transfected C2C12 myoblasts is due in part to the differential stabilization of mRNA which contains the contiguous skeletal alpha actin 3' UTR.
Table I IGF-I Levels in Stably Transfected C _. Myoblasts
Figure imgf000020_0001
In a similar manner, immunoperoxidase staining of myogenic cultures revealed the increased production of immunological reactive IGF-I in stable transfected myoblasts but not in the control EMSV-Hygromycin transfected myoblasts or in perfusϊon C2C12 cells. Antibodies against the A and D regions were used at dilutions of 1:1000. All of the transfected lines including SK202IGF-I were positively immunoperoxidase stained. Thus, it is clear that enhanced levels of IGF-I are being synthesized and exported from the stable myoblasts.
Example 5
Insertion of MVS into Transgenic Mice
Transgenic mice carrying SK202IGF-I-3'SVa or SK202IGF-I-SK were generated by standard oocyte injection (Brinster, et al, Proc. Natl. Acad. Sci.
USA Vol 82:4438-4442 (1958)) and bred to demonstrate stable transmission of transgenes to subsequent generations. Transgenics were identified by polymerase chain reaction or Southern genomic DNA blotting analysis from tail cut DNA. Transgenics were tested for muscle specific expression of the transferred IGF-I vector by RNA blotting of total RNA isolated from several tissues as shown in Figures 6 and 7. Independent transgenic mouse lines 5484, 5496, 5832, 5834 were generated with SK202IGF-I-3'SVa, containing the SV40 3' intron and poly A addition sequence. Mice from these strains were found to have weak expression primarily in heart tissue, but very low levels were found in skeletal muscle and non-myogenic tissues such as the kidney and brain (Figure 6). Independent transgenic mouse lines 3357, 3359 generated with SK733IGF-I-3'SK2. Mice from these strains were found to have elevated expression levels of IGF-I (Figure 7). These levels are comparable to the endogenous mouse alpha actin gene activity. These levels from SK733IGF-I-3'SK2 show at least 100-1000 fold greater accumulation of IGF-I mRNA in comparison to the levels produced by the SK202IGF-I-3'SVa vector. The addition of the skeletal alpha actin 3' UTR and contiguous noncoding regions allowed for a preferential increase in IGF-I RNA in skeletal muscle rather than cardiac. Thus, the 3' UTR and NCR of the skeletal actin have an important role in enhancing muscle specific gene expression.
Muscle tissue homogenates were analyzed by radioimmunoassay to determine the relative levels of IGF-I production. Homogenates were acid chromatographed to separate IGF-I from IGF binding proteins after acid chromatography fractions containing IGF-I were pooled, lyophilized, reconstituted and assayed by the protocol of Liu, F, Powell, D.R., Styne, D.M. and Hintz, R.L. (191) J. Clin. Endocrin. Metab. 72:905-911. The IGF-I antiserum was provided by the National Hormone and Pituitary Program.
In Table II, we detected a ten fold increase in the content of IGF-I in transgenic SK 7333 IGF-I SK muscle in comparison to controls. Thus, there is good correspondence between the increased IGF-I mRNA content with over an order of magnitude increase of expressed IGF-I in transgenic mouse muscle tissue. Table II
Muscle Content of IGF-I in Transgenic Mice IGF-I (100 ng/lOOug of muscle protein)
Control 11.5
SK 733 IGF-I-3' SK2 105.5 mouse line 3357
Example 6 Expression of MVS into Intact Muscle Intact plasmid DNA in a sterile 20% sucrose solution (wt/vol) can be injected into mature avian or mammalian muscle. Following a single injection the vector DNA is stable for at least 30 days as a non-integrated extrachromosomal circular DNA in muscle nuclei and, is transcriptionally active. Wolf et al., Science, 247: 1465-1468 (1990). However, greater than 99% of the injected DNA is degraded in muscle under the Wolff protocol (Wolff, et al, BioTechniques Vol. 11:4374-485, 1991). This protocol can be improved by increasing the uptake of plasmid DNA into muscle and reducing vector degradation. The procedure of the present invention uses MVS DNA coated with the relevant transcriptional regulatory factors, the human serum response factor and other human associated nuclear proteins, such as histones, and transcription initiation factors to enhance uptake and stability.
The regulatory proteins protect the DNA against muscle nucleases and facilitate the uptake of the protein coated DNA into myogenic nuclei.
The MVS forms a protein/DNA complex by the sequence specific binding of the serum response factor with the inner core CC(A/T)6GG of the serum response element and by the addition of histones. The interaction with the inner core of the promoter facilitates myogenic cell type restricted expression of the skeletal alpha actin gene. The serum response factor, transcription initiation factor, transregulatory factor and histones are added to the MVS by an in vitro binding reaction to form a reconstituted protein/DNA complex.
One method of post-natal gene therapy involves injecting myogenic vectors into adult muscle for the express purpose of expression of a particular polypeptide. Myogenic vector SK733IGF-I-SK2 was injected at about 100 μg/100 μl into the gastrocnemius muscle of hypophysectomized BALB/C mice. The entire muscle was removed between 1 to 4 weeks following injection and assayed for IGF-I mRNA content in comparison to the uninjected contralateral limb. As compared to controls, tube injected limbs should increase levels of IGF-I mRNA due to the expression of the MVS.
Injection of 20 ug of SK73IGF-I-SK2 plasmid DNA into the vastuslateralis muscle of Little mice caused an average increase of 60% in
IGF-I levels in comparison to controls, injected with only the plasmid vector pSK-3'SK, as assayed by IGF-I radioimmunoassay. Two animals injected with the SK7331IGF-I-SK2 vector doubled IGF-I levels as shown in Table III.
Table III Somatic Gene Transfer to Skeletal Muscle of Little Mice
Figure imgf000023_0001
Example 7 Treatment with Growth Hormone Growth hormone is produced and secreted from the anterior pituitary and promotes linear growth in prepuberty children. The secretion of growth hormone is regulated by a stimulatory (growth hormone releasing hormone) and an inhibitory (somatostatin) hypothalamic hormone. Growth hormone acts on the liver and other tissues to stimulate the production of insulin like growth factor I. This factor is responsible for the growth promoting effects of growth hormone. Further, this factor serves as an indicator of overall growth hormone secretion. Serum IGF-I concentration increases in response to endogenous and exogenous administered growth hormone. These concentrations are low in growth hormone deficiency. The injection of a MVS cont_-ining the sequence for IGF-I (for example SK 733 IGF-I Sk2) can be used to treat growth disorders. The injection of the MVS is a long-term inexpensive way to increase systemic blood concentration of IGF-I in patients with growth hormone deficiency.
Example 8 Treatment of Muscle Atrophy Due To Age Growth hormone levels decline with increasing age. The levels in healthy men and women above age of 55 are approximately one third lower than the levels in men and women 18 to 33. The decline in growth hormone and IGI-I production correlate with the decrease in muscle mass, termed senile muscle atrophy, and increase in adiposity that occur in healthy human subjects. Administering growth hormone three times a week to healthy 61 to 81 year old men who had serum levels below those of healthy younger men increased the Serum IGF-I levels to within the range found in young healthy adults. This increase level led to increased muscle mass and strength and reduced body fat.
The convenient cloning sites in the Myogenic Vector System are used to construct MVS vectors containing human growth hormone cDNA sequence and/or the human growth hormone releasing (secretory) hormone. The expression of an MVS driven growth hormone gene following intramuscular injections is another way to increase IGF-I serum levels. The MVS expression of the growth factor releasing hormone (GHRH) might be more advantageous than the expression of either IGF-I or the growth hormone vectors transcripts. Since GHRH is reduced in the elderly it appears to be responsible for the lack of GH secretion rather than the anterior pituitary capability of synthesizing growth hormone, thus the increased expression of GHRH from muscle would increase GHRH levels in systemic blood system and can allow for the natural diurnal secretion pattern of GH from the anterior pituitary. In this way, GHRH could act as the natural secretogogue allowing for elevated secretion or release of GH from the hypothalamus of the elderly.
Thus, the application of myogenic vector systems to express insulin-like growth factors through the injection of the SK 733 IGF-I Sk2 vector into adult muscle of the elderly is a long-term inexpensive way to increase systemic blood concentration of IGF-I in the elderly.
Example 9
Human Muscle Atrophies Induced by Spinal Cord Injuries. Denervation. and Neuromuscular Diseases
Insulin-like growth factors are one of the key factors that potentiate muscle development and muscle growth. Myoblasts naturally secrete IGF-
I/IGF-II as well as its cognate binding proteins during the onset of fusion.
This process coincides with the appearance of muscle specific gene products.
In terminally differentiated muscle, signals propagated from passive stretch induced hypertrophy induce the expression of IGF genes. Many of the actions of IGFs on muscle result from interactions with the IGF-I receptor. The receptor is a ligand activated tyrosine specific protein kinase. Insulin-like growth factors are also known neurotrophic agents which maintain neuronal muscular synapses, neuron integrity, and neuronal cell life under neurodegenerative conditions. It has been possible to increase muscle growth under otherwise stringent growth conditions in myogenic cultures and transgenic mice by injecting a MVS containing the IGF-I sequence. The direct injections into adult muscle allows direct transfer of growth promoting gene constructions into muscle and thus provides postnatal gene therapy. Since the MVS driven genes are relatively insensitive to the innerva tion state of the muscle, they provide a direct and rather broad application for remedying certain kinds of human muscle atrophies caused by spinal cord injuries and neuromuscular diseases. These diseases include the spinal cord muscular atrophies and aymtrophic lateral sclerosis (ALS). In this treatment, the product of the MVS acts as a neurotrophic agent secreted from injected muscle and as a hypertrophic agent to maintain muscle integrity.
Example 10 Atherosclerotic Cardiovascular Diseases Atherosclerotic cardiovascular disease is a major cause of mortality in the United States and the world. The atherosclerotic plaque, the basic underlying lesion in atherosclerosis, contains cholesterol esters that are derived from circulating lipids. these circulating lipids are essential to the development of atherosclerosis. The plasma concentration of high density Hpoprotein (HDL) is inversely related to the propensity for developing atherosclerosis. In the nascent state, HDL is secreted in the form of discoidal particles. These particles consist of a bilayer of phosphlipids onto which the apolipoproteins (apoA-I, ApoII and E) are embedded. HDL captures cholesterol esters by the action of an enzyme, lecithin-cholesterol acyltransferase. HDL is secreted from the liver, the small intestine and possibly other tissues.
The APO A-I cDNA is 878 bp and encodes 267 amino acids, including the 24 amino acid propropeptides. Increasing the circulating levels of HDL can influence or reverse cholesterol transport, and thus reduce the propensity for forming atherosclerotic plaques. The insertion of the human APO A-I coding sequences into the MVS serves as an expression vector for enhanced APO A-I expression following injection of plasmid DNA into skeletal muscle. The MVS APO A-I hybrid gene is effective for long term expression, biosynthesis and secretion of HDL in an ectopic site, and thus increases the content of total secretable HDL in blood plasma.
Example 11 Vitamin D Regulatable Myogenic Vector System The first tier of myogenic vectors designed with the skeletal actin promoters and 3' untranslated regions provide high levels of unregulated transcriptional activity. Under certain circumstances, it is desirable to control the vector's transcriptional activity and to switch gene transcription on and off by the systemic introduction of a simple inducer or ligand. It is also important that the regulation of the myogenic vector be controlled by natural inducer products that are neither considered toxic to humans nor are immunogenic. Two different Vitamin D regulatory systems are shown in Figures 8 and 9.
The cellular concentration of Vitamin D receptor (VDR) in muscle can be increased through the MVS system by injecting a hybrid skeletal actin VDR gene that would be under control of the actin promoter and the 3' UTR stabilizing sequences. The target, SEQ. ID. No. 2, is constructed to contain synthesized multimers of the Vitamin D regulatory element (VDRE). This target is linked to a minimal Herpes Simplex Virus (HSV) thymidine kinase promoter. Transcriptional activity emanating from the TK promoter is regulated by the presence of VDR and coactivated by the ligand, Vitamin D. Any polypeptide sequence cloned in tandem to the HSV promoter, as a cDNA, is driven from the target vector when Vitamin D is introduced into the muscle cells. The hybrid actin VDR gene and the target vector are linked on the same plasmid or coinjected on separate plasmids. Premeasured levels of
Vitamin D are administered by drinking a glass of milk or taking a Vitamin D pill. The levels are used to activate transcription of the target vector. Taking the ligand on every other day, will oscillate the promoter activity. Removal of the ligand, Vitamin D, from the diet down regulates or represses transcription from the target vector.
Example 12
Vaccine Production
The MVS system is well suited for directing the expression of an exogenous protein epitope in muscle, and thus, for generating vaccines in humans and animals. Targeted sequences are inserted into the cassette of a
MVS for expression of protein epitopes for mediating protective immunization. For example, the constant regions of the AIDS viral proteins
GP 120, GP 160 and GP 41 and for cell mediated immunity GP 24, the surface proteins of Hepatitis A, B, C, and C viruses, respiratory viruses including influenza, and the gastrointestinal rotoviruses are used. The MVS is then injected into the human or animal.
All patents and publications mentioned in the specification are indicative of the levels of those skilled in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
One skilled in the art will readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned as well as those inherent therein. The myogenic vector systems along with the methods, procedures treatments and vaccinations described herein are presently representative of preferred embodiments are exemplary and not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the invention or defined by this scope with the claims.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Schwartz, Robert J. DeMayo, Franco O'Malley, Bert W.
(ii) TITLE OF INVENTION: Myogenic Vector Systems
(iii) NUMBER OF SEQUENCES: 2
(iv) CORRESPONDENCE ADDRESS: (A) ADDRESSEE: Thomas D. Paul
(B) STREET: 1301 McRinney, Suite 5100
(C) CITY: Houston
(D) STATE: Texas
(E) COUNTRY: United States of America (F) ZIP: 77010-3095
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: Patentin Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US
(B) FILING DATE:
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Paul, Thomas D. (B) REGISTRATION NUMBER: 32,714
(C) REFERENCE/DOCKET NUMBER: D-5379
(ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 713/651-5325 ) (B) TELEFAX: 713/651-5246
(C) TELEX: 762829
(2) INFORMATION FOR SEQ ID N0:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 275 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
( i) SEQUENCE DESCRIPTION: SEQ ID NO:l:
TAAACATGTT TACATGATCA CTTTGCCAAC CACACTCAGG ATGACAATCT TGTAGGTTCC 60
AGGCTGCTGA GGACCTGCAC CAGCCATGCA ACTTTCTATT TTGTAACAAT TTCTGGTTAC 120
TGTTGCTGCA AAGCCCATGT GACACAGTGT ATGTAAAGTG TACATAAATT AATTTATTTT 180
ACCTCGTTTT GTTTGTTTTT AAAACCAATG CCCTGTGGAA GGAAACATAA AACTTCAAGA 240 AGCATTAAAT CATCAGTCAT TCTGTCACAC CCCTA 275
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2J
GGTGACTCAC CGGGTGAACG GGGCATT 27

Claims

CLAIMS What we claim is:
1. A myogenic vector system (MVS) for the expression of a nucleic acid sequence in myogenic tissue, comprising a promoter; a cassette whose 5' end is attached to the 3' end of the promoter, said cassette containing a nucleic acid sequence which is to be expressed; a myogenic specific 3' untranslated region (3'UTR) and a non-coding region (NCR) contiguous to the 3' end of the 3' UTR, said NCR containing a transcriptional termination signal, wherein the 5' end of the 3' UTR is attached to the 3' end of the cassette.
2. The MVS of claim 1, further comprising a 5' mRNA leader sequence inserted between the promoter and cassette, wherein the 5' end of said leader sequence is attached to the promoter and the 3' end is attached to the cassette.
3. A MVS for the expression of a nucleic acid sequence in myogenic tissues comprising a functional unit which expresses the nucleic acid sequence, wherein the functional unit is comprised of elements all sequentially linked together 5' to 3', said elements include the promoter; a 5' mRNA leader sequence; a first intron; an initiation ATG and NcoI cloning site; a cassette, wherein the cassette has an
EcoRl site at its 3' end and contains the nucleic acid sequence to be expressed; a myogenic specific 3' untranslated region (3' UTR); and a non-coding region (NCR) contiguous to the 3' end of the 3' UTR, said NCR containing a transcriptional termination signal.
4. The MVS of claim 1 or 3, wherein the promoter is myogenic specific.
5. The MVS of claim 2 or 3, wherein the leader sequence is myogenic specific.
6. The MVS of claim 1 or 3, wherein the promoter is selected from the group consisting of skeletal alpha actin gene promoter, fast myosin light chain 1 promoter, myosin heavy chain promoter, tropinin T promoter, muscle creatinine kinase promoter/enhancer, acetycholine receptor subunits, cytomegalovirus promoter, RSV promoter and Rous Sarcoma virus LTR.
7, The MVS of claim 1 or 3, wherein the promoter is a skeletal alpha actin gene promoter.
8. The MVS of claim 1, 3 or 59, wherein the 3' UTR and NCR are selected from the group consisting of skeletal alpha actin gene, fast myosin light chain 1 gene, myosin heavy chain gene, tropinin T gene and muscle creatinine kinase gene.
9. The MVS of claim 1, 3 or 59, wherein the 3' UTR and NCR are from the skeletal alpha actin gene.
10. The MVS of claim 1, 3 or 59 further comprising a regulator system for regulating the expression of the nucleic acid sequence.
11. The MVS of claim 10, wherein at least one of the serum response factors in the promoter region is a chimera, having its DNA binding domain replaced with a receptor DNA binding sequence.
12. The MVS of claim 11, wherein the receptor binding site is selected from the family of receptors consisting of vitamin, steroid, thyroid, orphan, hormone, retenoic acid, and thyroxine.
13. The MVS of claim 11, wherein the receptor binding site is a Vitamin D receptor.
14. The MVS of claim 1, 3 or 59, wherein the expressed nucleic acid sequence codes for a polypeptide.
15. The MVS of claim 1, 3 or 59, wherein the expressed nucleic acid sequence codes for the polypeptide selected from the group consisting of a hormone, growth factor, enzyme, apolipoprotein, clotting factor, tumor suppressor, tumor antigen, viral protein, bacterial surface protein, and parasitic cell surface protein.
16. The MVS of claim 1, 3 or 59, wherein the expressed nucleic acid sequence codes for insulin like growth factor I, insulin like growth factor II or insulin growth factor binding protein.
17. The MVS of claim 1, 3 or 59, wherein the expressed nucleic acid sequence codes for growth hormone releasing hormone.
18. The MVS of claim 1, 3 or 59, wherein the expressed nucleic acid sequence codes for apolipoprotein A-I.
19. The MVS of claim 10, wherein the expressed nucleic acid sequence codes for insulin like growth factor I, insulin like growth factor II or insulin growth factor binding protein.
20. The MVS of claim 10, wherein the expressed nucleic acid sequence codes for growth hormone releasing hormone.
21. The MVS of claim 10, wherein the expressed nucleic acid sequence codes for apolipoprotein A-I.
22. A method for introducing a continuous supply of polypeptide into a human, animal or tissue culture, comprising the step of injecting an effective amount of the MVS of claim 1 or 3 into skeletal muscle of said human or animal or into said tissue culture.
23. A method for introducing a regulated supply of polypeptide into a human or animal or tissue culture, comprising the step of injecting in an effective amount of the MVS of claim 10 into skeletal muscle of said human or animal or into said tissue culture.
24. A method for treating muscle atrophy in aging humans, comprising the step of injecting an effective amount of the MVS of claim 16 into skeletal muscle of said aging human.
25. A method for treating muscle atrophy in aging humans, comprising the step of injecting an effective amount of the MVS of claim 19 into skeletal muscle of said aging human.
26. A method for treating muscle atrophy in aging humans, comprising the step of injecting an effective amount of the MVS of claim 17 into skeletal muscle of said aging human.
27. A method for treating muscle atrophy in aging humans, comprising the step of injecting an effective amount of the MVS of claim 20 into skeletal muscle of said aging human.
28. A method of treating a human with muscle atrophy induced by spinal cord injuries or neuromuscular diseases, comprising the step of injecting an effective amount of the MVS of claim 16 into the skeletal muscle of the human.
29. The method of claim 28, wherein the human has amyotrophic lateral sclerosis.
30. A method of treating a human with muscle atrophy induced by spinal cord injuries or neuromuscular diseases, comprising the step of injecting an effective amount of the MVS of claim 19 into the skeletal muscle of the human.
31. The method of claim 30, wherein the human has amyotrophic lateral sclerosis.
32. A method of preventing or treating atherosclerotic cardiovascular disease in a human, comprising the step of injecting an effective amount of the MVS of claim 18 into skeletal muscle of the human.
33. A method of preventing or treating atherosclerotic cardiovascular disease in a human, comprising the step of injecting an effective amount of the MVS of claim 21 into skeletal muscle of the human.
34. A method of gene replacement, comprising the step of injecting an effective amount of the MVS of claim 1 or 3 into skeletal muscle, wherein the nucleic acid in the cassette contains the gene to correct the genetic defect.
35. The method of claim 34, wherein the cassette contains a sequence to code for the gene selected from the group consisting of glycogen phosphorylase, alpha-1-antitrypsin and dystrophin.
36. A method of gene replacement, comprising the step of injecting an effective amount of the MVS of claim 10 into skeletal muscle, wherein the nucleic acid in the cassette contains the gene to correct the genetic defect.
37. The method of claim 36, wherein the cassette contains a sequence to code for the gene selected from the group consisting of glycogen phosphorylase, alpha- 1-antitrypsin and dystrophin.
38. A method for vaccine production in a human or animal, comprising the step of injecting an effective amount of the MVS of claim 1 or 3 into skeletal muscle of the human or animal, wherein the nucleic acid sequence in the cassette codes for a polypeptide capable of eliciting an antibody response.
39. The method of claim 38, wherein the nucleic acid sequence is selected from the group which codes for a bacterial cell surface protein, a viral protein and a parasitic cell protein.
40. The method of claim 38, wherein the nucleic and sequence codes for an AIDS virus protein.
41. A method for vaccine production in a human or animal, comprising the step of injecting an effective amount of the MVS of claim 10 into skeletal muscle of the human or animal, wherein the nucleic acid sequence in the cassette codes for a polypeptide capable of eliciting an antibody response.
42. The method of claim 41, wherein the nucleic acid sequence is selected from the group which codes for a bacterial cell surface protein, a viral protein and a parasitic cell protein.
43. The method of claim 41, wherein the nucleic and sequence codes for an AIDS virus protein.
44. A method of treating growth disease, comprising the step of injecting an effective amount of the MVS of claim 1 or 3 into skeletal muscle of the diseased individual, wherein the nucleic acid sequence in the cassette contains the growth hormone sequence.
45. A method of treating growth disease, comprising the step of injecting an effective amount of the MVS of claim 10 into skeletal muscle of the diseased individual, wherein the nucleic acid sequence in the cassette contains the growth hormone sequence.
46. A regulatable myogenic vector system (MVS) for the expression of a specific nucleic acid sequence in myogenic tissue comprising: a first functional unit, elements of said first unit comprise a myogenic specific promoter; a nucleic acid sequence coding for a receptor; a myogenic specific 3' untranslated region (3' UTR) and a myogenic specific non-coding region (NCR); wherein the elements of the first unit are linked together sequentially starting from the
5' end with the promoter, followed by the receptor nucleic acid sequence, followed by the 3' UTR and contiguous NCR; a second functional unit, elements of said second unit comprise a response element corresponding to the receptor; a thymidine kinase promoter; the specific nucleic acid sequence of interest; a myogenic specific 3' UTR and a contiguous myogenic specific NCR; wherein the elements of the second unit are linked together sequentially starting from the 5' end with the response element followed by promoter followed by the specific sequence, followed by the 3' UTR and NCR; wherein the first unit expresses the receptor, said receptor forming an interaction between the response element, the receptor and an agent which binds to the receptor, said interaction up regulating the expression of the sequence of interest.
47. The MVS of claim 46 wherein the first and second functional units are on the same vector.
48. The MVS of claim 46, wherein the first and second functional units are on separate vectors.
49. The MVS of claim 46, wherein the receptor is selected from the family of receptors consisting of vitamin, steroid, thyroid, orphan, hormone, retinoic acid and thyroxine.
50. The method of claim 46 wherein the receptor is the Vitamin D receptor, the response element is the Vitamin D response element and agent is Vitamin D.
51. The MVS of claim 1, 3 or 46, further comprising a coating, said coating includes: a DNA initiation complex, said DNA initiation complex comprises a serum response factor (SRF), a transcription initiation factor (TIF) and a transregulatory factor (TRF), wherein the SRF is bound to the serum response element within the promoter, the TIF and TRF interact with the SRF and the TATA box within the promoter to form a stable complex; and histones, said histones bound nonspecifically to the remaining DNA in said MVS.
52. A method for introducing a polypeptide into a human, animal or tissue culture, comprising the step of injecting an effective amount of the coated MVS of claim 51 into skeletal muscle of said human or animal or tissue culture.
53. A method of treating human or animal disease, comprising the step of injecting an effective amount of the coated MVS of claim 51 into skeletal muscle of said human or animal.
54. A method of gene replacement, comprising the step of injecting an effective amount of the coated MVS of claim 51 into skeletal muscle, wherein the cassette contains the gene sequence to correct the genetic defect.
55. A method for vaccine production in a human or animal, comprising the step of injecting an effective amount of the coated MVS of claim 51 in skeletal muscle of the human or animal, wherein the cassette includes the sequence coding for a polypeptide capable of eliciting an antibody response.
56. A method for treating muscle atrophy, comprising the step of injecting an effective amount of the coated MVS of claim 51 into skeletal muscle.
57. The MVS of claim 10, further comprising a coating, said coating including: a DNA initiation complex, said DNA initiation complex comprising a serum response factor (SRF), transcription initiation factor (TIF) and transregulatory factor (TRF), wherein the SRF is attached to the serum response element within the promoter, the TIF and TRF interact with the SRF and the TATA box within the promoter to form a stable complex; and histones, said histones bound nonspecifically to the remaining DNA in said MVS.
58. The MVS of claim 11, further comprising a coating, said coating includes: a DNA initiation complex, said DNA initiation complex comprises a serum response factor (SRF), a transcription initiation factor (TIF) and a transregulatory factor (TRF), wherein the SRF is attached to the serum response element within the promoter, the
TIF and TRF interact with the SRF and the TATA box within the promoter to form a stable complex; and histones, said histones bound nonspecifically to the remaining DNA in said MVS.
59. A myogenic vector system (MVS) for the expression of a nucleic acid sequence in myogenic tissue, comprising: a 5' oligonucleotide, said oligonucleotide including a promoter; a 3' oligonucleotide containing a myogenic specific 3' untranslated region (3' UTR) and a non-coding region (NCR) contiguous to the 3' end of the 3' UTR, said NCR containing a transcriptional termination signal; and a linker, having a plurality of restriction endonuclease sites, said linker connecting the 5' oligonucleotide to the 3' oligonucleotide and said linker further providing a position for insertion of a cassette containing the nucleic acid to be expressed.
60. The MVS of claim 59, wherein the 5' oligonucleotide further includes a leader sequence.
61. The MVS of claim 1 or 3, wherein the nucleic acid sequence to be expressed is heterologous relative to the 3' UTR and NCR in the MVS.
62. A recombinant cell containing the vector of claims 1, 3, 10 or
59.
63. A vaccine comprising the vector of claim 1, 3, 10 or 59, wherein the nucleic acid in the cassette encoded an immunoglobin gene protein.
64. A pharmacological compound comprising the myogenic of claims 1, 3 10 or 59 and a pharmacological delivery vehicle.
65. A method for increasing milk production comprising the step of introducing a pharmacological dose of the myogenic vector of claim 1, 3, 10 or 59 into a milk producing animal, wherein the nucleic acid of said cassette encodes for growth hormone.
66. A method for increasing meat production comprising the step of introducing a pharmacological dose of the myogenic vector of clai 1, 3, 10 or 59 into a meat producing animal, wherein the nucleic acid of said cassette encodes for growth hormone.
PCT/US1992/009353 1991-11-06 1992-11-03 Myogenic vector systems WO1993009236A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU31246/93A AU660751B2 (en) 1991-11-06 1992-11-03 Myogenic vector systems
JP5508631A JPH07500967A (en) 1991-11-06 1992-11-03 Myogenic vector system
EP92925044A EP0635060A4 (en) 1991-11-06 1992-11-03 Myogenic vector systems.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US789,919 1991-11-06
US07/789,919 US5298422A (en) 1991-11-06 1991-11-06 Myogenic vector systems

Publications (1)

Publication Number Publication Date
WO1993009236A1 true WO1993009236A1 (en) 1993-05-13

Family

ID=25149108

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1992/009353 WO1993009236A1 (en) 1991-11-06 1992-11-03 Myogenic vector systems

Country Status (7)

Country Link
US (2) US5298422A (en)
EP (1) EP0635060A4 (en)
JP (1) JPH07500967A (en)
AU (1) AU660751B2 (en)
CA (1) CA2122617A1 (en)
PT (1) PT101042B (en)
WO (1) WO1993009236A1 (en)

Cited By (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993024640A2 (en) * 1992-06-04 1993-12-09 The Regents Of The University Of California Methods and compositions for in vivo gene therapy
WO1994028151A1 (en) * 1993-05-20 1994-12-08 Royal Free Hospital School Of Medicine Gene therapy for haemophilia
WO1995011307A1 (en) * 1993-10-22 1995-04-27 Institut Pasteur Nucleotide vector, composition containing such vector and vaccine for immunization against hepatitis
GB2293604A (en) * 1994-09-20 1996-04-03 British Tech Group Dermal fibroblast cells, convertable to myoblast cells, may comprise muscle-speicific DNA
WO1996010401A1 (en) * 1994-10-04 1996-04-11 Emory University Methods for regulation of insulin-like growth factor i receptor
WO1997011190A2 (en) * 1995-09-19 1997-03-27 Pharmadigm, Inc. Dna construct comprising a muscle specific regulatory element for immunization or gene therapy
WO1997049817A1 (en) * 1996-06-25 1997-12-31 The Ludwig Institute For Cancer Research Brain glycogen phosphorylase cancer antigen
WO1998020041A1 (en) * 1996-11-08 1998-05-14 Oklahoma Medical Research Foundation Endothelium specific expression regulated by epcr control elements
WO1998024922A1 (en) * 1996-12-02 1998-06-11 Valentis, Inc. Insulin-like growth factor i (igf-i) expression system and methods of use
WO1998033529A1 (en) * 1997-02-04 1998-08-06 Valentis, Inc. Treatment for urinary incontinence using gene therapy techniques
WO1999003981A1 (en) * 1997-07-17 1999-01-28 Ppl Therapeutics (Scotland) Limited Nucleic acid expression constructs incorporating a heterologous intron whose natural position is within the 5'untranslated region of its gene
WO1999005300A2 (en) * 1997-07-24 1999-02-04 Valentis, Inc. Ghrh expression system and methods of use
US5869292A (en) * 1995-11-13 1999-02-09 Immuno Ag Hybrid proteins with modified activity
WO1999014346A2 (en) * 1997-09-19 1999-03-25 Sequitur, Inc. SENSE mRNA THERAPY
US6013780A (en) * 1996-09-06 2000-01-11 Technion Research & Development Co. Ltd. VEGF145 expression vectors
FR2783839A1 (en) * 1998-09-25 2000-03-31 Aventis Pharma Sa Hybrid promoter useful for gene expression in smooth-muscle cells includes the enhancer region of a ubiquitous strong promoter/enhancer
WO2000018908A1 (en) * 1998-09-25 2000-04-06 Aventis Pharma S.A. Use of specific hybrid promoters for controlling tissue expression
US6136581A (en) * 1996-11-22 2000-10-24 Sugen, Inc. Kinase genes and uses
US6207401B1 (en) 1995-12-18 2001-03-27 Sugen, Inc. Diagnosis and treatment of AUR-1 and/or AUR-2 related disorders
US6228641B1 (en) 1997-05-20 2001-05-08 Sugen, Inc. Diagnosis and treatment of PTP04 related disorders
US6335010B1 (en) 1996-11-08 2002-01-01 University Of California At San Diego Gene therapy in coronary angioplasty and bypass
US6342593B1 (en) 1997-06-11 2002-01-29 Sugen, Inc. Diagnosis and treatment of ALP related disorders
US6380373B1 (en) 1995-09-15 2002-04-30 Baylor College Of Medicine Steroid receptor coactivator compositions and methods of use
US6388063B1 (en) 1997-06-18 2002-05-14 Sugen, Inc. Diagnosis and treatment of SAD related disorders
US6482605B1 (en) 1996-11-13 2002-11-19 Sugen, Inc. Protein tyrosine phosphatase PTP20 and related products and methods
US6489451B1 (en) 1997-04-10 2002-12-03 Hefei-Siu-Fung Ustc Pharmaceutical Co., Ltd. Antithrombosis enzyme from the snake venom of agkistrodon acutus
WO2002097099A1 (en) * 2001-05-29 2002-12-05 Valentis, Inc. Regulated expression of ghrh
US6495353B1 (en) 1998-01-21 2002-12-17 Sugen, Inc. Human orthologues of wart
US6506550B1 (en) 1997-07-11 2003-01-14 Brandeis University Method of including apoptosis by reducing the level of thiamin
US6534483B1 (en) 1998-08-14 2003-03-18 Valentis, Inc. Protected one-vial formulation for nucleic acid molecules, methods of making the same by in-line mixing, and related products and methods
US6541615B1 (en) 1996-11-15 2003-04-01 Max-Planck-Gellschaft Zur Foderung Der Wissenschaften E.V. SIRP proteins and uses thereof
US6596526B1 (en) 2000-06-09 2003-07-22 Baxter Aktiengesellschaft Furin polypeptides with improved characteristics
US6656716B1 (en) 1998-04-14 2003-12-02 Sugen, Inc. Polypeptide fragments of human PAK5 protein kinase
US6797513B2 (en) 1996-12-19 2004-09-28 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acid encoding CLK2 protein kinases
US6818440B2 (en) 1997-04-28 2004-11-16 Sugen, Inc. Diagnosis and treatment of alk-7 related disorders
US6821946B2 (en) 2000-05-10 2004-11-23 University College London Repair of nerve damage
US6861442B1 (en) 1998-12-30 2005-03-01 Sugen, Inc. PYK2 and inflammation
US7074589B1 (en) 1996-06-17 2006-07-11 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acids encoding BDP-1
US7098025B1 (en) 1997-07-25 2006-08-29 Ligand Pharmaceuticals Incorporated Human peroxisome proliferator activated receptor gamma (pparγ) gene regulatory sequences and uses therefor
US7115710B2 (en) 1997-06-11 2006-10-03 Sugen, Inc. Diagnosis and treatment of PTP related disorders
US7115728B1 (en) 1995-01-30 2006-10-03 Ligand Pharmaceutical Incorporated Human peroxisome proliferator activated receptor γ
US7119174B2 (en) 1995-12-18 2006-10-10 Sugen, Inc. Diagnosis and treatment of AUR1 and/or AUR2 related disorders
EP1726651A1 (en) 1996-03-11 2006-11-29 University College London Method of treating muscular disorders
EP1892299A2 (en) * 1997-09-19 2008-02-27 Sequitur, Inc. Sense mRNA therapy
US7544855B2 (en) 2004-04-23 2009-06-09 Buck Institute Transgenic mouse whose genome comprises an APP having a mutation at amino acid 664
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2015066377A1 (en) * 2013-10-30 2015-05-07 The Curators Of The University Of Missouri Method for scalable skeletal muscle lineage specification and cultivation
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9492386B2 (en) 2002-06-28 2016-11-15 Protiva Biotherapeutics, Inc. Liposomal apparatus and manufacturing methods
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9919031B2 (en) 2002-02-07 2018-03-20 The Board Of Trustees Of University Of Illinois Use of the insulin-like-growth factor 1 splice variant MGF for the prevention of myocardial damage
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US11479792B2 (en) 2017-07-13 2022-10-25 Upside Foods, Inc. Compositions and methods for increasing the efficiency of cell cultures used for food production
US11976302B2 (en) 2017-05-06 2024-05-07 Upside Foods, Inc. Compositions and methods for increasing the culture density of a cellular biomass within a cultivation infrastructure

Families Citing this family (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5925564A (en) * 1991-11-06 1999-07-20 Baylor College Of Medicine Expression vector systems and method of use
US6348327B1 (en) * 1991-12-06 2002-02-19 Genentech, Inc. Non-endocrine animal host cells capable of expressing variant proinsulin and processing the same to form active, mature insulin and methods of culturing such cells
JPH07509694A (en) * 1992-05-14 1995-10-26 ベイラー・カレッジ・オブ・メディシン Mutated steroid hormone receptors, their uses and molecular switches for gene therapy
US6416998B1 (en) 1992-09-02 2002-07-09 Baylor College Of Medicine Plasmid encoding a modified steroid hormone
US6264915B1 (en) 1992-09-13 2001-07-24 The President And Fellows Of Harvard College Process for detecting potential carcinogens
US5962427A (en) 1994-02-18 1999-10-05 The Regent Of The University Of Michigan In vivo gene transfer methods for wound healing
US20020193338A1 (en) * 1994-02-18 2002-12-19 Goldstein Steven A. In vivo gene transfer methods for wound healing
US6025183A (en) * 1994-02-28 2000-02-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Transgenic animal assay system for anti-cholinesterase substances
IL112820A0 (en) * 1994-03-07 1995-05-26 Merck & Co Inc Coordinate in vivo gene expression
US6995008B1 (en) 1994-03-07 2006-02-07 Merck & Co., Inc. Coordinate in vivo gene expression
US6551618B2 (en) * 1994-03-15 2003-04-22 University Of Birmingham Compositions and methods for delivery of agents for neuronal regeneration and survival
US5670347A (en) 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
WO1995034640A1 (en) * 1994-06-13 1995-12-21 Henry Ford Health System A novel neuronal-neonatal gene: neuronatin
US5444047A (en) * 1994-06-16 1995-08-22 Dipasquale; Gene Treatment of arthritic and post-surgical orthopedic conditions with Insulin-like Growth Factor-I
US20030152909A1 (en) * 1994-11-16 2003-08-14 Mitrani Eduardo N. In vitro micro-organs, and uses related thereto
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5728807A (en) * 1995-05-16 1998-03-17 Ramot-University Authority For Applied Research And Industrial Development, Ltd. Mutated proteins associated with ataxia-telangiectasia
US5858661A (en) 1995-05-16 1999-01-12 Ramot-University Authority For Applied Research And Industrial Development Ataxia-telangiectasia gene and its genomic organization
US5777093A (en) * 1995-05-16 1998-07-07 Ramot-University Authority For Applied Research & Industrial Development Ltd. cDNAs associated with ataxia-telangiectasia
FR2736642B1 (en) * 1995-07-10 1997-09-12 Pasteur Institut IMMUNOVECTORS, IN PARTICULAR ANTIBODIES AND FRAGMENTS OF ANTIBODIES FOR USE IN THE INTRACELLULAR AND INTRANUCLEAR TRANSPORT OF BIOLOGICALLY ACTIVE PRINCIPLES IN PARTICULAR OF HAPTENES, PROTEINS AND NUCLEIC ACIDS
KR19990044317A (en) * 1995-09-01 1999-06-25 라미 핀클러 ; 하나넬 크바탄스키 Manipulation and detection of amylophosphatase 2C-P2Cα-expression
US6140309A (en) * 1996-03-12 2000-10-31 University Of South Florida Vasoactive effects and free radical generation by β-amyloid peptides
US6143037A (en) * 1996-06-12 2000-11-07 The Regents Of The University Of Michigan Compositions and methods for coating medical devices
CA2259123C (en) 1996-07-01 2003-10-21 Jim A. Wright Oligonucleotides from the untranslated regions of ribonucleotide reductase and their use to modulate cell growth
EP0960204A4 (en) * 1996-07-26 2002-01-23 Sloan Kettering Inst Cancer Method and reagents for genetic immunization
US5898094A (en) * 1996-10-21 1999-04-27 University Of South Florida Transgenic mice expressing APPK670N,M671L and a mutant presenilin transgenes
AU6908998A (en) * 1996-10-29 1998-05-22 Baylor College Of Medicine Modified glucocorticoid receptors, glucocorticoid receptors/progesterone receptors hybrids
US6387700B1 (en) * 1996-11-04 2002-05-14 The Reagents Of The University Of Michigan Cationic peptides, Cys-Trp-(LYS)n, for gene delivery
US6635623B1 (en) 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
US20050096288A1 (en) * 1997-06-13 2005-05-05 Aragene, Inc. Lipoproteins as nucleic acid vectors
US7923250B2 (en) 1997-07-30 2011-04-12 Warsaw Orthopedic, Inc. Methods of expressing LIM mineralization protein in non-osseous cells
US6300127B1 (en) 1997-07-30 2001-10-09 Emory University Bone mineralization proteins, DNA, vectors, expression systems
EP1015471A1 (en) 1997-08-21 2000-07-05 Quark Biotech, Inc. Hypoxia-regulated genes
US7973156B2 (en) * 1997-08-21 2011-07-05 Quark Pharmaceuticals Inc. Hypoxia-regulated genes
US20030104973A1 (en) * 1997-08-21 2003-06-05 Quark Biotech, Inc. Hypoxia-regulated genes
WO1999018116A1 (en) * 1997-10-07 1999-04-15 Smithkline Beecham Corporation Method for modulating gene expression
US7687057B2 (en) 1998-01-09 2010-03-30 Yissum Research Development Company Of The Hebrew University Of Jerusalem In vitro micro-organs, and uses related thereto
DK1045918T3 (en) 1998-01-12 2008-12-08 Pedro Jose G protein-related kinase mutants in essential hypertension
US6395253B2 (en) 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
AU1128400A (en) 1998-10-22 2000-05-08 Medical College Of Georgia Institute, Inc. Long terminal repeat, enhancer, and insulator sequences for use in recombinant vectors
DE19907099A1 (en) * 1999-02-19 2000-09-07 Theragene Biomedical Lab Gmbh Novel nucleic acid construct useful in gene therapy comprising an hormone responsive element and transgene in which the hormone responsive element is not functionally linked to the transgene
US6759393B1 (en) * 1999-04-12 2004-07-06 Pfizer Inc. Growth hormone and growth hormone releasing hormone compositions
US20030131364A1 (en) * 1999-04-27 2003-07-10 Karen Duff Method for producing transgenic animal models with modulated phenotype and animals produced therefrom
US6787318B1 (en) * 1999-06-01 2004-09-07 Roskamp Research Institute, Llc Assay for evaluating the therapeutic effectiveness of agents in reducing Alzheimer's disease pathology
US20070160615A1 (en) * 1999-06-01 2007-07-12 Jun Tan Methods and compounds for disruption of CD40R/CD40L signaling in the treatment of Alzheimer's disease
WO2001000859A1 (en) * 1999-06-25 2001-01-04 Yissum Research Development Company Of The Hebrew University Of Jerusalem Method of inducing angiogenesis by micro-organs
US6770740B1 (en) 1999-07-13 2004-08-03 The Regents Of The University Of Michigan Crosslinked DNA condensate compositions and gene delivery methods
US6551996B1 (en) * 1999-07-26 2003-04-22 Baylor College Of Medicine Super-active porcine growth hormone releasing hormone analog
US20040192593A1 (en) 1999-07-26 2004-09-30 Baylor College Of Medicine Protease resistant ti-growth hormone releasing hormone
US6875582B1 (en) 1999-08-19 2005-04-05 Omniscience Pharmaceuticals, Inc. Methods and targets of antibiotic resistance
US6486134B2 (en) * 1999-09-17 2002-11-26 Leadergene Limited Gene treatment to enhance feed efficiency and growth rate of livestock
US6998232B1 (en) 1999-09-27 2006-02-14 Quark Biotech, Inc. Methods of diagnosing bladder cancer
AU778826B2 (en) * 1999-10-29 2004-12-23 Anges Mg, Inc. Gene therapy for diabetic ischemic disease
WO2001053323A2 (en) * 2000-01-24 2001-07-26 Ramot University Authority For Applied Research & Industrial Development Ltd. Plants tolerant of environmental stress conditions, methods of generating same and novel polynucleotide sequence utilized thereby
HUP0300810A2 (en) 2000-07-20 2003-08-28 M.G.V.S. Ltd. Artifical vascular grafts, and methods of producing and using same
US20040037828A1 (en) 2002-07-09 2004-02-26 Bar-Ilan University Methods and pharmaceutical compositions for healing wounds
AU2001288682A1 (en) 2000-08-30 2002-03-13 Chemocentryx, Inc. Inhibition of cmv infection and dissemination
WO2002024899A2 (en) 2000-09-25 2002-03-28 Valentis, Inc. Improved system for regulation of transgene expression
WO2002062296A2 (en) 2001-02-02 2002-08-15 Chemocentryx, Inc. Methods and compositions useful for stimulating an immune response
CA2441594A1 (en) * 2001-03-22 2002-10-03 Scentgene Pollination Ltd. Method of enhancing entomophilous
EP1578901A4 (en) * 2001-09-07 2006-03-29 Baylor College Medicine Linear dna fragments for gene expression
AR037038A1 (en) 2001-10-26 2004-10-20 Baylor College Medicine A COMPOSITION AND METHOD TO ALTER THE MAGRA BODY MASS AND THE OSEAS PROPERTIES IN A SUBJECT
WO2003048298A2 (en) * 2001-12-05 2003-06-12 Yissum Research Development Company Of The Hebrew University Of Jerusalem Nanoparticles containing polymeric nucleic acid homologs, pharmaceutical compositions and articles of manufacture containing same and methods of use thereof
CA2469310C (en) 2001-12-11 2013-01-29 Advisys, Inc. Plasmid mediated supplementation for treating chronically ill subjects
WO2003066825A2 (en) * 2002-02-07 2003-08-14 Baylor College Of Medicine Modified pituitary gland development in offspring from expectant mother animals treated with growth hormone releasing hormone therapy
US6992176B2 (en) 2002-02-13 2006-01-31 Technion Research & Development Foundation Ltd. Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
EP1359160A1 (en) * 2002-02-28 2003-11-05 Pfizer Products Inc. Androgen receptor overexpressing skeletal myoblasts
JP2005533102A (en) * 2002-07-12 2005-11-04 イッサム リサーチ ディベロップメント カンパニー オブ ザ ヘブリュー ユニバーシティー オブ エルサレム Method and apparatus for inducing biological processes by micro-organs
AU2003249208B2 (en) * 2002-07-16 2010-03-04 Vgx Pharmaceuticals, Llc Codon optimized synthetic plasmids
US20040146949A1 (en) * 2002-10-25 2004-07-29 Jun Tan Methods and compounds for disruption of CD40R/CD40L signaling in the treatment of alzheimer's disease
AU2003286911A1 (en) * 2002-11-04 2004-06-07 Dana-Farber Cancer Institute, Inc. In vivo imaging of e2f-regulated bioluminescent proteins
AU2003295366B2 (en) * 2002-11-04 2011-11-24 Advisys, Inc. Synthetic muscle promoters with activities exceeding naturally occurring regulatory sequences in cardiac cells
AU2003298675A1 (en) * 2002-11-22 2004-06-18 You-Jun Fei NaCT AS A TARGET FOR LIFESPAN EXPANSION AND WEIGHT REDUCTION
CA2513743C (en) * 2003-01-28 2013-06-25 Advisys, Inc. Reducing culling in herd animals growth hormone releasing hormone (ghrh)
WO2004081040A2 (en) 2003-03-12 2004-09-23 Advisys, Inc. Insulin-like growth factor (igf-i) plasmid-mediated supplementation for therapeutic applications
TW200424214A (en) * 2003-04-21 2004-11-16 Advisys Inc Plasmid mediated GHRH supplementation for renal failures
CA2575926C (en) * 2003-08-04 2014-02-25 Advisys, Inc. Canine specific growth hormone releasing hormone
WO2005059100A2 (en) * 2003-12-12 2005-06-30 New York University Methods and compositions relating to cystatin c
CA2592891C (en) * 2003-12-31 2013-05-28 Advisys, Inc. Reducing arthritis and lameness in subjects by growth hormone releasing hormone (ghrh) supplementation
WO2005073371A2 (en) * 2004-01-20 2005-08-11 Advisys, Inc. Enhanced secretion/retention of growth hormone releasing hormone (ghrh) from muscle cells by species-specific signal peptide
US20090005333A1 (en) * 2004-01-26 2009-01-01 Vgx Pharmaceuticlas, Inc. Reducing culling in herd animals growth hormone releasing hormone (ghrh)
US20060019241A1 (en) * 2004-04-20 2006-01-26 Vadivel Ganapathy Na+ and CI-coupled transport system for endogenous opioid peptides
WO2006014737A2 (en) * 2004-07-23 2006-02-09 Advisys, Inc. Growth hormone releasing hormone enhances the immune response induced by vaccination
US7846720B2 (en) * 2005-01-26 2010-12-07 Vgx Pharmaceuticals, Inc. Optimized high yield synthetic plasmids
NZ538097A (en) * 2005-02-07 2006-07-28 Ovita Ltd Method and compositions for improving wound healing
JP2009525110A (en) 2006-01-31 2009-07-09 マルチ ジーン バスキュラー システムズ, インコーポレイテッド Drug-eluting endovascular prostheses and methods of use
WO2007103683A2 (en) * 2006-03-01 2007-09-13 Roskamp Research Llc Compounds for inhibiting beta-amyloid production
WO2008006019A2 (en) * 2006-07-06 2008-01-10 Advisys Inc. Growth hormone releasing hormone treatment to decrease cholesterol levels
WO2008013918A2 (en) * 2006-07-26 2008-01-31 Myelin Repair Foundation, Inc. Cell cycle regulation and differentiation
US8124598B2 (en) * 2006-09-14 2012-02-28 Sharon Sageman 7-keto DHEA for psychiatric use
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
WO2008033375A2 (en) 2006-09-14 2008-03-20 Medgenics Ltd. Long lasting drug formulations
WO2008070875A2 (en) * 2006-12-08 2008-06-12 Roskamp Research Llc Polyhydroquinoline compounds and dihydropyridine compounds for inhibiting beta-amyloid production
WO2008152507A2 (en) 2007-03-16 2008-12-18 Multigene Vascular Systems, Inc. Compositions and methods for treating ophthalmic disorders
EP2514766A3 (en) 2007-03-29 2013-06-05 Technion Research & Development Foundation Ltd. Antibodies, methods and kits for diagnosing and treating melanoma
US20100160274A1 (en) * 2007-09-07 2010-06-24 Sharon Sageman 7-KETO DHEA for Psychiatric Use
US8811535B2 (en) * 2009-07-17 2014-08-19 Mitre Corporation Time-frequency space constructions of families of signals
US20110070199A1 (en) * 2009-09-21 2011-03-24 Scheiber Ii Lane Bernard Adaptable modified virus vector to deliver ribosomal ribonucleic acid combined with messenger ribonucleic acid as a medical treatment device to manage diabetes mellitus and other protein deficient dieases
CN103260649A (en) 2010-06-15 2013-08-21 迈德詹尼克斯医疗以色列有限公司 Long lasting drug formulations
US8993316B2 (en) 2011-11-16 2015-03-31 Brian P. Hanley Methods and compositions for gene therapy and GHRH therapy
WO2015059701A1 (en) 2013-10-24 2015-04-30 Medgenics Medical Israel Ltd. Micro-organs providing sustained delivery of a therapeutic polypeptide and methods of use thereof
MX2019002904A (en) 2016-09-14 2019-09-26 Modernatx Inc High purity rna compositions and methods for preparation thereof.
DE102021122096A1 (en) * 2021-08-26 2023-03-02 Forschungsinstitut für Nutztierbiologie Compositions comprising IGF-1, IGFBP-2 and insulin and their use

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4783405A (en) * 1983-01-18 1988-11-08 Eli Lilly And Company Recombinant DNA expression vectors useful in bacillus and other host cells

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1353839A (en) * 1963-04-10 1964-02-28 Resin tube with incorporated frame
EP0174608A1 (en) * 1984-09-13 1986-03-19 The Board Of Trustees Of The Leland Stanford Junior University Beta-actin gene and regulatory elements, preparation and use
GB8529014D0 (en) * 1985-11-25 1986-01-02 Biogen Nv Enhanced secretion of heterologous proteins
CA1294896C (en) * 1986-07-14 1992-01-28 Philip Salinas Intersticed nipple for elastomeric hoses and method
EP0832981A1 (en) * 1987-02-17 1998-04-01 Pharming B.V. DNA sequences to target proteins to the mammary gland for efficient secretion
US4873316A (en) * 1987-06-23 1989-10-10 Biogen, Inc. Isolation of exogenous recombinant proteins from the milk of transgenic mammals
JPH0791320B2 (en) * 1988-03-17 1995-10-04 ヤマサ醤油株式会社 Recombinant human cardiac myosin
US5093317A (en) * 1989-06-05 1992-03-03 Cephalon, Inc. Treating disorders by application of insulin-like growth factor
US5538722A (en) * 1989-06-13 1996-07-23 Stanford University Isolation, growth, differentiation and genetic engineering of human muscle cells
WO1991011522A1 (en) * 1990-01-26 1991-08-08 Baylor College Of Medicine MUTATED PROMOTER REGION FROM CHICKEN SKELETAL α-ACTIN GENE
AU7312891A (en) * 1990-02-12 1991-09-03 Board Of Regents, The University Of Texas System Satellite cell proliferation in adult skeletal muscle
HUT60523A (en) * 1990-02-22 1992-09-28 Biogen Inc Improved process for expressing polypeptides
GB9305091D0 (en) * 1993-03-12 1993-04-28 Thrombosis Res Inst Eukaryotic expression vectors driven by a novel strong mammalian cell promoter

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4783405A (en) * 1983-01-18 1988-11-08 Eli Lilly And Company Recombinant DNA expression vectors useful in bacillus and other host cells

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Methods in Enzymology, Vol. 185, issued 1990, R.J. KAUFMAN, "Vectors Used for Expression in Mammalian Cells", pages 487 to 511, see entire document. *
Nucleic Acids Research, Vol. 14, issued 04 November 1986, J.M. GRICHNIK et al., "Tissue Restricted and Stage Specific Transcription is Maintained within 411 Nucleotides Flanking the 5' End of the Chicken alpha-Skeletal Actin Gene", pages 1683 to 1701, see entire document. *
See also references of EP0635060A4 *

Cited By (137)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993025673A1 (en) * 1992-06-04 1993-12-23 The Regents Of The University Of California In vivo gene therapy with intron-free sequence of interest
WO1993024640A3 (en) * 1992-06-04 1994-04-28 Univ California Methods and compositions for in vivo gene therapy
WO1993024640A2 (en) * 1992-06-04 1993-12-09 The Regents Of The University Of California Methods and compositions for in vivo gene therapy
WO1994028151A1 (en) * 1993-05-20 1994-12-08 Royal Free Hospital School Of Medicine Gene therapy for haemophilia
US7825097B2 (en) 1993-10-22 2010-11-02 Institut Pasteur Nucleotide vector vaccine for immunization against hepatitis
WO1995011307A1 (en) * 1993-10-22 1995-04-27 Institut Pasteur Nucleotide vector, composition containing such vector and vaccine for immunization against hepatitis
FR2711670A1 (en) * 1993-10-22 1995-05-05 Pasteur Institut Nucleotide vector, composition containing it and vaccine for immunization against hepatitis.
GB2293604A (en) * 1994-09-20 1996-04-03 British Tech Group Dermal fibroblast cells, convertable to myoblast cells, may comprise muscle-speicific DNA
GB2293604B (en) * 1994-09-20 1996-09-11 British Tech Group Dermal fibroblast cells containing muscle-specific DNA and the use of dermal fibroblast cells in the treatment of muscular disorders
WO1996010401A1 (en) * 1994-10-04 1996-04-11 Emory University Methods for regulation of insulin-like growth factor i receptor
US7115728B1 (en) 1995-01-30 2006-10-03 Ligand Pharmaceutical Incorporated Human peroxisome proliferator activated receptor γ
US6380373B1 (en) 1995-09-15 2002-04-30 Baylor College Of Medicine Steroid receptor coactivator compositions and methods of use
WO1997011190A3 (en) * 1995-09-19 1997-06-26 Paradigm Biosciences Inc Dna construct comprising a muscle specific regulatory element for immunization or gene therapy
WO1997011190A2 (en) * 1995-09-19 1997-03-27 Pharmadigm, Inc. Dna construct comprising a muscle specific regulatory element for immunization or gene therapy
AU710756B2 (en) * 1995-09-19 1999-09-30 Inflabloc Pharmaceuticals, Inc. DNA construct comprising a muscle specific regulatory element for immunization or gene therapy
US5795872A (en) * 1995-09-19 1998-08-18 Pharmadigm, Inc. DNA construct for immunization
US6310196B1 (en) * 1995-09-19 2001-10-30 Joseph W. Ricigliano DNA construct for immunization or gene therapy
US5869292A (en) * 1995-11-13 1999-02-09 Immuno Ag Hybrid proteins with modified activity
US6051418A (en) * 1995-11-13 2000-04-18 Immuno Ag Hybrid proteins with modified activity
US5910481A (en) * 1995-11-13 1999-06-08 Immuno Ag Hybrid proteins with modified activity
US6156888A (en) * 1995-11-13 2000-12-05 Baxter Aktiengesellschaft Hybrid proteins with modified activity
US6130203A (en) * 1995-11-13 2000-10-10 Baxter Aktiengesellschaft Hybrid proteins with modified activity
US6716575B2 (en) 1995-12-18 2004-04-06 Sugen, Inc. Diagnosis and treatment of AUR1 and/or AUR2 related disorders
US7119174B2 (en) 1995-12-18 2006-10-10 Sugen, Inc. Diagnosis and treatment of AUR1 and/or AUR2 related disorders
US6207401B1 (en) 1995-12-18 2001-03-27 Sugen, Inc. Diagnosis and treatment of AUR-1 and/or AUR-2 related disorders
EP1726651A1 (en) 1996-03-11 2006-11-29 University College London Method of treating muscular disorders
US7074589B1 (en) 1996-06-17 2006-07-11 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acids encoding BDP-1
WO1997049817A1 (en) * 1996-06-25 1997-12-31 The Ludwig Institute For Cancer Research Brain glycogen phosphorylase cancer antigen
US6013780A (en) * 1996-09-06 2000-01-11 Technion Research & Development Co. Ltd. VEGF145 expression vectors
WO1998020041A1 (en) * 1996-11-08 1998-05-14 Oklahoma Medical Research Foundation Endothelium specific expression regulated by epcr control elements
US6200751B1 (en) 1996-11-08 2001-03-13 Oklahoma Medical Research Foundation Endothelial specific expression regulated by EPCR control elements
US6335010B1 (en) 1996-11-08 2002-01-01 University Of California At San Diego Gene therapy in coronary angioplasty and bypass
US6797501B2 (en) 1996-11-13 2004-09-28 Max-Planck-Gessellschaft Zur Fonderung De Wissenschaften E.V. Protein tyrosine phosphatase PTP20 and related products and methods
US6482605B1 (en) 1996-11-13 2002-11-19 Sugen, Inc. Protein tyrosine phosphatase PTP20 and related products and methods
US6541615B1 (en) 1996-11-15 2003-04-01 Max-Planck-Gellschaft Zur Foderung Der Wissenschaften E.V. SIRP proteins and uses thereof
US6136581A (en) * 1996-11-22 2000-10-24 Sugen, Inc. Kinase genes and uses
WO1998024922A1 (en) * 1996-12-02 1998-06-11 Valentis, Inc. Insulin-like growth factor i (igf-i) expression system and methods of use
US6797513B2 (en) 1996-12-19 2004-09-28 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acid encoding CLK2 protein kinases
WO1998033529A1 (en) * 1997-02-04 1998-08-06 Valentis, Inc. Treatment for urinary incontinence using gene therapy techniques
AU739224B2 (en) * 1997-02-04 2001-10-04 Valentis, Inc. Treatment for urinary incontinence using gene therapy techniques
US7223730B2 (en) 1997-04-10 2007-05-29 Hefei-Siu-Fung Ustc Pharmaceutical Co., Ltd. Antithrombosis enzyme from the snake venom of agkistrodon acutus
US6489451B1 (en) 1997-04-10 2002-12-03 Hefei-Siu-Fung Ustc Pharmaceutical Co., Ltd. Antithrombosis enzyme from the snake venom of agkistrodon acutus
US6818440B2 (en) 1997-04-28 2004-11-16 Sugen, Inc. Diagnosis and treatment of alk-7 related disorders
US6844177B2 (en) 1997-05-20 2005-01-18 Sugen, Inc. Diagnosis and treatment of PTP04 related disorders
US6228641B1 (en) 1997-05-20 2001-05-08 Sugen, Inc. Diagnosis and treatment of PTP04 related disorders
US6342593B1 (en) 1997-06-11 2002-01-29 Sugen, Inc. Diagnosis and treatment of ALP related disorders
US7115710B2 (en) 1997-06-11 2006-10-03 Sugen, Inc. Diagnosis and treatment of PTP related disorders
US6388063B1 (en) 1997-06-18 2002-05-14 Sugen, Inc. Diagnosis and treatment of SAD related disorders
US7803531B2 (en) 1997-07-11 2010-09-28 Brandeis University Method of inducing apoptosis by reducing the level of thiamin
US6506550B1 (en) 1997-07-11 2003-01-14 Brandeis University Method of including apoptosis by reducing the level of thiamin
WO1999003981A1 (en) * 1997-07-17 1999-01-28 Ppl Therapeutics (Scotland) Limited Nucleic acid expression constructs incorporating a heterologous intron whose natural position is within the 5'untranslated region of its gene
US6423693B1 (en) 1997-07-24 2002-07-23 Baylor College Of Medicine Growth hormone releasing hormone expression system and methods of use, including use in animals
WO1999005300A2 (en) * 1997-07-24 1999-02-04 Valentis, Inc. Ghrh expression system and methods of use
WO1999005300A3 (en) * 1997-07-24 1999-04-15 Genemedicine Inc Ghrh expression system and methods of use
US7098025B1 (en) 1997-07-25 2006-08-29 Ligand Pharmaceuticals Incorporated Human peroxisome proliferator activated receptor gamma (pparγ) gene regulatory sequences and uses therefor
WO1999014346A2 (en) * 1997-09-19 1999-03-25 Sequitur, Inc. SENSE mRNA THERAPY
EP1892299A2 (en) * 1997-09-19 2008-02-27 Sequitur, Inc. Sense mRNA therapy
EP1892299A3 (en) * 1997-09-19 2011-07-20 Life Technologies Corporation Sense mRNA therapy
WO1999014346A3 (en) * 1997-09-19 1999-05-27 Sequitur Inc SENSE mRNA THERAPY
US6495353B1 (en) 1998-01-21 2002-12-17 Sugen, Inc. Human orthologues of wart
US6656716B1 (en) 1998-04-14 2003-12-02 Sugen, Inc. Polypeptide fragments of human PAK5 protein kinase
US6680170B2 (en) 1998-04-14 2004-01-20 Sugen, Inc. Polynucleotides encoding STE20-related protein kinases and methods of use
US6534483B1 (en) 1998-08-14 2003-03-18 Valentis, Inc. Protected one-vial formulation for nucleic acid molecules, methods of making the same by in-line mixing, and related products and methods
WO2000018908A1 (en) * 1998-09-25 2000-04-06 Aventis Pharma S.A. Use of specific hybrid promoters for controlling tissue expression
FR2783839A1 (en) * 1998-09-25 2000-03-31 Aventis Pharma Sa Hybrid promoter useful for gene expression in smooth-muscle cells includes the enhancer region of a ubiquitous strong promoter/enhancer
US6861442B1 (en) 1998-12-30 2005-03-01 Sugen, Inc. PYK2 and inflammation
US6821946B2 (en) 2000-05-10 2004-11-23 University College London Repair of nerve damage
US6596526B1 (en) 2000-06-09 2003-07-22 Baxter Aktiengesellschaft Furin polypeptides with improved characteristics
WO2002097099A1 (en) * 2001-05-29 2002-12-05 Valentis, Inc. Regulated expression of ghrh
US10626398B2 (en) 2002-02-01 2020-04-21 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10196640B1 (en) 2002-02-01 2019-02-05 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
US10036025B2 (en) 2002-02-01 2018-07-31 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9796978B1 (en) 2002-02-01 2017-10-24 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9919031B2 (en) 2002-02-07 2018-03-20 The Board Of Trustees Of University Of Illinois Use of the insulin-like-growth factor 1 splice variant MGF for the prevention of myocardial damage
US11298320B2 (en) 2002-06-28 2022-04-12 Arbutus Biopharma Corporation Liposomal apparatus and manufacturing methods
US9492386B2 (en) 2002-06-28 2016-11-15 Protiva Biotherapeutics, Inc. Liposomal apparatus and manufacturing methods
US9504651B2 (en) 2002-06-28 2016-11-29 Protiva Biotherapeutics, Inc. Lipid compositions for nucleic acid delivery
US11318098B2 (en) 2002-06-28 2022-05-03 Arbutus Biopharma Corporation Liposomal apparatus and manufacturing methods
US7544855B2 (en) 2004-04-23 2009-06-09 Buck Institute Transgenic mouse whose genome comprises an APP having a mutation at amino acid 664
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015066377A1 (en) * 2013-10-30 2015-05-07 The Curators Of The University Of Missouri Method for scalable skeletal muscle lineage specification and cultivation
US10920196B2 (en) 2013-10-30 2021-02-16 The Curators Of The University Of Missouri Method for scalable skeletal muscle lineage specification and cultivation
IL245292B (en) * 2013-10-30 2022-10-01 Upside Foods Inc Method for scalable skeletal muscle lineage specification and cultivation
IL245292B2 (en) * 2013-10-30 2023-02-01 Upside Foods Inc Method for scalable skeletal muscle lineage specification and cultivation
US11976302B2 (en) 2017-05-06 2024-05-07 Upside Foods, Inc. Compositions and methods for increasing the culture density of a cellular biomass within a cultivation infrastructure
US11479792B2 (en) 2017-07-13 2022-10-25 Upside Foods, Inc. Compositions and methods for increasing the efficiency of cell cultures used for food production
US11708587B2 (en) 2017-07-13 2023-07-25 Upside Foods, Inc. Compositions and methods for increasing the efficiency of cell cultures used for food production

Also Published As

Publication number Publication date
CA2122617A1 (en) 1993-05-13
AU660751B2 (en) 1995-07-06
AU3124693A (en) 1993-06-07
US5298422A (en) 1994-03-29
PT101042A (en) 1994-02-28
US5756264A (en) 1998-05-26
PT101042B (en) 1999-11-30
JPH07500967A (en) 1995-02-02
EP0635060A4 (en) 1995-04-19
EP0635060A1 (en) 1995-01-25

Similar Documents

Publication Publication Date Title
AU660751B2 (en) Myogenic vector systems
US6423693B1 (en) Growth hormone releasing hormone expression system and methods of use, including use in animals
CA2311643C (en) Compositions and methods for inducing gene expression
RU2214280C2 (en) Methods and components for induction of tumor- -specific cytotoxicity
JP2002518018A (en) Erythropoietin analog-human serum albumin fusion
CA2215387A1 (en) Modulators of ob gene and screening methods therefor
CN113316639A (en) Treatment of gonadal-associated viruses for the treatment of pompe disease
CA2274314C (en) Insulin-like growth factor i (igf-i) expression system and methods of use
Bchini et al. Precocious mammary gland development and milk protein synthesis in transgenic mice ubiquitously expressing human growth hormone
US5476926A (en) Adipocyte-specific DNA sequences and use thereof in the production of transgenic animals exhibiting altered fat tissue metabolism
EP0672156B1 (en) Bovine heat shock promoter and uses thereof
AU669713B2 (en) Constitutive and inducible epidermal vector systems
EP0771874B1 (en) Transgenic protein production
AU770982B2 (en) Insulin-like growth factor I (IGF-I) expression system and methods of use
CA2058351A1 (en) Avian growth hormone receptor and binding protein
US20020028193A1 (en) Recombinant beta2-adrenergic receptor delivery and use in treating airway and vascular diseases
WO1997042337A1 (en) Gene expression in monocytes and macrophages
CN116390745A (en) Viral vectors encoding GLP-1 receptor agonist fusions and their use in the treatment of feline metabolic diseases
WO1999049060A1 (en) Use of growth hormone binding protein with a nuclear localization sequence (nls-ghbp)
AU5366298A (en) Insulin-like growth factor i (igf-i) expression system and methods of use
Arman Isolation of DNA-derived single-stranded aptamers specific for the growth hormone receptor (GHR) and characterization of the intracellular region of the GHR involved in signaling

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BG CA CS FI HU JP NO PL RO RU US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
EX32 Extension under rule 32 effected after completion of technical preparation for international publication

Free format text: UA

EX32 Extension under rule 32 effected after completion of technical preparation for international publication

Free format text: UA

WWE Wipo information: entry into national phase

Ref document number: 2122617

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1992925044

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1992925044

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1992925044

Country of ref document: EP