WO1990009443A1 - Animal model for benign prostatic disease - Google Patents

Animal model for benign prostatic disease Download PDF

Info

Publication number
WO1990009443A1
WO1990009443A1 PCT/US1990/000926 US9000926W WO9009443A1 WO 1990009443 A1 WO1990009443 A1 WO 1990009443A1 US 9000926 W US9000926 W US 9000926W WO 9009443 A1 WO9009443 A1 WO 9009443A1
Authority
WO
WIPO (PCT)
Prior art keywords
animal
gene
int
sequence
recombinant gene
Prior art date
Application number
PCT/US1990/000926
Other languages
French (fr)
Inventor
Philip Leder
William J. Muller
Original Assignee
President And Fellows Of Harvard College
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 President And Fellows Of Harvard College filed Critical President And Fellows Of Harvard College
Priority to KR1019900702293A priority Critical patent/KR910700338A/en
Publication of WO1990009443A1 publication Critical patent/WO1990009443A1/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
    • 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
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/20Animal model comprising regulated expression system
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases

Definitions

  • Transgenic animals carry a gene which has been introduced into the germline of the animal, or an ancestor of the animal, at an early (usually one cell) developmental stage.
  • transgenic animals whose germ and somatic cells contain an activated oncogene sequence introduced into the animal at an embryonic stage.
  • Wagner et al. (1981) Proc. Nat. Aca. Sci. 78, 5016; and Stewart et al. (1982) Science 217, 1046 describe transgenic mice containing human globin genes.
  • Constantini et al. (1981) Nature 294, 92; and Lacy et al. (1983) Cell 34, 343 describe transgenic mice containing rabbit globin genes.
  • McKnight et al. (1983) Cell 34, 335 describes transgenic mice containing the chicken transferrin gene. Brinster et al.
  • Science 222, 809 describes transgenic mice containing the human growth hormone gene fused to a metallothionein promoter sequence.
  • Neoplasia is believed to result at least in part from abnormal expression of an endogenous gene, either at abnormally high levels or in some altered or mutated form (Hayward et al., 1981, Nature 290:475-480; Payne et al., 1982, Nature 295:209-214; Reddy et al., 1982, Nature 300:149-152; Tabin et al., 1982, Nature 300:143-149; Taparowski et al., 1982, Nature 300:762-765). Nusse et al., 1982, Cell 31:99-109 and Peters et al., 1982, J. ViroL. 42:880-888
  • int-1 and int-2 which harbor an MMTV provirus in a majority of virally induced mammary tumors. These loci share no apparent homology with known cellular or viral oncogenes.
  • Dickson et al., 1984, Cell 37:529-536 showed that more than 50% of the mammary tumors arising in BR6 mice contain an acquired MMTV provirus integrated within a defined 25 kb domain of DN ⁇ on mouse chromosome 7.
  • Moore et al., 1986, EMBO J. 5:919-924 characterized the int-2 region and isolated a cDNA clone which contains the int-2 gene and which may encode a protein of approximately 27 kD.
  • FGF FGF-related oncogenes.
  • Basic and acidic FGF have been implicated in endothelial cell proliferation and migration during angiogenesis, in mesodermal induction in Xenopus
  • the invention features a male transgenic non-human vertebrate animal (preferably a mammal such as a rodent, e.g., a mouse) containing germ cells and somatic cells which contain a recombinant gene which is substantially homologous with a vertebrate gene in the int-2/FGF family which is capable of promoting (i.e., increases the probablility of developing) benign disease of the prostate, e.g., benign prostatic hyperplasia or hypertrophy (respectively, increase in cell number or cell size).
  • the recombinant gene (i.e., the gene as it exists prior to introduction into the animal) is introduced into the animal, or an ancestor of the animal, at an embryonic stage (preferably the one-cell, or fertilized oocyte, stage, and generally not later than about the 8-cell stage).
  • the recombinant gene preferably is substantially homologous with (i.e., greater than 50% homologous in terms of encoded amino acid sequence) a naturally occurring, most preferably endogenous, vertebrate gene in the int-2/FGF gene family of murine growth factor-encoding genes or their vertebrate
  • FGF acidic or basic fibroblast growth factor
  • FGF-5 the murine FGF-5 gene
  • EGF epidermal growth factor
  • IGF-1, IGF-2 the murine insulin-like growth factor-1 and -2
  • TGF ⁇ ⁇ -transforming growth factor
  • hst/KS3 gene The non-murine version of each of these genes will in most cases differ to some extent from its murine counterpart.
  • endogenous genes genes from other species can also be used, e.g., the human int-2 gene can be introduced into mice.
  • the recombinant gene can be not only a
  • transgenic animals can be made.
  • One method involves the use of a transfecting retrovirus containing the transgene.
  • Another method involves directly injecting the transgene into the embryo.
  • Yet another method employs the embryonic stem cell methodology known to workers in this field.
  • transcription of the recombinant gene is under the control of a promoter sequence different from the promoter sequence controlling transcription of the endogenous coding sequence.
  • Transcription of the recombinant gene can also be under the control of a synthetic promoter se ⁇ uence.
  • the promoter sequence controlling transcription of the recombinant gene is active (i.e., can promote gene
  • the promoter that controls transcription of the recombinant gene may be of viral origin; examples are promoters sometimes derived from mouse mammary tumor virus (MMTV) and cytomegalovirus (CMV).
  • MMTV mouse mammary tumor virus
  • CMV cytomegalovirus
  • founder means the animal into which the recombinant gene was introduced at the one cell mouse embryo stage.
  • F means the animal into which the recombinant gene was introduced at the one cell mouse embryo stage.
  • the animals of the invention can be used as models to test for agents potentially useful in the treatment of benign prostatic hyperplasia or hypertrophy in humans; benign prostate hyperplasia is a serious medical condition affecting a large percentage of the human male population at some time.
  • the agent to be tested can be administered to an animal of the invention and abnormal prostatic growth monitored.
  • the animals of the invention can also be used to test a material suspected of promoting benign prostatic hypertrophy or hyperplasia, by exposing the animal to the material and determining accelerated abnormal growth as an indicator of benign hyperplasia. These tests can be extremely sensitive because of the propensity of the transgenic animals of the invention to develop abnormal tissue growth. This sensitivity will permit potential drug treatments or suspect materials to be tested in much smaller amounts than the amounts used currently in the study of
  • the animals of the invention can also be used as a source of cells for cell culture.
  • Cells from the animals may advantageously exhibit desirable properties of both normal and transformed cultured cells; i.e., they will be normal or nearly normal morphologically and physiologically, but can, like cells such as NIH3T3 cells, be cultured for long, and perhaps indefinite, periods of time.
  • the promoter sequence controlling transcription of the recombinant gene sequence is inducible
  • cell growth rate and other culture characteristics can be controlled by adding or eliminating the inducing factor.
  • Fig. 1 is a diagrammatic representation of the MMTV-c-int-2 coding inserts from the plasmids constructed in Fig. 2.
  • Fig. 2 is a diagrammatic representation of
  • Fig. 3 (a)-(d) is the DNA sequence of the c-int-2 gene (from Moore et al., 1986, EMBO J. 5:919).
  • Fig. 4 is a comparison of amino acid sequences of proteins which are members of the FGF family, aligned to show maximal homology.
  • the murine int-2 gene was originally identified as a common integration site for MMTV and is implicated in mammary tumorigenesis in certain strains of mice (Peters et al., 1983, Cell 33:364-377; Dickson et al., 1984, Cell 37:529-536).
  • the int-2 gene is not detectably expressed in normal mammary glands, yet transcripts accumulate in many mammary tumors as a consequence of proviral insertion 5' or 3' of the int-2 gene
  • int-2 is highly restricted in normal uninfected cells; int-2-specific transcripts are found in normal mouse embryos of around 7.5 days of gestation, but are not round in normal adults tissues (Dickson et al., 1984, supra; Jakobovits et al., 1986, Proc. Nat. Aca. Sci. 83:7806). Thus, a role may exist in early mouse development for the int-2 gene product.
  • An expressible proto-oncogene of the invention was constructed by fusing int-2 coding sequences to viral
  • c-int-2 MMTV long terminal repeat
  • Fig. 1 illustrates the mouse c-int-2 gene fusion; restriction sites are shown; solid arrows below the constructions represent the promoter in the MMTV LTR; and the int-2 gene is shown by the boxed region labeled c-int 2.
  • the size (in Kb) of the major fragment produced by digestion with HindIII that will hybridize with the int-2 probe is given for each construction.
  • MMTV DNA fragment from the plasmid pA9 described in Huang et al. (1981) Cell 27:245, including the region required for glucocorticoid control, the MMTV promoter, and the cap site, is carried on the plasmid pMMTVneu NT (Muller et al., 1988, Cell 54:105).
  • this control region directs expression of an inducible protein called p21; the neu gene in pMMTVneu NT is controlled by this same region and is also inducible.
  • Fig. 2 illustrates the construction of the c-int-2 gene fusions.
  • the mouse c-int-2 gene whose DNA sequence is given in Fig. 3, was obtained from plasmid pKC3 (Moore et al., 1986, E.M.B.O. Journal 5:919).
  • the c-int-2 coding plasmid pKC3 was partially digested with HindIII and completely digested with ClaI, and then ligated to a ClaI + HindIII restriction fragment from pMMTVneu NT, bearing the MMTV regulatory
  • the MMTV LTR was isolated on a SalI + ClaI restriction fragment derived from pMMTVneu NT, and inserted into the unique ClaI site of pMMTVc-int-2 by ligation of the cohesive ClaI ends, treatment of the non-ligated, noncohesive ends with the Klenow fragment of DNA Polymerase I, and blunt end ligation, according to conventional techniques.
  • a plasmid was produced, pcMMTVc-int-2, which contains the entire MMTV LTR fused to the c-int-2 cDNA. In order to ensure proper expression of these gene fusions, these constructs also contain SV40-derived splicing and polyadenylation signals
  • MMTV-int-2 gene fusions were incorporated into the germ ceils of mice as follows.
  • pMMTVc-int-2 DNA was prepared for injection by digestion with 4 units each of ClaI and NaeI per ⁇ g of DNA for 1 hour at 37°C, electrophoresed through a 1% agarose gel, and purified as described by Sinn et al. (1987) Cell 49:465.
  • pcMMTVc-int-2 DNA was treated
  • each pup born was analyzed using DNA taken from the tail in a Southern hybridization. In each case, DNA was extracted from 1.5 cm sections of tail, as
  • nucleic acid pellet was resuspended in 200 ⁇ l of 10 mM Tris-Cl pH 7.4 , 0.1 mM EDTA, and 10 ⁇ g was digested with BamHI, electrophoresed through
  • RNAase ribonuclease
  • a probe illustrated as a solid box below the construct was used. Transcription from the MMTV promoter of the constructs will protect the probe and be revealed as two bands,
  • RNAase protection assay is described as follows.
  • Labelled single-stranded probe fragments were isolated on 8M urea 5% acrylamide gels, electroeluted, and hybridized to total RNA in a modification of the procedure of Berk et al. (1977) Cell 12, 721.
  • Hybridization temperatures were varied according to the GC content in the region of the probe expected to hybridize to mRNA.
  • RNAase A and RNAase T 1 were added to the hybridizations.
  • RNAase digestions were carried out at 37oC for 15 min.
  • the samples were then ethanol-precipitated and electrophoresed on thin 8M urea 5% acrylamide gels.
  • the tissues analyzed were prostate, seminal vesicle, vas deferens, and salivary gland
  • RNA from each of these tissues was analyzed.
  • the antisense probe used in this analysis is transgene specific and yields the same two protected fragments described above, corresponding to the SV40 region of the MMTV-c-int-2 fusion genes.
  • Table 1 shows results of RNase protection assays of MMTV-int-2 fusion gene specific transcripts in RNA from different tissue sources. Relative RNA levels are indicated by + (low), ++ (intermediate), or +++ (high). The results in presented in Table I show that transgene specific transcripts were detected in the
  • TG male NR transgene carrier
  • NR transgene NR
  • Table I also shows that the male NR transgene carrier (TG, NR, or transgene NR) expressed the transgene in the prostate and salivary glands, while the female transgenic NR animals expressed abundant levels of transgene specific transcripts in the mammary gland.
  • TG.NF mammary gland specific expression of the c-neu oncogene initially results in a lactation defect followed by the synchronous appearance of tumors involving mammary glands in the transgenic mouse.
  • transgenic lines (100% of NR males and the NS-F male) resulted in uniform enlargement of this gland.
  • Transgenic animals of the invention can be used for testing agents that may cure the disease, or relieve its symptoms, or for testing for agents that may promote prostate disease. Treatment
  • Transgenic animals of the invention are most useful as animal models for agents and procedures useful in treating or diagnosing benign prostatic hyperplasia or hypertrophy in humans. Treatments that potentially cure this disease, or relieve its symptoms, may be tested first in a trangenic animal which exhibits benign prostatic hyperplasia or hypertrophy by administering the potential treatment to the animal and
  • the animals of the invention can be used to test a material suspected of promoting prostate hyperplasia as
  • the transgenic animals most susceptible to developing abnormal growth are selected, by exposing the animals to a low dosage of a known agent, and selecting those which first develop benign prostatic hyperplasia or
  • the selected animals and their descendants are used as test animals by exposing them to the material suspected of promoting abnormal tissue growth and determining this growth as an indicator of benign prostatic disease. Less sensitive animals are used to test more strongly suspect materials.
  • Animals of the desired sensitivity can be selected by varying the type and concentration of a known agent, e.g. a carcinogen or a hormone, used in the selection process.
  • a known agent e.g. a carcinogen or a hormone
  • the selected test animals can consist of those which spontaneously develop benign prostatic
  • the transgenic animals of the invention can be used as a source of cells for cell culture. Tissues of transgenic animals are analyzed for the presence of the activated recombinant gene, either by directly analyzing DNA or RNA, or by assaying the tissue for the protein expressed by the gene. Cells of tissues carrying the gene can be cultured, using standard tissue culture techniques, and used, e.g., to study the causes of benign prostatic disease at the cellular and tissue levels.
  • any species of transgenic animal can be employed.
  • a species e.g., a primate such as the rhesus monkey, which is evolutionarily closer to humans than mice.
  • Non-mammals e.g., birds such as chickens, can be used as well.

Abstract

A male transgenic non human vertebrate animal containing germ cells and somatic cells which contain a recombinant gene which is substantially homologous with a vertebrate gene in the $i(int)-2/$i(FGF) family which is capable of promoting benign prostatic hyperplasia or hypertrophy in said transgenic vertebrate, said gene being introduced into the animal, or an ancestor of the animal, at an embryonic stage.

Description

ANIMAL MODEL FOR BENIGN PROSTATIC DISEASE
Background of the Invention
This invention relates to transgenic animals. Transgenic animals carry a gene which has been introduced into the germline of the animal, or an ancestor of the animal, at an early (usually one cell) developmental stage. Leder et al. U.S. Patent No. 4,736,866, hereby
incorporated by reference, describes transgenic animals whose germ and somatic cells contain an activated oncogene sequence introduced into the animal at an embryonic stage. Wagner et al. (1981) Proc. Nat. Aca. Sci. 78, 5016; and Stewart et al. (1982) Science 217, 1046 describe transgenic mice containing human globin genes. Constantini et al. (1981) Nature 294, 92; and Lacy et al. (1983) Cell 34, 343 describe transgenic mice containing rabbit globin genes. McKnight et al. (1983) Cell 34, 335 describes transgenic mice containing the chicken transferrin gene. Brinster et al. (1983) Nature 306, 332 describes transgenic mice containing a functionally rearranged immunoglobulin gene. Palmiter et al. (1982) Nature 300, 611 describes transgenic mice containing the rat growth hormone gene fused to a heavy metal-inducible metallothionein prpmoter sequence. Palmiter et al. (1982) Cell.29, 701 describes transgenic mice containing a thymidine kinase gene fused to a metalothionein promoter sequence. Palmiter et al. (1983).
Science 222, 809 describes transgenic mice containing the human growth hormone gene fused to a metallothionein promoter sequence.
The existence of potential oncogenes in the DNA of normal somatic cells has been demonstrated by Weinberg, 1981, Biochem. Biophys. Acta 651:25-35 and Bishop et al., 1982, The Molecular Biology of Tumor Viruses, Part III, RNA Tumor Viruses, Chp. 9, Weiss et al., eds., Cold Spring Harbor, N.Y., Cold Spring Harbor Laboratory. Neoplasia is believed to result at least in part from abnormal expression of an endogenous gene, either at abnormally high levels or in some altered or mutated form (Hayward et al., 1981, Nature 290:475-480; Payne et al., 1982, Nature 295:209-214; Reddy et al., 1982, Nature 300:149-152; Tabin et al., 1982, Nature 300:143-149; Taparowski et al., 1982, Nature 300:762-765). Nusse et al., 1982, Cell 31:99-109 and Peters et al., 1982, J. ViroL. 42:880-888
identified two cellular loci, int-1 and int-2, which harbor an MMTV provirus in a majority of virally induced mammary tumors. These loci share no apparent homology with known cellular or viral oncogenes. Dickson et al., 1984, Cell 37:529-536 showed that more than 50% of the mammary tumors arising in BR6 mice contain an acquired MMTV provirus integrated within a defined 25 kb domain of DNλ on mouse chromosome 7. Moore et al., 1986, EMBO J. 5:919-924 characterized the int-2 region and isolated a cDNA clone which contains the int-2 gene and which may encode a protein of approximately 27 kD.
There is demonstrable homology between the int-2 gene and members of the family of fibroblast growth factor
(FGF)-related oncogenes. Basic and acidic FGF have been implicated in endothelial cell proliferation and migration during angiogenesis, in mesodermal induction in Xenopus
(Gospodarowicz, 1986, Mol. Cell. Endocrinol. 46:187-204;
Folkman and Klagsbrun, 1987, Science 235:442-447; Slack et al., 1987, Nature 326:197-200), and in the formation of nonmammary human tumors (Delli Bovi et al., 1987, Cell 50:729-737 and Taira et al., 1987, Proc. Nat. Aca. Sci. 84:2980-2984).
Summary of the Invention
In general l, the invention features a male transgenic non-human vertebrate animal (preferably a mammal such as a rodent, e.g., a mouse) containing germ cells and somatic cells which contain a recombinant gene which is substantially homologous with a vertebrate gene in the int-2/FGF family which is capable of promoting (i.e., increases the probablility of developing) benign disease of the prostate, e.g., benign prostatic hyperplasia or hypertrophy (respectively, increase in cell number or cell size). The recombinant gene (i.e., the gene as it exists prior to introduction into the animal) is introduced into the animal, or an ancestor of the animal, at an embryonic stage (preferably the one-cell, or fertilized oocyte, stage, and generally not later than about the 8-cell stage). The recombinant gene preferably is substantially homologous with (i.e., greater than 50% homologous in terms of encoded amino acid sequence) a naturally occurring, most preferably endogenous, vertebrate gene in the int-2/FGF gene family of murine growth factor-encoding genes or their vertebrate
counterparts, including the murine acidic or basic fibroblast growth factor (FGF) genes, the murine FGF-5 gene, the murine epidermal growth factor (EGF) gene, the murine insulin-like growth factor-1 and -2 (IGF-1, IGF-2) genes, the murine
α-transforming growth factor (TGFα) gene, or the murine
hst/KS3 gene. The non-murine version of each of these genes will in most cases differ to some extent from its murine counterpart. As mentioned above, it is preferred to use endogenous genes, but genes from other species can also be used, e.g., the human int-2 gene can be introduced into mice. in addition, the recombinant gene can be not only a
vertebrate-derived gene or sequence thereof, but also the viral counterpart.
DNA sequences of the genes of the FGF family, aligned to show maximal homology, are shown in Fig. 4. Any recombinant gene or effective sequence thereof derived from this family might be used to produce the transgenic animals of the
invention.
There are several means by which transgenic animals can be made. One method involves the use of a transfecting retrovirus containing the transgene. Another method involves directly injecting the transgene into the embryo. Yet another method employs the embryonic stem cell methodology known to workers in this field.
Preferably, transcription of the recombinant gene is under the control of a promoter sequence different from the promoter sequence controlling transcription of the endogenous coding sequence. Transcription of the recombinant gene can also be under the control of a synthetic promoter seσuence. Preferably, the promoter sequence controlling transcription of the recombinant gene is active (i.e., can promote gene
expression) in the male urogenital tract of the males of the species to which the male animal of the invention belongs, and is active in prostate tissue. The promoter that controls transcription of the recombinant gene may be of viral origin; examples are promoters sometimes derived from mouse mammary tumor virus (MMTV) and cytomegalovirus (CMV).
Introduction of the recombinant gene at the fertilized oocyte stage ensures that the gene sequence will be present in all of the germ cells and somatic cells of the transgenic
"founder" animal. (As used herein, founder (abbreviated "F") means the animal into which the recombinant gene was introduced at the one cell mouse embryo stage.) The presence of the recombinant gene sequence in the germ cells of the transgenic founder animal in turn means that approximately half of the founder animal's descendants will carry the activated
recombinant gene sequence in all of their germ cells and somatic cells. Introduction of the recombinant gene sequence at a later embryonic stage might result in the gene's absence from some somatic cells of the founder animal, but the
descendants of such an animal that inherit the gene will carry the activated recombinant gene in all of their germ cells and somatic cells. The animals of the invention can be used as models to test for agents potentially useful in the treatment of benign prostatic hyperplasia or hypertrophy in humans; benign prostate hyperplasia is a serious medical condition affecting a large percentage of the human male population at some time. The agent to be tested can be administered to an animal of the invention and abnormal prostatic growth monitored. The animals of the invention can also be used to test a material suspected of promoting benign prostatic hypertrophy or hyperplasia, by exposing the animal to the material and determining accelerated abnormal growth as an indicator of benign hyperplasia. These tests can be extremely sensitive because of the propensity of the transgenic animals of the invention to develop abnormal tissue growth. This sensitivity will permit potential drug treatments or suspect materials to be tested in much smaller amounts than the amounts used currently in the study of
abnormal tissue growth, and thus will minimize one source of criticism of current methods, that their validity is
questionable because the amounts of the tested material used is greatly in excess of amounts to which humans are likely to be exposed. Until now, there have been no satisfactory animal models in which benign prostatic hypertrophy or hyperplasia can be made to occur in a reliable and predictable fashion in a substantial proportion of animals.
The animals of the invention can also be used as a source of cells for cell culture. Cells from the animals may advantageously exhibit desirable properties of both normal and transformed cultured cells; i.e., they will be normal or nearly normal morphologically and physiologically, but can, like cells such as NIH3T3 cells, be cultured for long, and perhaps indefinite, periods of time. Further, where the promoter sequence controlling transcription of the recombinant gene sequence is inducible, cell growth rate and other culture characteristics can be controlled by adding or eliminating the inducing factor. Other features and advantages of the invention will be apparent from the description of the preferred embodiments thereof, and from the claims.
Description of the Preferred Embodiments The drawings will first briefly be described.
Drawings
Fig. 1 is a diagrammatic representation of the MMTV-c-int-2 coding inserts from the plasmids constructed in Fig. 2.
Fig. 2 is a diagrammatic representation of
construction of the MMTV-c-int-2-containing plasmids,
pMMTVc-int-2 and pcMMTVc-int-2.
Fig. 3 (a)-(d) is the DNA sequence of the c-int-2 gene (from Moore et al., 1986, EMBO J. 5:919).
Fig. 4 is a comparison of amino acid sequences of proteins which are members of the FGF family, aligned to show maximal homology.
Int-2 Gene Expression
The murine int-2 gene was originally identified as a common integration site for MMTV and is implicated in mammary tumorigenesis in certain strains of mice (Peters et al., 1983, Cell 33:364-377; Dickson et al., 1984, Cell 37:529-536). The int-2 gene is not detectably expressed in normal mammary glands, yet transcripts accumulate in many mammary tumors as a consequence of proviral insertion 5' or 3' of the int-2 gene
(Dickson et al., 1984, supra; Moore et al., 1986, supra; Peters et al., 1986, Nature 320:628-631). Expression of int-2 is highly restricted in normal uninfected cells; int-2-specific transcripts are found in normal mouse embryos of around 7.5 days of gestation, but are not round in normal adults tissues (Dickson et al., 1984, supra; Jakobovits et al., 1986, Proc. Nat. Aca. Sci. 83:7806). Thus, a role may exist in early mouse development for the int-2 gene product. MMTV-int-2 Fused Genes
An expressible proto-oncogene of the invention was constructed by fusing int-2 coding sequences to viral
regulatory sequences; in this case, the mouse int-2 gene
(c-int-2) and the MMTV long terminal repeat (LTR) were fused, according to conventional techniques, described in Maniatis et al. (1982) Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory). Fig. 1 illustrates the mouse c-int-2 gene fusion; restriction sites are shown; solid arrows below the constructions represent the promoter in the MMTV LTR; and the int-2 gene is shown by the boxed region labeled c-int 2. The size (in Kb) of the major fragment produced by digestion with HindIII that will hybridize with the int-2 probe is given for each construction. These MMTV-int-2 constructions were made as follows.
An MMTV DNA fragment from the plasmid pA9, described in Huang et al. (1981) Cell 27:245, including the region required for glucocorticoid control, the MMTV promoter, and the cap site, is carried on the plasmid pMMTVneu NT (Muller et al., 1988, Cell 54:105). In the pA9 construct, this control region directs expression of an inducible protein called p21; the neu gene in pMMTVneu NT is controlled by this same region and is also inducible.
Fig. 2 illustrates the construction of the c-int-2 gene fusions. The mouse c-int-2 gene, whose DNA sequence is given in Fig. 3, was obtained from plasmid pKC3 (Moore et al., 1986, E.M.B.O. Journal 5:919). The c-int-2 coding plasmid pKC3 was partially digested with HindIII and completely digested with ClaI, and then ligated to a ClaI + HindIII restriction fragment from pMMTVneu NT, bearing the MMTV regulatory
sequences. Recombinant DNA clones containing the ΗMTV LTR fragment upstream of and directing the expression of the c-int-2 gene were verified by restriction enzyme analysis of plasmid DNA. The MMTV-c-int-2 fusion gene can be inducibly expressed in this construct. pMMTVc-int-2 does not contain MMTV LTR sequences upstream of the ClaI site; therefore, a second construct was made which contains the entire MMTV LTR linked to c-int-2
sequences. As shown in Fig. 2, the MMTV LTR was isolated on a SalI + ClaI restriction fragment derived from pMMTVneu NT, and inserted into the unique ClaI site of pMMTVc-int-2 by ligation of the cohesive ClaI ends, treatment of the non-ligated, noncohesive ends with the Klenow fragment of DNA Polymerase I, and blunt end ligation, according to conventional techniques. A plasmid was produced, pcMMTVc-int-2, which contains the entire MMTV LTR fused to the c-int-2 cDNA. In order to ensure proper expression of these gene fusions, these constructs also contain SV40-derived splicing and polyadenylation signals
(designated "SV40" in Figs. 1 and 2) downstream of the c-int-2 coding region.
These constructions were verified by multiple restriction enzyme digestions and were found to be free of detectable rearrangements.
Production of Transgenic Mice Containing MMTV-c-int-2 Fusions
The above MMTV-int-2 gene fusions were incorporated into the germ ceils of mice as follows. pMMTVc-int-2 DNA was prepared for injection by digestion with 4 units each of ClaI and NaeI per μg of DNA for 1 hour at 37°C, electrophoresed through a 1% agarose gel, and purified as described by Sinn et al. (1987) Cell 49:465. pcMMTVc-int-2 DNA was treated
identically except that it was digested only with Nael. Each DNA segment was separately injected into the pronuclei of fertilized one-cell mouse eggs derived from the FVB/NHd inbred strain (Taconic Laboratory, Germantown, NJ); this resulted in about 500 copies of linearized plasmid per pronucleus.
Following micro-injection, viable eggs were transferred to the oviducts of pseudopregnant Swiss-Webster mice (Taconic Farms, Germantown, PA), as described by Wagner et al. (1981) Proc. Nat. Aca. Sci. 78:5016. Mice were housed in an environmentally controlled facility maintained on a 10 hour dark: 14 hour light cycle. The eggs in the foster females were allowed to develop to term.
Analysis of Transgenic Mice
At four weeks of age, each pup born was analyzed using DNA taken from the tail in a Southern hybridization. In each case, DNA was extracted from 1.5 cm sections of tail, as
described by Davis et al. (1980) Meth. Enzymol. 65:405, with the modification that one chloroform extraction was performed prior to ethanol precipitation. The nucleic acid pellet was resuspended in 200 μl of 10 mM Tris-Cl pH 7.4 , 0.1 mM EDTA, and 10 μg was digested with BamHI, electrophoresed through
1.0% agarose, and transferred to nitrocellulose, as described by Southern (1975) J. Mol. Biol. 98:503. Filters were
hybridized overnight to probes in the presence of 101 dextran sulfate and washed twice in 2X SSC, 0.1% SDS at 64°C. The c-int-2 DNA probe was labeled by nick-translation with 32P dCTP, according to Rigby et al., 1977, J. Mol. Biol. 113:237.
The Southern hybridization indicated that four founder mice had retained an MMTV-int-2 fusion. Two founder animals, the NR-F female and the NS-F male, had integrated the int-2 gene from the pMMTV-c-int2 construct, and the other two, the NW-F male and the NX-F male, had integrated the int-2 gene from the pcMMTV-c-int2 construct. The NR-F transgenic female gave both male and female offspring containing the MMTV-c-int-2 gene fusion. Because they exhibited prostate hyperplasia, the NS-F and NW-F male transgenic offspring were sterile.
Transcription of the MMTV-c-int-2 Gene in Transgenic Mice
Transcription of the newly acquired genes in tissues was determined by extracting RNA from the tissues and assaying the RNA in a ribonuclease (RNAase) protection procedure as follows. The excised tissue was rinsed in 5.0 ml cold Hank's buffered saline and total RNA was isolated by the method of Chrigwin et al. (1979) Biochemistry 18:5294, using the CsCl gradient modification. RNA pellets were washed twice by reprecipitation in ethanol and quantitated by absorbance at 260 nm. An appropriate single stranded, uniformly labeled RNA probe was prepared as described by Melton et al. (1984) Nucl. Acids Res. 12:7035. To test for transcription of the pMMTV-c-int-2 gene fusions of Fig. 1, for example, a probe illustrated as a solid box below the construct was used. Transcription from the MMTV promoter of the constructs will protect the probe and be revealed as two bands,
approximately 200 and 125 base pairs long, corresponding to a transcribed but noncoding SV40 sequence used in the construction of the MMTV-c-int-2 fusion genes; transcription of the endogenous int-2 gene, if it is expressed in the cell, will produce RNA that will not be protected, since the SV40 sequence is not present in normal cells. The RNAase protection assay is described as follows.
Labelled single-stranded probe fragments were isolated on 8M urea 5% acrylamide gels, electroeluted, and hybridized to total RNA in a modification of the procedure of Berk et al. (1977) Cell 12, 721. The hybridization mixture contained 50,000 cpm to 100,000 cpm of probe (SA = 108 cpm/μg), 10 μg total cellular RNA, 50% formamide, 500 mM NaCl, 20 mM Tris pH 7.5 1 mM EDTA, as described in Melton et al. (1984, supra). Hybridization temperatures were varied according to the GC content in the region of the probe expected to hybridize to mRNA. The hybridizations were terminated by the addition of 1500 units of RNAase A and RNAase T1 (Sigma, St. Louis, MO). RNAase digestions were carried out at 37ºC for 15 min. The samples were then ethanol-precipitated and electrophoresed on thin 8M urea 5% acrylamide gels.
The tissues analyzed were prostate, seminal vesicle, vas deferens, and salivary gland
10μg of total RNA from each of these tissues was analyzed. The antisense probe used in this analysis is transgene specific and yields the same two protected fragments described above, corresponding to the SV40 region of the MMTV-c-int-2 fusion genes. Table 1 shows results of RNase protection assays of MMTV-int-2 fusion gene specific transcripts in RNA from different tissue sources. Relative RNA levels are indicated by + (low), ++ (intermediate), or +++ (high). The results in presented in Table I show that transgene specific transcripts were detected in the
prostate, seminal vesicles, vas deferens, and salivary glands derived from the NS-F animal. Table I also shows that the male NR transgene carrier (TG, NR, or transgene NR) expressed the transgene in the prostate and salivary glands, while the female transgenic NR animals expressed abundant levels of transgene specific transcripts in the mammary gland. In the MMTV/c-neu strain, TG.NF, mammary gland specific expression of the c-neu oncogene initially results in a lactation defect followed by the synchronous appearance of tumors involving mammary glands in the transgenic mouse.
Expression of the MMTV/int-2 fusion gene in the prostate gland in males derived from two independent
transgenic lines (100% of NR males and the NS-F male) resulted in uniform enlargement of this gland. This
enlargement was closely associated with low fertility exhibited by male transgenic animals. Histological analyses of the enlarged prostates revealed tissue hyperplasia and hypertrophy without evidence of malignant transformation; the prostate growths were shown to be benign by their inability to grow after reimplantation into nu/nu mice.
Use
Transgenic animals of the invention can be used for testing agents that may cure the disease, or relieve its symptoms, or for testing for agents that may promote prostate disease. Treatment
Transgenic animals of the invention are most useful as animal models for agents and procedures useful in treating or diagnosing benign prostatic hyperplasia or hypertrophy in humans. Treatments that potentially cure this disease, or relieve its symptoms, may be tested first in a trangenic animal which exhibits benign prostatic hyperplasia or hypertrophy by administering the potential treatment to the animal and
observing the effects, and comparing the treated animals to untreated controls.
Testing
The animals of the invention can be used to test a material suspected of promoting prostate hyperplasia as
follows. If the animals are to be used to test materials thought to be only weakly effective in promoting abnormal tissue growth, the transgenic animals most susceptible to developing abnormal growth are selected, by exposing the animals to a low dosage of a known agent, and selecting those which first develop benign prostatic hyperplasia or
hypertrophy. The selected animals and their descendants are used as test animals by exposing them to the material suspected of promoting abnormal tissue growth and determining this growth as an indicator of benign prostatic disease. Less sensitive animals are used to test more strongly suspect materials.
Animals of the desired sensitivity can be selected by varying the type and concentration of a known agent, e.g. a carcinogen or a hormone, used in the selection process. When extreme sensitivity is desired, the selected test animals can consist of those which spontaneously develop benign prostatic
hyperplasia or hypertrophy.
Tissue Culture
The transgenic animals of the invention can be used as a source of cells for cell culture. Tissues of transgenic animals are analyzed for the presence of the activated recombinant gene, either by directly analyzing DNA or RNA, or by assaying the tissue for the protein expressed by the gene. Cells of tissues carrying the gene can be cultured, using standard tissue culture techniques, and used, e.g., to study the causes of benign prostatic disease at the cellular and tissue levels.
Other Embodiments
Other embodiments are within the following claims. For example, any species of transgenic animal can be employed. In some circumstance, for instance, it may be desirable to use a species, e.g., a primate such as the rhesus monkey, which is evolutionarily closer to humans than mice. Non-mammals, e.g., birds such as chickens, can be used as well.
Table 1. Transgene Expression in HHTV/c-int-2 Mice
Tissue
Strain Sex Mammary Salivary Prostate Seminal Vas
vesicles Deferens
NS-F male ND + ++ +++ ++
TG.NR female +++ + ND ND ND male ND + ++ + -

Claims

Claims 1. A male transgenic non-human vertebrate animal containing germ cells and somatic cells which contain a recombinant gene which is substantially homologous with a vertebrate gene in the int-2/FGF family which is capable of promoting benign prostatic hyperplasia or hypertrophy in said transgenic vertebrate, said gene being introduced into the animal, or an ancestor of the animal, at an embryonic stage.
2. The animal of claim, 1, wherein a chromosome of said animal includes an endogenous coding sequence
substantially the same as a coding sequence of said
recombinant gene.
3. The animal of claim 2 , said recombinant gene being integrated into a chromosome of said animal at a site different from the location of said endogenous coding sequence.
4. The animal of claim 2 wherein transcription of said recombinant gene sequence is under the control of a promoter sequence different from the promoter sequence controlling the transcription of said endogenous coding sequence.
5. The animal of claim 4 wherein said promoter sequence controlling transcription of said recombinant gene is inducible.
6. The animal of claim 5 wherein said promoter is active in the male urogenital tract of the species of animal to which said transgenic animal belongs.
7. The animal of claim 1 wherein transcription of said recombinant gene is under the control of a viral promoter sequence .
8. The animal of claim 9 wherein said viral promoter sequence comprises a sequence of an MMTV promoter.
9. The animal of claim 1 wherein said animal is a mammal.
10. The mammal of claim 9, wherein said mammal is a rodent.
PCT/US1990/000926 1989-02-17 1990-02-16 Animal model for benign prostatic disease WO1990009443A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1019900702293A KR910700338A (en) 1989-02-17 1990-02-16 Animal model for benign prostate disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/312,641 US5175383A (en) 1989-02-17 1989-02-17 Animal model for benign prostatic disease
US312,641 1989-02-17

Publications (1)

Publication Number Publication Date
WO1990009443A1 true WO1990009443A1 (en) 1990-08-23

Family

ID=23212372

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/000926 WO1990009443A1 (en) 1989-02-17 1990-02-16 Animal model for benign prostatic disease

Country Status (7)

Country Link
US (1) US5175383A (en)
EP (1) EP0458908A1 (en)
JP (1) JPH04505704A (en)
KR (1) KR910700338A (en)
AU (1) AU5278590A (en)
CA (1) CA2046915A1 (en)
WO (1) WO1990009443A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994003594A1 (en) * 1992-08-07 1994-02-17 University Of Manitoba Androgen regulation with dna sequences of rat probasin gene
WO1999013068A2 (en) * 1997-09-12 1999-03-18 Washington University Transgenic animal model of prostate cancer

Families Citing this family (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5714146A (en) * 1992-08-26 1998-02-03 Board Of Regents Of The University Of Washington IL-4 bone therapy
US6264915B1 (en) 1992-09-13 2001-07-24 The President And Fellows Of Harvard College Process for detecting potential carcinogens
US5981175A (en) * 1993-01-07 1999-11-09 Genpharm Internation, Inc. Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
US5489743A (en) * 1993-01-19 1996-02-06 Amgen Inc. Transgenic animal models for thrombocytopenia
AU6586494A (en) * 1993-04-14 1994-11-08 United States Of America As Represented By The Secretary Of The Navy, The Transgenic animal model for autoimmune diseases
US5523226A (en) 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US6001589A (en) * 1993-06-11 1999-12-14 Pbl Biomedical Laboratories, Inc. Method of identifying proteins modified by disease states related thereto
WO1994029344A1 (en) * 1993-06-11 1994-12-22 Pestka Biomedical Laboratories, Inc. Super proteins including interferons and interleukins
US5849995A (en) * 1993-09-27 1998-12-15 The University Of British Columbia Mouse model for Huntington's Disease and related DNA sequences
US5602309A (en) * 1993-10-04 1997-02-11 University Of Kentucky Research Foundation Transgenic mice which overexpress nerve growth factor
EP0648841B1 (en) 1993-10-13 2001-09-19 Northeastern Ohio Universities Genomic DNA of human Cholesterol 7alpha-hydroxylase and methods for using it
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
US5858774A (en) * 1994-05-12 1999-01-12 The Research Foundation Of State University Of New York Antisense DNA constructs for expression of hybrid MRNAs driven by inducible, tissue-specific promoters
US5675060A (en) * 1994-05-19 1997-10-07 Institut National De La Sante Et De La Recherche Medicale Transgenic arthritic mice expressing a T-cell receptor transgene
WO1995034640A1 (en) * 1994-06-13 1995-12-21 Henry Ford Health System A novel neuronal-neonatal gene: neuronatin
US20030152909A1 (en) * 1994-11-16 2003-08-14 Mitrani Eduardo N. In vitro micro-organs, and uses related thereto
KR100268713B1 (en) * 1994-11-21 2000-10-16 서정선 Transgenic mouse deficient in t-cells
KR100268714B1 (en) * 1994-11-21 2000-10-16 서정선 Diabetes-inducing transgenic mouse
US5859308A (en) * 1994-12-29 1999-01-12 University Of Medicine And Denistry Of New Jersey Transgenic animals and related aspects
US5814300A (en) * 1995-03-03 1998-09-29 Cephalon, Inc. Gene-targeted non-human mammals deficient in the SOD-1 gene
AU5531796A (en) * 1995-04-11 1996-10-30 Trustees Of Columbia University In The City Of New York, The Reagent specific for apolipoprotein-j polymorphisms and uses thereof
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
US5728807A (en) * 1995-05-16 1998-03-17 Ramot-University Authority For Applied Research And Industrial Development, Ltd. Mutated proteins associated with ataxia-telangiectasia
US6187991B1 (en) 1995-05-23 2001-02-13 Pfizer Inc Transgenic animal models for type II diabetes mellitus
US6248555B1 (en) 1995-08-31 2001-06-19 The General Hospital Corporation Genetic alterations related to familial alzheimer's disease
KR19990044317A (en) * 1995-09-01 1999-06-25 라미 핀클러 ; 하나넬 크바탄스키 Manipulation and detection of amylophosphatase 2C-P2Cα-expression
US5859311A (en) * 1995-11-27 1999-01-12 University Of Kentucky Research Foundation Transgenic mice which overexpress neurotrophin-3 (NT-3) and methods of use
US6140309A (en) * 1996-03-12 2000-10-31 University Of South Florida Vasoactive effects and free radical generation by β-amyloid peptides
US6011019A (en) * 1996-03-12 2000-01-04 University Of South Florida Vasoactive effects and free radical generation by β-amyloid peptides
US20060021070A1 (en) * 1996-04-01 2006-01-26 University of Massachusetts, a Public Institution of Higher Education of the Commonwealth of MA Production of chimeric bovine or porcine animals using cultured inner cell mass
US5919901A (en) * 1996-04-08 1999-07-06 Bayer Corporation Neuropeptide Y receptor Y5 and nucleic acid sequences
US5965392A (en) * 1996-04-08 1999-10-12 Bayer Corporation Neuropeptide Y receptor Y5 and nucleic acid sequences
US5824838A (en) * 1996-05-09 1998-10-20 Cedars-Sinai Medical Center Transgenic mouse model for pituitary disorders associated with LIF overexpression and/or GH underexpression, and its use for testing therapeutic drugs for the conditions
EP0954574A2 (en) 1996-07-01 1999-11-10 Jim A. Wright Oligonucleotides from the untranslated regions of housekeeping genes and methods of using same to modulate cell growth
US5898094A (en) * 1996-10-21 1999-04-27 University Of South Florida Transgenic mice expressing APPK670N,M671L and a mutant presenilin transgenes
US7008765B1 (en) * 1997-04-10 2006-03-07 The Johns Hopkins University PCA3, PCA3 genes, and methods of use
CA2298410A1 (en) 1997-07-24 1999-02-04 Duke University Intracellular inhibitors of gq protein signaling
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
WO1999009046A1 (en) 1997-08-21 1999-02-25 Quark Biotech, Inc. Hypoxia-regulated genes
US7321023B2 (en) * 1997-11-07 2008-01-22 Incyte Corporation SP16 protein
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
US6583333B1 (en) 1998-05-12 2003-06-24 Cold Spring Harbor Laboratory Lymphoma-susceptible transgenic mice and methods for studying drug sensitivity of lymphomas
US6548737B1 (en) 1998-05-15 2003-04-15 Barnes-Jewish Hospital Transgenic mice deficient in natural killer cells
ATE338120T2 (en) 1998-11-27 2006-09-15 Ucb Sa COMPOSITIONS AND METHODS FOR INCREASE BONE MINERALIZATION
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
US7105315B2 (en) * 1999-06-16 2006-09-12 Incyte Genomics, Inc. Transmembrane protein differentially expressed in cancer
US6864235B1 (en) * 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US6911429B2 (en) * 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
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
EP1190082A4 (en) * 1999-06-25 2003-07-02 Yissum Res Dev Co Method of inducing angiogenesis by micro-organs
JP2003506038A (en) * 1999-07-29 2003-02-18 ダナ−ファーバー キャンサー インスティテュート インク. An inducible cancer model to study the molecular basis of host tumor cell interactions in vivo
CA2374745A1 (en) * 1999-08-09 2001-02-15 Incyte Genomics, Inc. Proteases and protease inhibitors
US6875582B1 (en) 1999-08-19 2005-04-05 Omniscience Pharmaceuticals, Inc. Methods and targets of antibiotic resistance
AU7353500A (en) * 1999-09-10 2001-04-10 Incyte Genomics, Inc. Apoptosis proteins
US6998232B1 (en) 1999-09-27 2006-02-14 Quark Biotech, Inc. Methods of diagnosing bladder cancer
EP2275559A3 (en) 1999-09-28 2011-03-23 Shire Human Genetic Therapies, Inc. Optimized messenger RNA
PT1222266E (en) 1999-09-29 2006-07-31 Diagnocure Inc PCA3 MESSENGER RNA IN BENEFICIAL AND MALIGNAL PROSTATE TISSUES
CA2426669A1 (en) * 1999-10-22 2001-05-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education .alpha.1-3 galactosyltransferase gene and promoter
US7385106B2 (en) * 2000-01-24 2008-06-10 Ramot At Tel Aviv University Ltd. Plants tolerant of environmental stress conditions, methods of generating same and novel polynucleotide sequence utilized thereby
CA2398743C (en) 2000-01-28 2015-06-23 The Government Of The United States Of America Novel mhc class ii restricted t cell epitopes from the cancer antigen, ny eso-1
WO2001066742A2 (en) * 2000-03-03 2001-09-13 Incyte Genomics, Inc. G-protein coupled receptors
WO2001098323A2 (en) 2000-06-16 2001-12-27 Incyte Genomics, Inc. G-protein coupled receptors
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
US7608704B2 (en) 2000-11-08 2009-10-27 Incyte Corporation Secreted proteins
AU2002233614B2 (en) * 2001-03-22 2007-05-24 Scentgene Pollination Ltd. Method of enhancing entomophilous
EP1967525B1 (en) 2001-05-08 2012-11-14 Darwin Molecular Corporation A method for regulating immune function in primates using the foxp3 protein
IL159756A0 (en) 2001-07-12 2004-06-20 Univ Massachusetts IN VIVO PRODUCTION OF SMALL INTERFERING RNAs THAT MEDIATE GENE SILENCING
EP1900815B1 (en) 2001-07-12 2016-09-07 University of Massachusetts In vivo production of small interfering RNAs that mediate gene silencing
US8178128B2 (en) * 2001-12-05 2012-05-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Nanoparticles containing polymeric nucleic acid homologs
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
US7378492B2 (en) * 2002-02-20 2008-05-27 Incyte Corporation CD40-related receptor that binds CD40L
US20040005571A1 (en) * 2002-07-03 2004-01-08 Hawley R. Scott Axs gene and protein and methods related thereto
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
US20070276126A1 (en) * 2002-08-13 2007-11-29 Incyte Corporation Cell adhesion and extracellular matrix proteins
ES2338111T3 (en) * 2002-08-21 2010-05-04 Revivicor, Inc. PIG ANIMALS THAT LACK OF ANY EXPRESSION OF ALPHA 1.3 FUNCTIONAL GALACTOSILTRANSPHERASE.
US20070219353A1 (en) * 2002-09-03 2007-09-20 Incyte Corporation Immune Response Associated Proteins
WO2004033636A2 (en) * 2002-10-04 2004-04-22 Incyte Corporation Protein modification and maintenance molecules
US20050164275A1 (en) * 2002-10-18 2005-07-28 Incyte Corporation Phosphodiesterases
AU2003284968A1 (en) * 2002-10-25 2004-05-13 University Of South Florida Methods and compounds for disruption of cd40r/cd40l signaling in the treatment of alzheimer's disease
EP1557670A4 (en) * 2002-10-30 2007-03-07 Ishihara Sangyo Kaisha REGULATION OF INTERACTION BETWEEN RAPL AND Rap1
AU2003286911A1 (en) * 2002-11-04 2004-06-07 Dana-Farber Cancer Institute, Inc. In vivo imaging of e2f-regulated bioluminescent proteins
US20070009886A1 (en) * 2002-11-12 2007-01-11 Incyte Corporation Carbohydrate-associated proteins
WO2004044165A2 (en) * 2002-11-13 2004-05-27 Incyte Corporation Lipid-associated proteins
AU2003298675A1 (en) * 2002-11-22 2004-06-18 You-Jun Fei NaCT AS A TARGET FOR LIFESPAN EXPANSION AND WEIGHT REDUCTION
US20070009516A1 (en) * 2002-11-26 2007-01-11 Tran Uyen K Immune response-associated proteins
ES2427853T3 (en) 2003-02-07 2013-11-04 Diagnocure Inc. Procedure to detect prostate cancer in a sample
US7829682B1 (en) 2003-04-30 2010-11-09 Incyte Corporation Human β-adrenergic receptor kinase nucleic acid molecule
EP2314692B1 (en) 2003-06-02 2021-02-24 University of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
CA2528012C (en) 2003-06-02 2015-11-24 University Of Massachusetts Methods and compositions for controlling efficacy of rna silencing
WO2005010039A1 (en) * 2003-07-31 2005-02-03 Pfizer Products Inc. Bsep polypeptide variants and uses thereof
CA2548080A1 (en) * 2003-11-05 2005-05-26 University Of Pittsburgh Porcine isogloboside 3 synthase protein, cdna, genomic organization, and regulatory region
WO2005059100A2 (en) * 2003-12-12 2005-06-30 New York University Methods and compositions relating to cystatin c
WO2005089411A2 (en) 2004-03-17 2005-09-29 Revivicor, Inc. Tissue products from animals lacking functional alpha 1,3 galactosyl transferase
SI1733056T1 (en) 2004-03-31 2013-10-30 The General Hospital Corporation Method to determine responsiveness of cancer to epidermal growth factor receptor targeting treatments
US20060019241A1 (en) * 2004-04-20 2006-01-26 Vadivel Ganapathy Na+ and CI-coupled transport system for endogenous opioid peptides
CA2491067A1 (en) * 2004-12-24 2006-06-24 Stichting Katholieke Universiteit Mrna rations in urinary sediments and/or urine as a prognostic marker for prostate cancer
US20070061899A1 (en) * 2005-04-13 2007-03-15 The Trustees Of Columbia University In The City Of New York Transgenic mammal with constitutive, tissue-specific cytokine expression and use as a cancer model
CN103044432B (en) 2005-12-23 2016-08-03 阿里亚德医药股份有限公司 Bicyclic heteroaryl compounds
WO2007088418A1 (en) 2006-01-31 2007-08-09 Multi Gene Vascular Systems, Inc. Drug-eluting intravascular prostheses and methods of use
CN101400349A (en) * 2006-03-01 2009-04-01 罗斯坎普研究有限责任公司 Compounds for inhibiting beta-amyloid production
CA2664318C (en) 2006-09-14 2017-05-23 Andrew L. Pearlman Long lasting drug formulations
US8454948B2 (en) 2006-09-14 2013-06-04 Medgenics Medical Israel Ltd. Long lasting drug formulations
US9127084B2 (en) 2006-09-14 2015-09-08 Medgenics Medical Israel 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
US7943590B2 (en) * 2007-03-16 2011-05-17 Multi-Gene Vascular Systems Ltd. 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
AU2009248923B2 (en) 2008-05-21 2015-01-29 Takeda Pharmaceutical Company Limited Phosphorous derivatives as kinase inhibitors
US8241846B1 (en) 2008-09-08 2012-08-14 Institut De Recherches Cliniques De Montreal Hedgehog pathway modulation and uses thereof for treating, preventing and/or diagnosing cancer
EP2365809B8 (en) 2008-11-12 2018-10-10 Ariad Pharmaceuticals, Inc. Pyrazinopyrazines and derivatives as kinase inhibitors
CN110322924A (en) 2010-04-29 2019-10-11 加利福尼亚大学董事会 Utilize the approach recognition methods (PARADIGM) of the data integration about gene group model
US9646134B2 (en) 2010-05-25 2017-05-09 The Regents Of The University Of California Bambam: parallel comparative analysis of high-throughput sequencing data
EP2577538A1 (en) 2010-05-25 2013-04-10 The Regents of the University of California Bambam: parallel comparative analysis of high-throughput sequencing data
TW201522629A (en) 2013-10-24 2015-06-16 Medgenics Medical Israel Ltd Micro-organs providing sustained delivery of a therapeutic polypeptide and methods of use thereof
EA035145B1 (en) 2014-10-21 2020-05-06 Ариад Фармасьютикалз, Инк. Crystalline forms of 5-chloro-n4-[-2-(dimethylphosphoryl)phenyl]-n2-{2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]pyrimidine-2,4-diamine
CN112425568A (en) * 2020-12-11 2021-03-02 上海市计划生育科学研究所 Method for establishing benign prostatic hyperplasia BPH dog model with EMT characteristics

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4736866A (en) * 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4736866A (en) * 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US4736866B1 (en) * 1984-06-22 1988-04-12 Transgenic non-human mammals

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Cell (Cambridge, USA), Volume 46, Issued 26 September 1986, BROWN et al., "A retrovirus vector expressing the putative mammary oncogene int-1 causes partial transformation of a mammary epithelial cell line", page 1001-1009. See the entire dcument. *
Cell (Cambridge, USA), Volume 55, Issued 18 November 1988, TSUKAMOTO et al., "Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice", pages 619-625. See the entire document. *
Nature (London, VK), Volume 320, Issued 17 April 1986, PETERS et al., "Concertedactivation of two potential proto-oncogenes in carcinomas induced by mouse mammary tumor virus", pages 628-631. *
Proceedings of the National Academy of Sciences (Washington, USA), Volume 83, Issued October 1986, JAKOBOVITS et al., "Two proto-oncogebes, int-1 and int-2, are independently regulated during mouse development", pages 7806-7810. *
See also references of EP0458908A4 *
The EMBO Journal (Oxford, England), Volume 5, No.5, Issued 1986, CASEY et al., "Sequence, topography and protein coding potential of mouse int-2: a putative oncogene activated by mouse mammary tumors virus", pages 919-924. See the entire document. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994003594A1 (en) * 1992-08-07 1994-02-17 University Of Manitoba Androgen regulation with dna sequences of rat probasin gene
US5783681A (en) * 1992-08-07 1998-07-21 University Of Manitoba Androgen regulation with DNA sequences of rat probasin gene
US5952488A (en) * 1992-08-07 1999-09-14 University Of Manitoba Androgen regulation with DNA sequences of rat probasin gene
WO1999013068A2 (en) * 1997-09-12 1999-03-18 Washington University Transgenic animal model of prostate cancer
WO1999013068A3 (en) * 1997-09-12 1999-05-06 Univ Washington Transgenic animal model of prostate cancer

Also Published As

Publication number Publication date
KR910700338A (en) 1991-03-14
US5175383A (en) 1992-12-29
CA2046915A1 (en) 1990-08-18
EP0458908A1 (en) 1991-12-04
AU5278590A (en) 1990-09-05
JPH04505704A (en) 1992-10-08
EP0458908A4 (en) 1991-09-30

Similar Documents

Publication Publication Date Title
US5175383A (en) Animal model for benign prostatic disease
CA1341442C (en) Transgenic animals
Pierce et al. Inhibition of mammary duct development but not alveolar outgrowth during pregnancy in transgenic mice expressing active TGF-beta 1.
McAndrew et al. Targeting of transforming growth factor-alpha expression to pituitary lactotrophs in transgenic mice results in selective lactotroph proliferation and adenomas
US5087571A (en) Method for providing a cell culture from a transgenic non-human mammal
Andres et al. Ha-ras oncogene expression directed by a milk protein gene promoter: tissue specificity, hormonal regulation, and tumor induction in transgenic mice.
Behringer et al. Dwarf mice produced by genetic ablation of growth hormone-expressing cells.
AU631571B2 (en) Method of gene transfer into chickens and other avian species
Bailleul et al. Skin hyperkeratosis and papilloma formation in transgenic mice expressing a ras oncogene from a suprabasal keratin promoter
AU683227B2 (en) Tetracycline repressor-mediated binary regulation system for control of gene expression in transgenic animals
Lira et al. Identification of rat growth hormone genomic sequences targeting pituitary expression in transgenic mice.
Kollias et al. Ectopic expression of Thy-1 in the kidneys of transgenic mice induces functional and proliferative abnormalities
Held et al. T antigen expression and tumorigenesis in transgenic mice containing a mouse major urinary protein/SV40 T antigen hybrid gene.
Botteri et al. Unexpected thymic hyperplasia in transgenic mice harboring a neuronal promoter fused with simian virus 40 large T antigen
DiMattia et al. The Pit-1 gene is regulated by distinct early and late pituitary-specific enhancers
WO1998033887A1 (en) Growth differentiation factor-8
US20010053358A1 (en) Growth differentiation factor-11
US5789651A (en) Isolation and characterization of Agouti: a diabetes/obesity related gene
Allison et al. Tissue-specific and hormonal regulation of the gene for rat prostatic steroid-binding protein in transgenic mice
WO1992006104A1 (en) Adipocyte-specific dna sequences and use for the production of transgenic animals exhibiting altered fat metabolism
EP1190042A2 (en) Methods for manipulating the avian genome
Soares et al. Tissue-specific and hormonally regulated expression of a rat α2u globulin gene in transgenic mice
JP2003513645A (en) Melanocortin-3 receptor deficient cells, non-human transgenic animals and methods for selecting compounds that regulate body weight
US20100107265A1 (en) Double-muscling in mammals
EP0648841B1 (en) Genomic DNA of human Cholesterol 7alpha-hydroxylase and methods for using it

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BB BG BR CA DK FI HU JP KP KR LK MC MG MW NO RO SD SU

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE BF BJ CF CG CH CM DE DK ES FR GA GB IT LU ML MR NL SE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 1990905076

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2046915

Country of ref document: CA

WWP Wipo information: published in national office

Ref document number: 1990905076

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1990905076

Country of ref document: EP