CA2127450C - Transgenic animal models for alzheimer's disease - Google Patents

Transgenic animal models for alzheimer's disease Download PDF

Info

Publication number
CA2127450C
CA2127450C CA002127450A CA2127450A CA2127450C CA 2127450 C CA2127450 C CA 2127450C CA 002127450 A CA002127450 A CA 002127450A CA 2127450 A CA2127450 A CA 2127450A CA 2127450 C CA2127450 C CA 2127450C
Authority
CA
Canada
Prior art keywords
promoter
app
app770
protein
construct
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002127450A
Other languages
French (fr)
Other versions
CA2127450A1 (en
Inventor
Samuel Wadsworth
Benjamin Snyder
Vermuri B. Reddy
Chamer Wei
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Elan Pharmaceuticals LLC
Original Assignee
Elan Pharmaceuticals LLC
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 Elan Pharmaceuticals LLC filed Critical Elan Pharmaceuticals LLC
Publication of CA2127450A1 publication Critical patent/CA2127450A1/en
Application granted granted Critical
Publication of CA2127450C publication Critical patent/CA2127450C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction 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
    • 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
    • A01K67/0278Humanized animals, e.g. knockin
    • 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
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • 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
    • 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
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0312Animal model for Alzheimer's disease
    • 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

Abstract

The construction of transgenic animal models for testing potential treatments for Alzheimer's disease are described. The models are characterized by a greater similarity to the conditions existing in naturally occurring Alzheimer's disease, based on expression of all three forms of the .beta.-amyloid precursor protein (APP), APP695, APP751, and APP770, as well as various point mutations based on naturally occurring mutations, such as the London and Indiana familial Alzheimer's disease (FAD) mutations at amino acid 717, and predicted mutations in the APP gene. The APP gene constructs are prepared using the naturally occurring promoter, as well as inducible promoters such as the mouse metallothionine promoter, which can be regulated by addition of heavy metals such as zinc to the animal's water or diet, and promoters such as the rat neuron specific enolase promoter, human .beta. actin gene promoter, human platelet derived growth factor B (PDGF-B) chain gene promoter, rat sodium channel gene promoter, mouse myelin basic protein gene promoter, human copper-zinc superoxide dismutase gene promoter and mammalian POU-domain regulatory gene promoter. The constructs are introduced into animal embryos using standard techniques such as microinjection. Animal cells can be isolated from the transgenic animals or prepared using the same contructs with standard techniques such as lipofection or electroporation. The transgenic animals, or animal cells, are used to screen for compounds altering the pathological course of Alzheimer's disease as measured by their effect on the amount and histopathology of APP and .beta.-amyloid peptide in the animals, as well as by behavioral alterations.

Description

V~,O 93/ 14200 Transgenic Animal Models for Alzheimer's Disease Background of the Invention ' Transgenic technology is described for the production of animals that exhibit symptoms of human Alzheimer's disease through the expression of the Alzheimer's precursor protein or a modified version thereof .
Alzheimer's Disease (AD) is a degenerative disorder of the brain first described by Alios Alzheimer in 1907 after examining one of his patients who suffered drastic reduction in cognitive abilities and had generalized dementia ("The early story of Alzheimer's Disease", edited by Bick K., Amaducci L., and Pepeu G. (Raven Press, New York 1987). It is the leading cause of dementia in elderly persons. AD
patients have increased problems with memory loss and intellectual functions which progress to the point where they cannot function as normal individuals.
With the loss of intellectual skills the patients exhibit personality changes, socially inappropriate actions and schizophrenia ("A guide to the understanding of Alzheimer's Disease and related disorders", edited by Jorm AF.; (New York University Press, New York 1987). AD is devastating for both victims and their families, for there is no effective palliative or preventive treatment for the inevitable neurodegeneration. The most common problems in the Alzheimer's patient are inability to dress unaided, restlessness by day, urinary incontinence and sleep disturbances. The family members report embarrassment, anxiety, depression, and a decreased social life.
The impact of AD on society and on the national economy is enormous. It is expected that the demented elderly population in the United States will increase by 41% by the year 2000. It is expensive for the health care systems that must provide institutional and ancillary care for the patients at an estimated '~
annual cost of $40 billion (Jorm, 1987; Fisher, LM:
New York Times, August 23, 1989 D1 "Alzheimer's Disease", edited by Reisberg, B.; (The Free Press, New York & London 1983). These factors imply preventive action must be taken to decrease AD incidence by allocating resources into AD research.
At a macroscopic level, the brains of AD patients are usually smaller, sometimes weighing less than 1,000 grams. At a microscopic level, the histopathological symptoms of AD include neurofibrillary tangles (NFT), neuritic plaques, and degeneration of neurons. AD patients exhibit degeneration of nerve cells in the frontal and temporal cortex of the cerebral cortex, pyramidal neurons of hippocampus, neurons in the medial, medial central, and cortical nuclei of the amygdala, noradrenergic neurons in the locus coeruleus, and the neurons in the basal forebrain cholinergic system.
Loss of neurons in the cholinergic system leads to a consistent deficit in cholinergic presynaptic markers in AD (Reisberg, 1983; "Alzheimer's Disease and related disorders, research and development" edited by Kelly WE; (Charles C. Thomas, Springfield, IL. 1984).
AD is associated with neuritic plaques measuring up to 200 ~m in diameter in the cortex, hippocampus, subiculum, hippocampal gyrus, and amygdala. One of the principal constituents of neuritic plaques is amyloid, which is stained by Congo red (Reisberg, 1983; Kelly, 1984). Amyloid plaques are extracellular, pink - or rust-colored in bright field, and birefringent in polarized light. The plaques are composed of polypeptide fibrils and are often present around blood vessels, reducing blood supply to various neurons in the brain.
Various factors such as genetic predisposition, infectious agents, toxins, metals, and head trauma have all been suggested as possible mechanisms of AD

WO 93/14200 i ~ ~ PCT/US92/11276 neuropathy. However, available evidence strongly indicates two distinct types of genetic predisposition for AD. First, molecular analysis has provided evidence for mutations in the amyloid precursor protein (APP) gene in certain AD-stricken families (Goate, et al. Nature 349:704-706 (1991); Murrell, J, et al. Science 254; 97-99, 1991; Chartier-Harlin, M-C, et al. Nature 353, 844-846 (1991)). Second, in certain other families with a clear genetic predisposition to AD, the mutation maps to chromosome 21 but is distinct from the APP locus (Tanzi, R.E., et al. Nature, 331;528-530 (1988)).
Amyloid plaques are abundantly present in AD
patients and in Down's Syndrome individuals surviving to the age of 40. The plaques are also present in the normal aging brain, although at a lower number. These plaques are made up of the amyloid B peptide (I3 peptide) (Glenner and Wong, et al., Biochem. Biophys.
Res. Comm. 120:885-890 (1984)), which is also the main protein constituent in cerebrovascular deposits and neurofibrillary tangles. The /3 peptide is a filamentous material that is arranged in beta-pleated sheets and has a molecular weight of 4.2-4.5 kd. It is a hydrophobic peptide comprising 39-42 amino acids.
The determination of its amino acid sequence led to the cloning of the APP cDNA (Kang, et al., Nature 325:733-735 (1987); Goldgaber, et al., Science 235:877-880 (1987); Robakis et al., Proc. Natl. Acad.
Sci. 84:4190-4194 (1987); Tanzi, et al., Nature 331:528-530 (1988) and genomic APP DNA (Lemaire et al., Nucl. Acids Res. 17:517-522 (1989); Yoshikai, et al., Gene 87, 257-263 (1990). Three forms of APP
cDNAs (APP695, APP751, and APP770) have been isolated, and arise from a single precursor RNA by alternate splicing. The gene spans more than 175 Kb with 18 exons (Yoshikai, et al., 1990). APP contains three extracellular domains, a transmembrane region and a ~~C~~ i cytoplasmic domain. The 8 peptide consists of 28 amino acids just outside the membrane and 14 residues of the hydrophobic transmembrane domain. Thus, the B
peptide is a cleavage product of APP normally found in brain and other tissues such as heart, kidney and spleen. B peptide deposits, however, are usually found only in the brain, although Joachim et al., Nature 341:226-228 (1989) have reported f3 peptide deposits outside the brain in the skin, intestine, and subcutaneous tissues of most AD patients.
The larger alternate forms of APP (APP751, APP770) consist of all of APP695 plus one or two additional domains. APP751 consists of all of APP695 plus an additional 56 amino acids which has homology to the Kunitz family of serine protease inhibitors (KP1) (Tanzi et al., 1988; Weidemann, et al., Cell 57:115-126 (1989); Kitaguchi, et al., Nature 331:530-532 (1988); Tanzi et al., Nature 329, 156 (1987).
APP770 contains APP751 and an additional 19 amino acid domain homologous to the neuron cell surface antigen OX-2 (Weidemann, et al., Cell 57:115-126 (1989);
Kitaguchi et al., 1988). APP is post-translationally modified by the removal of the leader sequence and by the addition of sulfate and sugar groups.
Van Broeckhaven, et al., Science 248:1120-1122 (1990) have demonstrated that the APP gene is tightly linked to hereditary cerebral hemorrhage with amyloidosis (HCHWA-D) in two Dutch families. This was confirmed by the finding of a point mutation in the APP coding region in two Dutch patients (Levy et al., Science 248:1124-1128 (1990). The mutation substituted a glutamine for glutamic acid at position 22 of the f3 peptide (position 618 of APP695). In addition, certain families are genetically predisposed to Alzheimer's disease, a condition referred to as familial Alzheimer's disease (FAD), through mutations resulting in an amino acid replacement at position 717 WO 93/ 14200 ~ ~ ~ PCT/US92/ 11276 of the full length protein (Goate, et al., (1991);
Murrell et al., 1991; Chartier-Harlin et al., 1991).
These mutations co-segregate with the disease within the families and are absent in families with late-onset AD.
There are no proven animal models to study AD, although aging nonhuman primates seem to develop amyloid plaques of f3 peptide in brain parenchyma and in the walls of some meningeal and cortical vessels.
Although aged primates and canines can serve as animal models, they are expensive to maintain and need lengthy study periods. There are no spontaneous animal mutations with sufficient similarities to AD to be useful as experimental models. Various models have been proposed in which some AD-like symptoms may be induced by electrolysis, transplantation of AD brain samples, aluminum chloride, kainic acid or choline analogs (Kisner, et al., Neurobiol. Aging 7;287-292 (1986); Mistry, J.S., et al., J Med Chem 29;337-343 (1986)). Flood, et al. (Proc. Natl. Acad. Sci.
88:3363-3366 (1986), reported amnestic effects in mice of four synthetic peptides homologous to the !3 peptide. Because none of these share with AD either common symptoms, biochemistry or pathogenesis, they are not likely to yield much useful information on etiology or treatment.
Transgenic mice with the human APP promoter linked to E. coli f3-galactosidase (Wirak, D.O., et al., The EMBO J 10;289-296 (1991)) as well as transgenic mice expressing the human APP751 cDNA
(Quon, D, et al. Nature 352, 239-241 (1991)) or subfragment of the cDNA including the f3 peptide (Wirak, D.O., et al., Science 253, 323-325 (1991);
Sandhu, F.A., et al., J. Biol. Chem. 266, 21331-21334 (1991); Kawabata, S. Nature 354, 4?6-478 (1991)) have been produced. Results obtained in the different studies appear to depend upon the source of promoter ~~~ t'~
and the protein coding sequence used. For example, Wirak, et al. (1991) found that in transgenic mice expressing a form of the B peptide, intracellular deposits of "amyloid-like" material, reactive with antibodies prepared against APP were observed but did not find other histopathological disease symptoms.
The intracellular nature of the antibody-reactive material and the lack of other symptoms suggest that this particular transgenic animal is not a faithful model system for Alzheimer's disease. Kawabata et al.
(1991) report the production of amyloid plaques, neurofibrillary tangles, and neuronal cell death in their transgenic animals. In each of these studies, the same peptide fragment, the B peptide plus the 56 remaining C terminal amino acids of APP, was expressed. Wirak et al. (1991) used the human APP
promoter while Kawabata, et al. (1991) used the human thy-1 promoter. In transgenic mice expressing the APP751 cDNA from the neuron-specific enolase promoter of Quon, D., et al., Nature 352, 239-241 (1991), extracellular deposits of material reactive with antibody prepared against APP were observed. What was not shown was whether the deposits contained full-length APP751 or B peptide or both, thus precluding any correlation of the deposits with those present in Alzheimer's disease. Quon et al. (1991) also state that the protein encoded by the APP695 cDNA expressed from the neuron-specific enolase promoter, does not form extracellular immunoreactive deposits. These results raise the possibility that although the f3 pep~_3e is included within the APP695 precursor, use of the neuron-specific enolase promoter in conjunction with the APP695 cDNA may not present an effective Alzheimer's disease model. Furthermore, the presence of APP immunoreactive deposits is not correlated with the age or gene dosage in their particular transgenic model.
Alzheimer's disease is a complex syndrome involving pathological and behavioral aspects. A
useful disease model should take these complexities into account. There are multiple proteins expressed from the gene with certain forms predominating in a given tissue. In the brain, the 695 form is predominant, but the mRNAs for additional forms are also present (Golde et al., Neuron 4; 253-267 (1990)).
It is not known whether the ratio of the different forms changes with the age of the individual. The various protein forms result from alternative splicing such that the KI domain and/or the OX-2 domain may or may not be present in the mature protein. Moreover, the Ii-peptide results from post-translational processing of the precursor protein. This process can change in time as an individual ages, and can be affected by mutations not directly affecting the structure of the f3-peptide: for example, the familial Alzheimer's disease (FAD) mutations at amino acid position 717 in the full length protein (Groate, et al., 1991; Murrell, et al., 1991; Chartier-Harlin, et al., 1991). Given these considerations, the production of universal animal models for Alzheimer's disease necessitates the construction of animal models that take into account the effects of known mutations on the phenotype resulting from the expression of these forms, and the possibility of the ratio of the different forms changing during the lifetime of the animal.
It is therefore an object of the present invention to provide an animal model for Alzheimer's disease that is constructed using transgenic technology.
It is a further object of the present invention to provide transgenic animals that accurately reflect the expression of different forms of the amyloid precursor protein.

~~,~~ ~~Cl _g_ It is a still further object of the present invention to provide transgenic animals characterized by certain genetic abnormalities in the expression of the amyloid precursor protein.
summary of the Invention The construction of transgenic animal models for testing potential treatments for Alzheimer's disease is described. The models are characterized by a greater similarity to the conditions existing in naturally occurring Alzheimer's disease, based on the ability to control expression of one or more of the three forms of the f3-amyloid precursor protein (APP), APP695, APP751, and APP770, or subfragments thereof, as well as various point mutations based on naturally occurring mutations, such as the FAD mutations at amino acid 717, and predicted mutations in the APP
gene. The APP gene constructs are prepared using the naturally occurring APP promoter of human, mouse, or rat origin, as well as inducible promoters such as the mouse metallothionine promoter, which can be regulated by addition of heavy metals such as zinc to the animal's water or diet. Neuron-specific expression of constructs is achieved by using the rat neuron specific enolase promoter.
The constructs are introduced into animal embryos using standard techniques such as microinjection or embryonic stem cells. Cell culture based models can also be prepared by two methods. Cell cultures can be isolated from the transgenic animals or prepared from established cell cultures using the same constructs with standard cell transfection techniques.
The specific constructs that are described employ the following protein coding sequences: the APP770 cDNA; the APP770 CDNA bearing a mutation at amino acid 717; the APP751 cDNA containing the KI protease inhibitor domain without the OX2 domain in the WO 93/14200 PCl"/US92/11276 -g-construct; the APP751 cDNA and bearing a mutation at amino acid 717; the APP695 cDNA; the APP695 cDNA
bearing a mutation at amino acid 717; the APP leader sequence followed by the Ii peptide region plus the remaining carboxy terminal 56 amino acids of APP; the APP leader sequence followed by the t3 peptide region plus the remaining carboxy terminal 56 amino acids with the addition of a mutation at amino acid 717; the APP leader sequence followed by the 13 peptide region;
the I3 peptide region plus the remaining carboxy terminal 56 amino acids of APP; the 8 peptide region plus the remaining carboxy terminal 56 amino acids of APP with the addition of a mutation at amino acid 717;
a combination genomic-cDNA APP gene construct; and a combination genomic-cDNA APP gene construct, with the addition of a mutation at amino acid 717, operably linked to promoters selected from the following: the human APP gene promoter, mouse APP gene promoter, rat APP gene promoter, metallothionine gene promoter, rat neuron specific enolase gene promoter, human 13 actin gene promoter, human platelet derived growth factor B
(PDGF-B) chain gene promoter, rat sodium channel gene promoter, mouse myelin basic protein gene promoter, human copper-zinc superoxide dismutase gene promoter, and mammalian POU-domain regulatory gene promoter.
Additional constructs include a human yeast artificial chromosome construct controlled by the human APP
promoter; a human yeast artificial chromosome construct controlled by the human APP promoter with the addition of a mutation at amino acid 717; the endogenous mouse or rat APP gene modified through the process of homologous recombination between the APP
gene in a mouse or rat embryonic stem (ES) cell and a vector carrying the human APP cDNA of the wild-type such that sequences in the resident rodent chromosomal APP gene beyond the recombination point (the preferred site for recombination is within APP exon 9) are WO 93/142(i0 PCT/US92/11276 replaced by the analogous human sequences; the endogenous mouse or rat APP gene modified through the process of homologous recombination between the APP
gene in a mouse or rat ES cell and a vector carrying the human APP cDNA bearing a mutation at amino acid position 717 such that sequences in the resident rodent chromosomal APP gene beyond the recombination point (the preferred site for recombination is within APP exon 9) are replaced by the analogous human sequences bearing a mutation at amino acid 717. These constructs can be introduced into the transgenic animals and then combined by mating of animals expressing the different constructs.
The transgenic animals, or animal cells, are used to screen for compounds altering the pathological course of Alzheimer's Disease as measured by their effect on the amount and histopathology of APP and 8 peptide in the animals, as well as by behavioral alterations.
Brief Description of the Drawings The boxed portions of the drawings indicate the amino acid coding portions of the constructs. Filled portions indicate the various domains of the protein as indicated in the Figure Legend. Lines indicate sequences in the clones that are 5' or 3' untranslated sequences, flanking genomic sequences, or introns.
The break in the line to the left of the constructs in Figs. 7 and 8 indicates the presence of a long DNA
sequence.
Figure la is a schematic of the APP 770 cDNA
coding sequence.
Figure 1b is a schematic of the APP770 cDNA
coding sequence bearing a mutation at position 717, indicated by FAD.
Figure 2a is a schematic of the APP751 cDNA coding sequence.

Figure 2b is a schematic of the APP751 cDNA
coding sequence bearing a mutation at position 717, indicated by FAD.
Figure 3a is a schematic of the APP695 coding sequenca.
Figure 3b is a schematic of the APP695 cDNA coding sequence bearing a mutation at position 717, indicated by FAD.
Figure 4a is a schematic of a coding sequence for the carboxy terminal portion of APP.
Figure 4b is a schematic of a coding sequence for the carboxy terminal portion of APP bearing a mutation at position 717, indicated by FAD.
Figure 5 is a schematic of a coding sequence for the B peptide portion of APP.
Figure 6a is a schematic of a combination genomic/cDNA coding sequence allowing alternative splicing of the KI and OX2 exons.
Figure 6b is a schematic of a combination genomic/cDNA coding sequence bearing a mutation at position 717 and allowing alternative splicing of the KI and OX2 exons.
Figure 7a is a schematic of a human APP YAC
coding sequence.
Figure 7b is a schematic of a human APP YAC
coding sequence bearing a mutation at position 717.
Figure 8 is a schematic of genetic alteration of the mouse APP gene by homologous recombination between the mouse APP gene in a mouse ES cell and a vector carrying the human APP cDNA (either of the wild-type or FAD mutant form) directed to the exon 9 portion of the gene. As a result of this recombination event, sequences in the resident mouse chromosomal APP gene beyond the recombination point in exon 9 are replaced by the analogous human sequences.

Detailed Description of the Invention The constructs and transgenic animals and animal cells are prepared using the methods and materials described below.
sources of materials.
Restriction endonucleases are obtained from conventional commercial sources such as New England Biolabs (Beverly, MA.), Promega Biological Research Products (Madison, WI.), and Stratagene (LaJolla CA.), etc. Radioactive materials are obtained from conventional commercial sources such as Dupont/NEN or Amersham. Custom-designed oligonucleotides for site-directed mutagenesis are available from any of several commercial providers of such materials such as Bio-synthesis Inc., Lewisville, TX. Kits for carrying out site-directed mutagenesis are available from commercial suppliers such as Promega Biolo-:ical Research Products, Stratagene, etc. Clones of cDNA
including the APP695, APP751, and APP770 fonas of APP
mRNA were obtained directly from Dr. Dmitry Goldgaber, NIH. Libraries of DNA are available from commercial providers such as Stratagene, La Jolla, CA., or Clontech, Palo Alto, CA. PC12 and 3T3 cells were obtained from ATCC (~CRL1721 and ,~CCL92 respectively).
An additional PC12 cell line was obtained from Dr.
Charles Marotta of Harvard Medical School, Massachusetts General Hospital, and McLean Hospital.
Standard cell culture media appropriate to the cell line are obtained from conventional commercial sources such as Gibco/BRL. Murine stem cells, strain D3, were obtained from Dr. Rolf Kemler (Doetschman, et al., J.
Embrvol. Exp. Morphol. 87, 27 (1985) ) . LipofectiriMfor DNA transfection and the drug 6418 for selection of stable transformants are available from Gibco/BRL.

d Isolation of the human APP promoter.
' A cosmid library, constructed from human placental DNA in the pWElS cosmid vector, was screened by hybridization with a 32P-labeled probe prepared by nick-translation (Maniatis, et al. Molecular Cloning:
a laboratory manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 1989)) of the APP770 cDNA
clone. Clones that hybridized with the probe were picked, purified, and characterized by restriction mapping, hybridization, and DNA sequencing. From one such clone containing a long 5' flanking region, a NotI to NruI restriction DNA fragment of approximately 25 kb was isolated. This fragment terminates 2 nucleotides before the initiator methionine codon of the Alzheimer's protein-coding region. This fragment, or a subfragment thereof, is the source of the human APP promoter for the constructs described herein.
Analogous DNA fragments isolated using the same methods from mouse or rat genomic libraries are the source of mouse or rat promoters.
Definition of APP cDNA clones.
The cDNA clone APP-695 is of the form of cDNA
described by Kang, et al., Nature 325:733-735 ((1987), and represents the most predominant form of Alzheimer's protein in the brain. The cDNA clone APP-751 is of the form described by Ponte, P, Nature 331, 525-527 (1988). The cDNA clone APP-770 is of the form described by Kitaguchi, et al. Nature 331:530-532 (1988). This form contains an insert of 225 nucleotides relative to the 695 form. The 225 nucleotide insert encodes for the KI domain as well as the OX-2 domain.
Definition of the APP g~enomic locus.
Characterization of phage and cosmid clones of human genomic DNA clones listed in the table below originally established a minimum size of at least 100 kb for the Alzheimer's gene. There are a total of 18 exons in the APP gene (Lemaire et al., Nucl. Acid Res, 17;517-522, 1989; Yoshikai et al., 1990). These results taken together indicate that the minimum size of the Alzheimer's gene is 175 kb.
I. Table of Alzheimer's Cosmid and Lambda Clones Name of Insert Library Clone Size (Rb1 Assiqned APP Region 1 GPAPP47A 35 25 Kb promoter & 9 Kb intron Cosmid 2 GPAAP36A 35 12 Kb promoter &
22 Kb intron 3 GAPP30A 30-35 5' coding region 4GAPP43A 30-35 exons 9, 10 and 11 1 GAPP6A 12 exon 6 2 GAPP6B 18 exons 4 and 5 3 GAPP20A 20 exon 6 4 GAPP20B 17 exons 4 and 5 Lambda 5 GAPP28A 18 exons 4 and 5 6 GAPP3A 14 exon 6 7 GAPP4A 19 exon 6 8 GAPP10A 16 exons 9, 10 and 11 9 GAPP16A 21 exon 6 Construction of Transg~enes.
The clones bearing various portions of the human APP gene sequence shown in Figs. 1-5 are constructed in an analogous manner. First, the polyA addition signal from SV40 virus as a 253 base pair BcZI to BamHI fragment (Reddy et al., Science 200;494-502 (1978) is cloned into a modified vector from the pUC
series. Next, the cDNA coding sequences (770, 751, or 695) are inserted. Correct orientation and content of the fragments inserted is determined through restriction endonuclease mapping and limited sequencing.
The clones bearing various carboxy terminal portions of the human APP gene sequence shown in Figs.
4 and 5 are constructed through several steps in addition to those indicated above. First, an APP770 cDNA clone is digested with Asp718 which cleaves after position 56 (numbering system of Kang et al., 1987).
The resulting 5' extension is filled in using the Klenow enzyme (Maniatis et al., 1989) and ligated to a hexanucleotide of the following sequence: AGATCT, the recognition site for eglII. After cleavage with BglII, which also cuts after position 1769, and re-ligation, the translational reading frame of the protein is preserved. The truncated protein thus encoded contains the leader sequence, followed by approximately 6 amino acids that precede the B
peptide, followed by the Ii peptide, and the 56 terminal amino acids of APP. The clone in Fig. 5 is created by the introduction through site directed mutagenesis of nucleotide 1913 in the clone of Fig. 4a (numbering system of Kang et al., 1987) to a T thus creating a termination codon directly following the last amino acid codon of the (3 peptide. Each of the APP cDNA sequence clones shown in Figs. 1-5 contains a single NruI site 2 nucleotides upstream from the initiator methionine codon that is used for attachment of the different promoters used to complete each construct.
Expression clones identical to these but bearing mutations at the amino acid 717 of the full length protein, the site of the FAD mutation, are also constructed. Mutations at amino acid 717 are created by site-directed mutagenesis (Vincent, et al., Genes &
bevel. 3, 334-347 (1989)) and include mutations of the wild-type val codon to one of the following codons;
ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, gln.
The preferred method for construction of the combination cDNA/genomic expression clones in Figure 6 is as follows. The TaqI site at position 860 (numbering system of Kang, et al., 1987) in an APP770 WO 93/14200 ~ ~ ~~~~ PCT/US92/11276 ~r~ ~

cDNA clone is converted to an XhoI site by site-directed mutagenesis. Cleavage of the resulting plasmid with XhoI cuts at the new XhoI site and a pre-existing site at 930y and releases the KI and OX-2 coding sequence.
The plasmid thus generated serves as the acceptor for the KI and OX-2 alternative splicing cassette.
The alternative splicing cassette is created through a series of cloning steps. First, the TaqI site at position 860 (numbering system of Kang, et al., 1987) in the genomic clone containing exon 6 and adjacent downstream intron is converted to an XhoI site by site-directed mutagenesis. Cleavage of the resulting plasmid with XhoI cuts at the new XhoI site and an XhoI site within the adjacent intron. This fragment is cloned into the XhoI site in a plasmid vector.
Second, the genomic clone containing exon 9 and adjacent upstream intron is cleaved with XhoI
(position 930) and cloned into the XhoI site of a plasmid vector. These two junction exon/intron fragments are released from their respective plasmid backbones by cleavage with XhoI and either BamHI or BglII, and cloned into the XhoI site of a plasmid vector. The resulting XhoI fragment is cleaved with either BamHI or BgZII and the genomic 6.6 kb BamHI
segment (Kitaguchi et al., 1988) containing the KI and OX-2 coding region along with their flanking intron sequences are inserted. After cleavage with XhoI, this DNA segment is inserted into the XhoI site of the modified APP770 cDNA constructed above. These cloning steps generate a combination cDNA/genomic expression clone that allows cells in a transgenic animal to regulate the inclusion of the KI and OX-2 domains by a natural alternative splicing mechanism. An analogous gene bearing a mutation at amino acid 717 is constructed by using the mutated form of APP770 cDNA
described above.

Activity of Gene Promoters.
Different promoter sequences are used to control expression of APP coding sequences. The ability to regulate expression of the APP gene in transgenic animals is believed to be useful in evaluating the roles of the different APP gene products in AD. The ability to regulate expression of the APP gene in cultured cells is believed to be useful in evaluating expression and processing of the different APP gene products and may provide the basis for cell cultured drug screens.
The metallothionine (MT) promoter is well characterized, has been employed in transgenic animals, and its expression can be regulated through modulation of zinc and glucocorticoid hormone levels (Palmiter et al., Nature 300, 611-615 (1982)).
The human APP promoter is also characterized with regard to expression in the CNS (Wirak et al., 1991).
It is believed that this promoter is useful for accurately reproducing temporal and spatial expression of human APP sequences in the CNS of transgenic rodents. In addition to the human APP promoter, the APP promoter from mouse and rat is used in conjunction with the various wild-type and mutant APP coding sequences. Although the human APP promoter has been shown to have activity in the appropriate regions of the brain of transgenic mice (Wirak et al., 1991), it is believed that the use of a mouse APP promoter in a transgenic mouse or a rat APP promoter in a transgenic rat will offer an even more precise pattern of expression in the CNS of transgenic animals.
As an alternative for the control of human APP
expression in neurons, the rat neuron specific enolase gene promoter is used. This promoter has been shown to direct expression of coding sequences in neurons (Forss-Petter et al., Neuron 5;197-197 (1990)).

Other alternatives for use in controlling human APP expression in neurons include the human I3 actin gene promoter (Ray et al., Genes and Develoument 5:2265-2273 (1991)), the human platelet derived growth factor B (PDGF-B) chain gene promoter (Sasahara et al., Cell 64:217-227 (1991)), the rat sodium channel gene promoter (Maue et al., Neuron 4:223-231 (1990)), the human copper-zinc superoxide dismutase gene promoter (Ceballos-Picot et al., Brain Res. 552:198-214 (1991)), and promoters for members of the mammalian POU-domain regulatory gene family (Xi et al., Nature 340:35-42 (1989)). The POU-domain is the region of similarity between the four mammalian transcription factors Pit-1, Oct-1, Oct-2, and unc-86, and represents a portion of the DNA-binding domain.
These promoters are known or believed to result in expression specifically within the neurons of transgenic animals.
Expression of human APP in non-neuronal brain cells can be directed by the promoter for mouse myelin basic protein (Readhead et al., Cell 48:703-712 (1987)).
Yeast Artificial Chromosomes.
The constructs shown in Figure 7 are constructed as follows. Large segments of human genomic DNA, when cloned into certain vectors, can be propagated as autonomously-replicating units in the yeast cell.
Such vector-borne segments are referred to as yeast artificial chromosomes (YAC; Burke et al. Science 236, 806 (1987)). A human YAC library is commercially available (Clontech, Palo Alto, CA) with an average insert size of 250,000 base pairs (range of 180,000 to 500,000 base pairs). A YAC clone of the Alzheimer's gene can be directly isolated by screening the library with the human APP770 cDNA. The inclusion of all of the essential gene regions in the clone can be confirmed by PCR analysis.

2~ 2~ 45 0 The YAC-APP clone, shown in Figure 7a, is established in embryonic stem (ES) cells by selecting for neomycin resistance encoded by the YAC vector. ES
cells bearing the YAC-APP clone are used to produce transgenic mice by established methods described below under "Transgenic Mice" and "Embryonic Stem Cell Methods". The YAC-APP gene bearing a mutation at amino acid 7I7 (Fig. 7b) is produced through the generation of a YAC library using genomic DNA from a person affected by a mutation at amino acid 717. The clone is identified and established in ES cells as described above.
Genetic Alteration of the Mouse APP Gene.
The nucleotide sequence homology between the human and murine Alzheimer's protein genes is approximately 85~. Within the (3 peptide-coding region, there are three amino acid differences between the two sequences. The val residue that is mutated at amino acid 717 is conserved between mouse, rat, and man. Wild-type rodents do not develop Alzheimer's disease nor do they develop deposits or plaques in their CNS analogous to those present in human Alzheimer's patients. Therefore, it is possible that the human but not the rodent form of f3 peptide is capable of causing disease. Homologous recombination (Capecchi, MR Science 244, 1288-1292 (1989)) can be used to convert the mouse Alzheimer's gene in situ to a gene encoding the human !3 peptide. This recombination is directed to a site downstream from the KI and OX-2 domains, for example, within exon 9, so that the natural alternative splicing mechanisms appropriate to all cells within the transgenic animal can be employed in expressing the final gene product.
Both wild-type (Fig. 8, schematic "a") and mutant (Fig. 8, schematic "b") forms of human cDNA are used to produce transgenic models expressing either the wild-type or mutant forms of APP. The recombination WO 93/14200 ~ ~ ~~ ~ ~~ PCT/US92/11276 ., vector is constructed from a human APP cDNA (695 or 770 form), either wild-type or mutant at amino acid 717. Cleavage of the recombination vector, for example, at the XhoI site within exon 9, promotes homologous recombination within the directly adjacent sequences (Capecchi, 1989). The endogenous APP gene resulting from this event is normal up to the point of recombination, within exon 9 in this example, and consists of the human cDNA sequence thereafter.
Mutant Forms of APP Proteins Expression clones identical to these but bearing mutations at the amino acid 717 of the full length protein, the site of FAD mutations, are also constructed. Mutations at amino acid 717 are created by site-directed mutagenesis (Vincent, et al., 1989) and include mutations of the wild-type val codon to one of the following codons; ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, gln. Mutations of val-717 to ile, phe, and gly, have been described (Goate et al., 1991; Murrell, et al., 1991; Chartier-harlin et al., 1991). None of these naturally-occurring mutations are charged or bulky amino acids.
Therefore it is believed that replacement of val-717 with the other amino acids listed may also promote the FAD syndrome and have properties that are useful for animal AD models.
Preparation of Constructs for Transfections and Microiniections DNA clones for microinjection are cleaved with appropriate enzymes, such as Sall, Notl, etc., and the DNA fragments electrophoresed on 1% agarose gels in TBE buffer (Maniatis et al., 1989). The DNA bands are visualized by staining with ethidium bromide, excised, and placed in dialysis bags containing 0.3 M sodium acetate, pH 7Ø DNA is electroeluted into the dialysis bags, extracted with phenol-chloroform (1:1), and precipitated by two volumes of ethanol. The DNA

is redissolved in 1 ml of low salt buffer (0.2 M NaCl, 20 mM TrisTM, pH 7.4, and 1 mM EDTA) and purified on an Elutip-DrM column. The column is first primed with 3 . ml of high salt buffer (1 M NaCl, 20 mM TrisTM, pH 7.4, and 1 mM EDTA) followed by washing with 5 ml of low salt buffer. The DNA solutions are passed through the column for three times to bind DNA to the column matrix. After one wash with 3 ml of low salt buffer, the DNA is eluted with 0.4 ml of high salt buffer and precipitated by two volumes of ethanol. DNA
concentrations are measured by absorption at 260 nm in a W spectrophotometer. For microinjection, DNA
concentrations are adjusted to 3 ~,g/ml in 5 mM TrisTM, pH 7.4 and 0.1 mM EDTA. Other methods for purification of DNA for microinjection are also described in Hogan, et al., Manipulating the mouse embrvo (cold Spring Harbor Laboratory, Cold Spring Harbor, NY (1986), in Palmiter, et al., Nature 300, 611 (1982), in "The Qiagenologist, Application Protocols", 3rd edition, published by Qiagen, Inc., Chatsworth, CA., and in Maniatis, et al., Molecular Cloning: a laboratory manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 1989).
Construction of Transg~enic Animals.
Animal Sources Animals suitable for transgenic experiments can be obtained from standard commercial sources such as Charles River (Wilmington, MA), Taconic (Germantown, NY), Harlan Sprague Dawley (Indianapolis, IN), etc.
Swiss Webster female mice are preferred for embryo retrieval and transfer. B6D2F~ males can be used for mating and vasectomized Swiss Webster studs can be used to stimulate pseudopregnancy. Vasectomized mice and rats can be obtained from the supplier.
Microin~ection Procedures The procedures for manipulation of the rodent embryo and for microinjection of DNA are described in detail in Hogan et al. Manipulating the mouse embryo, Cold Spring Harbor Laboratory, Cold Spring harbor, NY
(1986), Transqenic Mice Female mice six weeks of age are induced to superovulate with a 5 IU injection (0.1 cc, ip) of pregnant mare serum gonadotropin (PMSG; Sigma) followed 48 hours later by a 5 IU injection (0.1 cc, ip) of human chorionic gonadotropin (hCG; Sigma).
Females are placed with males immediately after hCG
injection. Twenty-one hours after hCG, the mated females are sacrificed by COz asphyxiation or cervical dislocation and embryos are recovered from excised oviducts and placed in Dulbecco's phosphate buffered saline with 0.5% bovine serum albumin (BSA; Sigma).
Surrounding cumulus cells are removed with hyaluronidase (1 mg/ml). Pronuclear embryos are then washed and placed in Earle's balanced salt solution containing 0.5% BSA (EBSS) in a 37.5°C incubator with a humidified atmosphere at 5% C02, 95% air until the time of injection.
Randomly cycling adult female mice are paired with vasectomized males. Swiss Webster or other comparable strains can be used for this purpose.
Recipient females are mated at the same time as donor females. At the time of embryo transfer, the recipient females are anesthetized with an intraperitoneal injection of 0.015 ml of 2.5% avertin per gram of body weight. The oviducts are exposed by a single midline dorsal incision. An incision is then made through the body wall directly over the oviduct. The ovarian bursa is then torn with watchmakers forceps. Embryos to be transferred are placed in DPBS and in the tip of a transfer pipet (about 10-12 embryos). The pipet tip is inserted into the infundibulum and the embryos A

transferred. After the transfer, the incision is closed by two sutures.
TransQenic Rats The procedure for generating transgenic rats is similar to that of mice (Hammer et al., Cell 63;1099-112 (1990)). Thirty day-old female rats are given a subcutaneous injection of 20 IU of PMSG (0.1 cc) and 48 hours later each female placed with a proven male.
At the same time, 40-80 day old females are placed in cages with vasectomized males. These will provide the foster mothers for embryo transfer. The next morning females are checked for vaginal plugs. Females who have mated with vasectomized males are held aside until the time of transfer. Donor females that have mated are sacrificed (C02 asphyxiation) and their oviducts removed, placed in DPBS (Dulbecco's phosphate buffered saline) with 0.5% BSA and the embryos collected. Cumulus cells surrounding the embryos are removed with hyaluronidase (1 mg/ml). The embryos are then washed and placed in EBSS (Earle's balanced salt solution) containing 0.5% BSA in a 37.5°C incubator until the time of microinjection.
Once the embryos are injected, the live embryos are moved to DPBS for transfer into foster mothers.
The foster mothers are anesthetized with ketamine (40 mg/kg, ip) and xylazine (5 mg/kg, ip). A dorsal midline incision is made through the skin and the ovary and oviduct are exposed by an incision through the muscle layer directly over the ovary. The ovarian bursa is torn, the embryos are picked up into the transfer pipet, and the tip of the transfer pipet is inserted into the infundibulum. Approximately 10-12 embryos are transferred into each rat oviduct through the infundibulum. The incision is then closed with sutures, and the foster mothers are housed singly.

Embryonic Stem fESI Cell Methods Introduction of cDNA into ES cells:
Methods for the culturing of ES cells and the subsequent production of transgenic animals, the introduction of DNA into ES cells by a variety of methods such as electroporation, calcium phosphate/DNA
precipitation, and direct injection are described in detail in Teratocarcinomas and embryonic stem cells, a practical approach, ed. E.J. Robertson, (IRL Press 1987 ), Selection or the aesired clone of transgene-containing ES cells is accomplished through one of several means.
In cases involving random gene integration, an APP
clone is co-precipitated with a gene encoding neomycin resistance. Transfection is carried out by one of several methods described in detail in Lovell-Badge, in Teratocarcinomas and embryonic stem cells, a practical approach, ed. E.J. Robertson, (IRL Press 1987) or in Potter et al Proc. Natl. Acad. Sci. USA
81, 7161 (1984). Calcium phosphate/DNA precipitation, direct injection, and electroporation are the preferred methods. In these procedures, 0.5 X 106 ES
cells are plated into tissue culture dishes and transfected with a mixture of the linearized APP clone and 1 mg of pSV2neo DNA (Southern and Berg, J. Mol.
Appl. Gen. 1:327-341 (1982)) precipitated in the presence cf 50 mg lipofectiriMin a final volume of 100 ~,1. The cells are fed with selection medium containing 10% fetal bovine serum in DMEM supplemented with 6418 (between 200 and 500 ~,g/ml). Colonies of cells resistant to 6418 are isolated using cloning rings and expanded. DNA is extracted from drug resistant clones and Southern blotting experiments using an APP770 cDNA probe are used to identify those clones carrying the APP sequences. In some experiments, PCR methods are used to identify the clones of interest.

WO 93/ 14200 212 7 4 5 ~ P~T/US92/ 11276 DNA molecules introduced into ES cells can also be integrated into the chromosome through the process of homologous recombination, described by Capecchi, (1989). Direct injection results in a high efficiency of integration. Desired clones are identified through PCR of DNA prepared from pools of injected ES cells.
Positive cells within the pools are identified by PCR
subsequent to cell cloning (2immer and Gruss, ature 338, 150-153 (1989). DNA introduction by electroporation is less efficient and requires a selection step. Methods for positive selection of the recombination event (i.e., neo resistance) and dual positive-negative selection (i.e., neo resistance and gancyclovir resistance) and the subsequent identification of the desired clones by PCR have been described by Joyner et al., atu a 338, 153-156 (1989) and Capecchi, (1989).
Embr~ro RecoverLr and ES cell Injection Naturally cycling or superovulated female mice mated with males are used to harvest embryos for the implantation of ES cells. It is desirable to use the C57B strain for this purpose when using mice. Embryos of the appropriate age are recovered approximately 3.5 days after successful mating. Mated females are sacrificed by COZ asphyxiation or cervical dislocation and embryos are flushed from excised uterine horns and placed in Dulbecco's modified essential medium plus 10% calf serum for injection with ES cells.
Approximately 10-20 ES cells are injected into blastocysts using a glass microneedle with an internal diameter of approximately 20 um.
Transfer of Embryos to Pseudoprectnant Females Randomly cycling adult female mice are paired with vasectomized males. Mouse strains such as Swiss Webster, ICR or others can be used for this purpose.

_26_ Recipient females are mated such that they will be at 2.5 to 3.5 days post-mating when required for implantation with blastocysts containing ES cells. At the time of embryo transfer, the recipient females are anesthetized with an intraperitoneal injection of 0.015 ml of 2.5% avertin per gram of body weight. The ovaries are exposed by making an incision in the body wall directly over the oviduct and the ovary and uterus are externalized. A hole is made in the uterine horn with a 25 gauge needle through which the blastocysts are transferred. After the transfer, the ovary and uterus are pushed back into the body and the incision is closed by two sutures. This procedure is repeated on the opposite side if additional transfers are to be made.
Identification of Transqenic Mice and Rats.
Tail samples (1-2 cm) are removed from three week old animals. DNA is prepared and analyzed by both Southern blot and PCR to detect transgenic founder (Fo) animals and their progeny (F1 and Fzj.
Pathological Studies The various Fo, F" and FZ animals that carry the microinjected transgene are sacrificed by COZ
asphyxiation and analyzed by immunohistology for neuritic plaques and neurofibrillary tangles (NFTs) in the brain. Brains of mice and rats from each transgenic line are fixed in 4% paraformaldehyde and sectioned on a cryostat. Sections are stained with antibodies reactive with the APP and/or the f3 peptide.
Secondary antibodies conjugated with fluorescein, rhodamine, horse radish peroxidase, or alkaline phosphatase are used to detect the primary antibody.
These experiments permit identification of amyloid plaques and the regionalization of these plaques to specific areas of the brain. Plaques ranging in size from 9 to 50 um characteristically occur in the brains of AD patients in the cerebral cortex, but also may be observed in deeper grey matter including the amygdaloid nucleus, corpus striatum and diencephalon.
Sections are also stained with other antibodies diagnostic of Alzheimer's plaques, recognizing antigens such as Alz-50, tau, A2B5, neurofilaments, neuron-specific enolase, and others that are characteristic of Alzheimer's plaques (Wolozin, et al., Science 232, 648 (1986); Hardy and Allsop, Trends in Pharm. Sci. 12, 383-388 (1991); Selkoe, Ann. Rev.
Neurosci. 12, 463-490 (1989); Arai et al., Proc. Natl.
Acad. Sci. USA 87, 2249-2253 (1990); Majocha et al., Amer. Assoc. Neuropatholoay Abs; 99,22 (1988); Masters et al., Proc. Natl. Acad. Sci. 82,4245-4249; Majocha et al., Can J Biochem Cell Biol 63;577-584 (1985)).
Staining with thioflavins and Congo red is also carried out to analyze co-localization of B peptide deposits within neuritic plaques and NFTs.
Analysis of APP and f3 Peptide Expression:
mRNA: mRNA is isolated by the acid guanidinium thiocyanate-phenol: chloroform extraction method (Chomczynski and Sacchi, Anal Biochem 162,156-159 (1987)) from cell lines and tissues of transgenic animals to determine expression levels by Northern blots.
Protein: APP and f3 peptide are detected by using polyclonal and monoclonal antibodies that are specific to the extra-cytoplasmic domain, f3 peptide region, and C-terminus.
Western Blot Analysis: Protein fractions are isolated from tissue homogenates and cell lysates and subjected to Western blot analysis as described by Harlow et al., Antibodies: A laboratory manual, (Cold Spring Harbor, NY, 1988); Brown et al., J. Neurochem.
40;299-308 (1983); and Tate-Ostroff et al., Proc Natl Acad Sci 86;745-749 (1989)). Only a brief description is given below.

The protein fractions are denatured in Laemmli sample buffer and electrophoresed on SDS-Polyacrylamide gels. The proteins are be then transferred to nitrocellulose filters by electroblotting. The filters are blocked, incubated with primary antibodies, and finally reacted with enzyme conjugated secondary antibodies. Subsequent incubation with the appropriate chromogenic substrate reveals the position of APP proteins.
Pathological and Behavioral Studies Patholoaical Studies Immunohistology and thioflavin S staining are conducted as described elsewhere herein.
In situ Hybridizations: Radioactive or enzymatically labeled probes are used to detect mRNA
in situ. The probes are degraded approximately to 100 nucleotides in length for better penetration of cells.
The procedure of Chou et al. J. Psych. Res. 24,27-50 (1990) for fixed and paraffin embedded samples is briefly described below although similar procedures can be employed with samples sectioned as frozen material. Paraffin slides for in situ hybridization are dewaxed in xylene and rehydrated in a graded series of ethanols and finally rinsed in phosphate buffered saline (PBS). The sections are post-fixed in fresh 4% paraformaldehyde. The slides are washed with PBS twice for 5 minutes to remove paraformaldehyde.
Then the sections are permeabilized by treatment with a 20 ~Cg/ml proteinase K solution. The sections are re-fixed in 4% paraformaldehyde, and basic molecules that could give rise to background probe binding are acetylated in a 0.1 M triethanolamine, 0.3 M acetic anhydride solution for 10 minutes. The slides are washed in PBS, then dehydrated in a graded series of ethanols and air dried. Sections are hybridized with antisense probe, using sense probe as a control.
After appropriate washing, bound radioactive probes 21 2 7 4 5 0 _ -29-are detected by autoradiography or enzymatically labeled probes are detected through reaction with the appropriate chromogenic substrates.
Behavioral Studies Behavioral tests designed to assess learning and memory deficits are employed. An example of such as test is the Morris Water maze (Morris, Learn Motivat.
12;239-260 (1981)). In this procedure, the animal is placed in a circular pool filled with water, with an escape platform submerged just below the surface of the water. A visible marker is placed on the platform so that the animal can find it by navigating toward a proximal visual cue. Alternatively, a more complex form of the test in which there are no formal cues to mark the platform's location will be given to the animals. In this form, the animal must learn the platform's location relative to distal visual cues.
The procedures applied to test transgenic mice is similar for transgenic rats.
Screening' of Compounds for Treatment of Alzheimer's Disease The transgenic animals and animal cells are used to screen compounds for a potential effect in the treatment of Alzheimer's disease using standard methodology. The compound is administered to the animals or introduced into the culture media over a period of time and in various dosages, then the animals or animal cells examined for alterations in APP expression, histopathology, and/or behavior using the procedures described above.
Example 1: Expression of pMTAPP-1 in NIIi3T3 and PC12 Cells.
The clone, pMTAPP-1 is an example of the expression vector shown in Fig. la where the promoter used is the metallothionine promoter. Stable cell lines were derived by transfecting NIH3T3 and PC12 cell lines (ATCC #CCL92 and CRL1721). 0.5 x 106 of w'0 93/14240 PCT/US92/11276 NIH3T3 or PC12 cells were plated into 100 mm dishes and transfected with a mixture of 5 mg of the Sal1 fragment and 1 mg of pSV2neo DNA (46) precipitated in the presence of 50 mg lipofectinTM(Gibco, BRL) in a final volume of 100 ~1. Polylysine-coated plates were used for PC12 cells, which normally do not adhere well to tissue culture dishes. The cells were fed with selection medium containing l0% fetal bovine serum in DMEM or RPMI and supplemented with 6418. Five hundred mg/ml (biological weight) and 250 mg/ml of 6418 were used to select colonies form NIH3T3 and PC12 cells, respectively. Fifteen days after transfection, colonies of cells resistant to 6418 were isolated by cloning rings and expanded in T flasks. The presence 'of APP cDNA in the cells was detected by PCR using the procedure of Mullis and Faloona, Methods Enzymol.
155;335-350 (1987) Expression of APP in 25 colonies from each cell line was analyzed by immunostaining (Majocha et al., 1988). Cells were grown to subconfluence and fixed in a solution containing 4% paraformaldehyde, 0.12 M
NaCl, and 20 mm Na3P04, pH 7Ø They were incubated overnight with a primary monoclonal antibody against a synthetic t3 peptide sequence (Masters et al., 1985) provided by Dr. Ronald Majocha, Massachusetts General Hospital, Boston, MA, followed by a generalized anti-mouse antibody conjugated to biotin (Jackson ImmunoResearch Labs, PA). Immunostaining was then performed by adding avidin-horseradish peroxidase (HRP) (Vector Labs, Burlingame, CA) and diaminobenzidine as the chromogen (Majocha et al., 1985). The results indicated that the pMTAPP-1 vector was expressing APP in both NIH3T3 and PC12 cells.
Example 2: Expression of pEAPP-1 in PC12 Cells.
pEAPP-1 is an example of the 25 kb human APP gene promoter linked to and controlling expression of a human APP770 cDNA like the construct in Figure 1A.
DNA from this construct was transfected into PC12 cells as described above. Certain clones of pEAPP-1 transfected cells exhibited a differentiation phenotype morphologically similar to that exhibited by PC12 cells treated with nerve growth factor (NGF).
PC12 cells normally are fairly round and flat cells.
Those transfected with pEAPP-1 have cytoplasmic extensions resembling neurites. PC12 cells treated with NGF extend very long neuritic extensions.
Thirteen PC12 cell clones transfected with pEAPP-1 were selected and propagated. Eight of these cell clones exhibited the spontaneous differentiation phenotype with clones 1-8, 1-1, and 1-4 exhibiting the strongest phenotypes. Staining of pEAPP-1 transfected PC12 cells with antibody against the t3 peptide as described above indicated that those cells exhibiting the differentiation were also expressing APP. Because PC12 cells transfected with the pMTAPPl clone did not exhibit this phenotype even though the APP770 cDNA is expressed, these results suggest that expression of APP770 from the human promoter has novel properties regarding the physiology of the cell.
Example 3: Expression of pMTA4 in PC12 Cells.
pMTA4 is an example of the type of construct shown in Figure 4A where the promoter used is the metallothionine promoter. The protein encoded by this construct differs slightly from that depicted in Figure 4. An APP770 cDNA clone was digested with Asp718 which cleaves after position 56 (number system of Kang, et al., 1987). The resulting 5' extension was filled in using the Klenow enzyme (Maniatis). The same DNA preparation was also cleaved with EcoRI which also cuts after position 1795 and the resulting 5' extension was filled in using the Klenow enzyme (Maniatis). Self-ligation of this molecule results in an expression clone in which the truncated protein 4 ~~ -32-thus encoded contains the leader sequence, followed by a shortened version of the !3 peptide starting with the sequence phe-arg-val-gly-ser-of the 13 peptide followed by the 56 terminal amino acids of APP. DNA from this construct was transfected into PC12 cells as described above.
Example 4: Generation of Transgenic Mice expressing APP under the control of the MT-1 promoter.
Transgenic mice were made by microinjecting pMTAPPl vector DNA into pronuclear embryos. pMTAPPl is an example of the type of construct shown in Fig.
la which is operably linked to the metallothionine promoter. The procedures for microinjection into mouse embryos are described in "Manipulating the mouse embryo" by B. Hogan et al. (1986). Only a brief description of the procedures is described below.
Mice were obtained from Taconic Laboratories (German Town, New York). Swiss Webster female mice were used for embryo retrieval and implantation.
B6D2F, males were used for mating and vasectomized Swiss webster studs were used to simulate pseudopregnancy.
Embryo Recovery: Female mice, 4 to 8 weeks of age, were induced to superovulate with 5 IU of pregnant mare's serum gonadotropin (PMSG; Sigma) followed 48 hours later by 5 IU of human chorionic gonadotropin (hCG; Sigma). Females were placed with males immediately after hCG injection. Embryos were recovered from excised oviducts of mated females 21 hours after hCG in Dulbecco's phosphate buffered saline with 0.5o bovine serum albumin (BSA; Sigma).
Surrounding cumulus cells were removed with hyaluronidase (1 mg/ml). Pronuclear embryos were then washed and placed in Earle's balanced salt solution containing 0.4% BSA (EBSS) in a 37.5°C incubator with a humidified atmosphere at 7% COZ, 5% Oz, and 88% NZ
until the time of injection.
Microinj ection: Elutip-Due''' purified Sal1 DNA was dissolved in 5 mM Tris (pH 7.4) and 0.1 mM EDTA at 3 ug/ml concentration for microinjection. Microneedles and holding pipettes were pulled from Fisher coagulation tubes (Fisher) on a DKI model 720 pipette pulley. Holding pipettes were then broken at approximately 70 ~m (0.D.) and fire polished to an I.D. of about 30 ~.m on a Narishige microforge (model MF-83). Pipettes were mounted on NarishigeT""
micromanipulators which were attached to a Nikon ~DiaphotMmicroscope. The air-filled injection pipette was filled with DNA solution through the tip after breaking the tip against the holding pipette.
Embryos, in groups of 30 to 40, were placed in 100 ~cl drops of EBBS under paraffin oil for micromanipulation. An embryo was oriented and held with the holding pipette. The injection pipette was then inserted into the male pronucleus (usually the larger one). If the pipette did not break through the membrane immediately the stage was tapped to assist in penetration. The nucleus was then injected and the injection was monitored by swelling of the nucleus.
Following injection, the group of embryos was placed in EBSS until transfer to recipient females.
Transfer: Randomly cycling adult female mice were paired with vasectomized Swiss Webster males.
Recipient females were mated at the same time as donor females. At the time of transfer, the females were anesthetized with avertin. The oviducts were exposed by a single midline dorsal incision. An incision was then made through the body :all directly over the oviduct. The ovarian bursa was then torn with watch makers forceps. Embryos to be transferred were placed in DPBS and in the tip of a transfer pipet (about 10-12 embryos). The pipet tip was inserted into the ~J !.

infundibulum and embryos were transferred. After the transfer, the incision was closed by two sutures.
Analysis Of Mice For Transctene Intectration: At three weeks of age tail samples about 1 cm long were excised for DNA analysis. The tail samples were digested by incubating with shaking overnight at 55°C
in the presence of 0.7 ml 5 mM Tris, pH 8.0, 100 mM
EDTA, 0.5% SDS and 350 ~,g of proteinase K. The digested material was extracted once with an equal volume of phenol and once with an equal volume of phenol: chloroform (1:1 mixture). The supernatants were mixed with 70 ~1 3 M sodium acetate (pH 6.0) and the DNAs were precipitated by adding equal volume of 100% ethanol. The DNAs were spun down in a microfuge, washed once with 70% ethanol, dried and dissolved in 100 ,~1 TE buffer (10 mM tris pH 8.0 and 1 mM EDTA).
10-20 ~,1 of DNAs were restricted with BamHl, electrophoresed on 1% agarose gels, blotted onto nitrocellulose paper, and hybridized with 3zP-labeled APP cDNA fragment. Transgenic animals were identified by autoradiography of the hybridized nitrocellulose filters. The DNAs were also analyzed by PCR carried out by synthetic primers to generate an 800 by fragment.
A total of 671 pronuclear embryos were microinjected out of which 73 live and 6 dead pups were born. DNA analysis identified 9 transgenic mice (5 females and 4 males) which were bred to generate F, and F~ transgenics. These animals can be analyzed for expression of mRNA and protein of APP in different tissues and for analysis of behavioral and pathological abnormalities as described above.

Claims (19)

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. A mammalian cell containing a nucleic acid construct comprising a promoter operatively linked to a combination cDNA/genomic expression clone of amyloid precursor protein (APP) comprising:
(a) exon 6 and an amount of the adjacent downstream intron sufficient for splicing;
(b) KI and OX-2 coding regions and an amount of each of their upstream and downstream introns sufficient for splicing; and (c) exon 9 and an amount of the adjacent upstream intron sufficient for splicing wherein the construct is transcribed and alternatively spliced in the mammalian cell to form mRNA molecules which encode and are translated into APP695, APP751 and APP770.
2. The mammalian cell of claim 1 wherein the construct includes a promoter selected from the group consisting of human APP promoter, mouse APP promoter, rat APP promoter, metallothionine promoter, rat neuron specific enolase promoter, human .beta. actin gene promoter, human platelet derived growth factor B (PDGFB) chain gene promoter, rat sodium channel gene promoter, mouse myelin basic protein gene promoter, human copper-zinc superoxide dismutase gene promoter, and mammalian POU-domain regulatory gene promoter.
3. The mammalian cell of claim 1 wherein the construct is transcribed by the mammalian cell to yield mRNA encoding (a) APP770, APP695, and APP751, or (b) APP770, APP695, and APP751 having a mutation that predisposes to familial Alzheimer's disease.
4. The cell of claim 1 wherein a codon encoding valine at position 717 in the APP770 protein is mutated to encode an amino acid selected from the group consisting of ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, and gln.
5. The cell of claim 1 wherein the promoter is an APP promoter of the same species of origin as the cell.
6. A method for making a transgenic rodent model for Alzheimer's disease comprising:
(a) introducing inta a rodent cell or embryo a nucleic acid construct comprising a promoter operatively linked to a combination cDNA/genomic expression clone of amyloid precursor protein comprising:
(i) exon 6 and an amount of the adjacent downstream intron sufficient for splicing;
(ii) KI and OX-2 coding regions and an amount of each of their upstream and downstream introns sufficient for splicing; and (iii) exon 9 and an amount of the adjacent upstream intron sufficient for splicing;
wherein the construct is transcribed and alternatively spliced in the mammalian cell to form mRNA molecules which encode and are translated into APP695, APP751 and APP770; and (b) implanting the cell or embryo into a female rodent.
7. The method of claim 6 wherein the transgenic rodent has altered expression or processing of APP.
8. A method for screening compounds for an effect on Alzheimer's disease comprising exposing:
a transgenic rodent or mammalian cells transcribing a nucleic acid construct comprising a promoter operatively linked to a combination cDNA/genomic expression clone of amyloid precursor protein comprising:
(i) exon 6 and an amount of the adjacent downstream intron sufficient for splicing;

(ii) KI and OX-2 coding regions and an amount of each of their upstream and downstream intron sufficient for splicing; and (iii) exon 9 and an amount of the adjacent upstream intron sufficient for splicing, wherein the construct is transcribed and alternatively spliced in the mammalian cell to form mRNA molecules which encode and are translated into APP695, APP751 and APP770, to the compound to be tested and determining if there is altered expression of processing of amyloid precursor protein.
9. The method of claim 8 using a transgenic rodent, further comprising determining if the expression or processing of amyloid precursor protein, histopathology, or behavior of the transgenic rodent is altered after administration of the compound to the rodent.
10. The method of claim 8 wherein using a transgenic rodent.
11. A nucleic acid construct comprising a promoter operatively linked to a combination cDNA/genomic expression clone of amyloid precursor protein (APP) comprising:
(a) exon 6 and an amount of the adjacent downstream intron sufficient for splicing;
(b) KI and OX-2 coding regions and an amount of each of their upstream and downstream introns sufficient for splicing; and (c) exon 9 and an amount of the adjacent upstream intron sufficient for splicing;
wherein the construct is transcribed and alternatively spliced in a mammalian cell to form mRNA molecules which encode and are translated into APP695, APP751 and APP770, and further wherein the APP construct has the wild type amino acid sequence or a mutation that predisposes to familial Alzheimer's disease.
12. The construct of claim 11 wherein the construct includes a promoter selected from the group consisting of human APP promoter, mouse APP
promoter, rat APP promoter, metallothionine promoter, rat neuron specific enolase promoter, human .beta. actin gene promoter, human platelet derived growth factor B (PDGF-B) chain gene promoter, rat sodium channel gene promoter, mouse myelin basic protein gene promoter, human copper-zinc superoxide dismutase gene promoter, and mammalian POU-domain regulatory gene promoter.
13. The construct of claim 11, wherein the codon encoding the valine residue at position 717 in the wild type APP770 protein is mutated to encode an amino acid selected from the group consisting of ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, and gln.
14. The method of claim 6 wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode an amino acid selected from the group consisting of ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, and gln.
15. The method of claim 14, wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode phe.
16. The method of claim 8 wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode an amino acid selected from the group consisting of ile, phe, gly, tyr, leu, ala, pro, trp, met, ser, thr, asn, and gln.
17. The method of claim 16, wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode phe.
18. The cell of claim 4, wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode phe.
19. The construct of claim 13, wherein the codon encoding valine at position 717 in the APP770 protein is mutated to encode phe.
CA002127450A 1992-01-07 1992-12-29 Transgenic animal models for alzheimer's disease Expired - Lifetime CA2127450C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US81758492A 1992-01-07 1992-01-07
US817,584 1992-01-07
US91546992A 1992-07-16 1992-07-16
US915,469 1992-07-16
PCT/US1992/011276 WO1993014200A1 (en) 1992-01-07 1992-12-29 Transgenic animal models for alzheimer's disease

Publications (2)

Publication Number Publication Date
CA2127450A1 CA2127450A1 (en) 1993-07-22
CA2127450C true CA2127450C (en) 2007-04-17

Family

ID=27124190

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002127450A Expired - Lifetime CA2127450C (en) 1992-01-07 1992-12-29 Transgenic animal models for alzheimer's disease

Country Status (12)

Country Link
US (2) US5720936A (en)
EP (1) EP0620849B1 (en)
JP (3) JPH07506720A (en)
AT (1) ATE243746T1 (en)
AU (1) AU671093B2 (en)
CA (1) CA2127450C (en)
DE (1) DE69233109T2 (en)
DK (1) DK0620849T3 (en)
ES (1) ES2204899T3 (en)
IL (1) IL104304A (en)
PT (1) PT620849E (en)
WO (1) WO1993014200A1 (en)

Families Citing this family (184)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5586226A (en) * 1990-05-16 1996-12-17 Canon Kabushiki Kaisha Control method and device for a unicolor printer
US5912410A (en) * 1990-06-15 1999-06-15 Scios Inc. Transgenic non-human mice displaying the amyloid-forming pathology of alzheimer's disease
DE69233774D1 (en) * 1991-01-21 2009-12-10 Elan Pharm Inc Exam and model for Alzheimers disease
PT620849E (en) * 1992-01-07 2003-11-28 Elan Pharm Inc TRANSGENIC ANIMAL MODELS FOR ALZHEIMER'S DISEASE
US6610493B1 (en) 1993-06-17 2003-08-26 Brigham And Women's Hospital Screening compounds for the ability to alter the production of amyloid-β peptide
US6264915B1 (en) 1992-09-13 2001-07-24 The President And Fellows Of Harvard College Process for detecting potential carcinogens
US5436153A (en) * 1992-12-02 1995-07-25 Sprecher; Cindy A. Human amyloid protein precursor homolog and Kunitz-type inhibitor
WO1994012637A2 (en) * 1992-12-02 1994-06-09 Zymogenetics, Inc. Novel human amyloid protein precursor homologue and kunitz-type inhibitors
ES2155099T3 (en) 1993-10-27 2001-05-01 Elan Pharm Inc TRANSGENIC ANIMALS THAT HOST APP ALELOS THAT PRESENT A SWEDISH MUTATION.
JPH07132033A (en) * 1993-11-12 1995-05-23 Hoechst Japan Ltd Transgenic animal for alzheimer's disease model
US5877399A (en) * 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
CA2182311A1 (en) 1994-01-27 1995-08-03 Karen Hsiao Transgenic non-human mammals with progressive neurologic disease
FR2716460B1 (en) * 1994-02-21 2002-08-09 Rhone Poulenc Rorer Sa Animal model of Alzheimer's disease, preparation and uses.
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
WO1995034640A1 (en) * 1994-06-13 1995-12-21 Henry Ford Health System A novel neuronal-neonatal gene: neuronatin
CA2198451A1 (en) * 1994-09-01 1996-03-07 Gurparkash Singh Transgenic animal expressing a familial form of human amyloid precursor protein
US7427392B1 (en) 1994-11-14 2008-09-23 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of alzheimer's disease by measuring amyloid-β peptide (x-≧41) and tau
US6114133A (en) * 1994-11-14 2000-09-05 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of Alzheimer's disease by measuring amyloid-β peptide (x-≧41)
JP2000504202A (en) * 1994-12-05 2000-04-11 メルク エンド カンパニー インコーポレーテッド Transgenic animals lacking native amyloid precursor protein
US6187992B1 (en) 1994-12-05 2001-02-13 Merck & Co., Inc. Transgenic mouse having a disrupted amyloid precursor protein gene
US5777194A (en) * 1995-04-26 1998-07-07 Cephalon, Inc. Gene-targeted mice with humanized Aβ sequence and Swedish FAD mutation
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
US5858661A (en) 1995-05-16 1999-01-12 Ramot-University Authority For Applied Research And Industrial Development Ataxia-telangiectasia gene and its genomic organization
US6187991B1 (en) 1995-05-23 2001-02-13 Pfizer Inc Transgenic animal models for type II diabetes mellitus
CA2221986A1 (en) * 1995-06-07 1996-12-19 Athena Neurosciences, Inc. Method for identifying alzheimer's disease therapeutics using transgenic animal models
AU6168396A (en) * 1995-06-07 1996-12-30 Athena Neurosciences, Inc. Method for identifying alzheimer's disease therapeutics usin g transgenic animal models
US6717031B2 (en) 1995-06-07 2004-04-06 Kate Dora Games Method for selecting a transgenic mouse model of alzheimer's disease
US5894078A (en) * 1996-02-26 1999-04-13 Advanced Bioconcept, Inc. Transgenic mouse expressing C-100 app
US6140309A (en) * 1996-03-12 2000-10-31 University Of South Florida Vasoactive effects and free radical generation by β-amyloid peptides
WO1997046664A1 (en) * 1996-06-06 1997-12-11 University Of Washington Perlecan transgenic animals and methods of identifying compounds for the treatment of amyloidoses
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
WO1998003644A1 (en) * 1996-07-24 1998-01-29 Novartis Ag Transgenic animal model for alzheimer disease
US20010016951A1 (en) 1996-07-24 2001-08-23 Bernd Sommer Transgenic animal model for alzheimer disease
WO1998003643A2 (en) * 1996-07-22 1998-01-29 Smithkline Beecham Pharma Gmbh Transgenic animals with mutant human app or a4ct sequences
JP2000517185A (en) 1996-08-29 2000-12-26 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Novel metalloprotease family KUZ
US6566089B1 (en) * 1996-09-04 2003-05-20 Tularik Inc. Cell-based drug screens for regulators of gene expression
WO1998015172A2 (en) * 1996-10-10 1998-04-16 Institut Pasteur Transgenic or mutated animal as model for a neuron deficit
US5898094A (en) * 1996-10-21 1999-04-27 University Of South Florida Transgenic mice expressing APPK670N,M671L and a mutant presenilin transgenes
US7973156B2 (en) 1997-08-21 2011-07-05 Quark Pharmaceuticals Inc. Hypoxia-regulated genes
US6710226B1 (en) 1997-12-02 2004-03-23 Neuralab Limited Transgenic mouse assay to determine the effect of Aβ antibodies and Aβ Fragments on alzheimer's disease characteristics
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US6913745B1 (en) 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US6743427B1 (en) 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
DK1045918T3 (en) 1998-01-12 2008-12-08 Pedro Jose G protein-related kinase mutants in essential hypertension
CA2321934C (en) * 1998-02-25 2012-02-21 Paul Q. Anziano Manganese superoxide dismutase exon 3-deleted isoforms and nucleic acid molecules encoding the isoforms
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US6548261B1 (en) 1998-12-30 2003-04-15 Case Western Reserve University Alzheimer model for drug screening
EP1026251A3 (en) * 1999-02-03 2000-09-06 Pfizer Products Inc. Transgenic animals expressing human P25
US6374130B1 (en) 1999-04-06 2002-04-16 Eric M. Reiman Methods for tracking the progression of Alzheimer's disease identifying treatment using transgenic mice
US6670195B1 (en) 1999-05-26 2003-12-30 New York University Mutant genes in Familial British Dementia and Familial Danish Dementia
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
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
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
US6541244B1 (en) * 1999-06-07 2003-04-01 Cedars-Sinai Medical Center Suppressor of cytokine signaling (SOCS)-3 promoter and methods for its use in genetic therapy in humans
US7569660B1 (en) 1999-06-09 2009-08-04 The University Of Chicago Recombinant prion-like proteins and materials comprising same
US6875582B1 (en) 1999-08-19 2005-04-05 Omniscience Pharmaceuticals, Inc. Methods and targets of antibiotic resistance
US6864355B1 (en) 2000-05-02 2005-03-08 Yale University Inhibition of NF-κB activation by blockade of IKKβ-NEMO interactions at the NEMO binding domain
AU2288601A (en) 1999-12-23 2001-07-03 Millennium Pharmaceuticals, Inc. P2Y12 receptor
NZ520065A (en) 2000-01-12 2006-02-24 Univ Yale Nogo receptor-mediated blockade of axonal growth
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
US6982361B1 (en) * 2000-02-25 2006-01-03 The Regents Of The University Of California Method of screening a compound for anxiolytic activity in an Apolipoprotein e knockout animal
DE60124080T2 (en) 2000-03-23 2007-03-01 Elan Pharmaceuticals, Inc., San Francisco COMPOUNDS AND METHOD FOR THE TREATMENT OF ALZHEIMER'S DISEASE
US6992081B2 (en) 2000-03-23 2006-01-31 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
EP1274720A4 (en) 2000-04-12 2004-08-18 Human Genome Sciences Inc Albumin fusion proteins
US20030096864A1 (en) * 2000-06-30 2003-05-22 Fang Lawrence Y. Compounds to treat alzheimer's disease
PE20020276A1 (en) 2000-06-30 2002-04-06 Elan Pharm Inc SUBSTITUTE AMINE COMPOUNDS AS ß-SECRETASE INHIBITORS FOR THE TREATMENT OF ALZHEIMER
JP2004502664A (en) * 2000-06-30 2004-01-29 イーラン ファーマスーティカルズ、インコーポレイテッド Compound for treating Alzheimer's disease
US6846813B2 (en) * 2000-06-30 2005-01-25 Pharmacia & Upjohn Company Compounds to treat alzheimer's disease
EP1666452A2 (en) 2000-06-30 2006-06-07 Elan Pharmaceuticals, Inc. Compounds to treat Alzheimer's disease
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
US20030087286A1 (en) * 2000-09-06 2003-05-08 Hodge Timothy A. Isolation of eukaryotic genomic DNA using magnetically responsive solid functionalized particles
US7494817B2 (en) * 2000-09-06 2009-02-24 Transnet Yx, Inc. Methods for genotype screening using magnetic particles
US20050272085A1 (en) * 2000-09-06 2005-12-08 Hodge Timothy A Methods for forensic and congenic screening
EP1978110B1 (en) * 2000-09-06 2010-05-26 Transnetyx, Inc. Computer-based method and system for screening genomic DNA
US20050266494A1 (en) * 2000-09-06 2005-12-01 Hodge Timothy A System and method for computer network ordering of biological testing
US20030082605A1 (en) * 2000-09-06 2003-05-01 Hodge Timothy A. Genomic DNA detection method and system thereof
US20050239125A1 (en) * 2000-09-06 2005-10-27 Hodge Timothy A Methods for genotype screening
US6900367B2 (en) * 2000-09-29 2005-05-31 Novartis Transgenic Drosophila melanogaster expressing a β42 in the eye
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US20020170078A1 (en) * 2001-03-08 2002-11-14 Isabelle Mansuy Calcineurin-related transgenic mammals, compositions and methods
US7081561B2 (en) 2001-03-16 2006-07-25 The Regents Of The University Of California Gene-targeted animal model of apolipoprotein E4 domain interaction and uses thereof
ATE396174T1 (en) * 2001-06-01 2008-06-15 Elan Pharm Inc HYDROXYALKYLAMINE DERIVATIVES AS BETA-SECRETASE INHIBITORS AND THE USE THEREOF FOR THE TREATMENT OF ALZHEIMER'S DISEASE OR SIMILAR DISEASES
MXPA03011521A (en) 2001-06-13 2004-03-18 Elan Pharm Inc Aminediols as agents for the treatment of alzheimer's disease.
US6982264B2 (en) * 2001-06-27 2006-01-03 Elan Pharmaceuticals, Inc. Substituted alcohols useful in treatment of Alzheimer's disease
US20070213407A1 (en) * 2001-06-29 2007-09-13 Elan Pharmaceuticals And Pharmacia & Upjohn Company Llc Compounds to treat Alzheimer's disease
CA2453451A1 (en) * 2001-07-10 2003-01-23 Elan Pharmaceuticals, Inc. Aminediols for the treatment of alzheimer's disease
US7067542B2 (en) * 2001-07-10 2006-06-27 Pharmacia & Upjohn Company Diaminediols for the treatment of Alzheimer's disease
US7297553B2 (en) * 2002-05-28 2007-11-20 Nanosphere, Inc. Method for attachment of silylated molecules to glass surfaces
CA2455497A1 (en) * 2001-08-31 2003-03-06 Neurochem (International) Limited Amidine derivatives for treating amyloidosis
US20060014186A1 (en) * 2001-09-04 2006-01-19 Hodge Timothy A Methods for genotype screening of a strain disposed on an adsorbent carrier
BR0213139A (en) 2001-10-04 2004-08-10 Elan Pharm Inc Compound, method of treating or preventing disease and preparing a compound, use of a compound and pharmaceutical composition
NZ533107A (en) * 2001-11-08 2007-04-27 Upjohn Co N, N'-substituted-1,3-diamino-2-hydroxypropane derivatives
WO2003043618A2 (en) * 2001-11-19 2003-05-30 Elan Pharmaceuticals, Inc. Amino diols useful in the treatment of alzheimer's disease
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
EP2277888A3 (en) 2001-12-21 2011-04-27 Human Genome Sciences, Inc. Fusion proteins of albumin and erythropoietin
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
AU2003216370A1 (en) * 2002-02-27 2003-09-09 Merck And Co., Inc. Assays to monitor amyloid precursor protein processing
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
WO2004058686A1 (en) * 2002-04-30 2004-07-15 Elan Pharmaceuticals, Inc. Hydroxypropyl amides for the treatment of alzheimer’s disease
UY27967A1 (en) * 2002-09-10 2004-05-31 Pfizer 2-HINDROXI-1,3-DIAMINOALCANE OIL
EP1558737A4 (en) 2002-10-18 2008-06-11 Lg Life Sciences Ltd Gene families associated with cancers
AU2003293155A1 (en) * 2002-11-27 2004-06-23 Elan Pharmaceutical, Inc. Substituted ureas and carbamates
US7341847B2 (en) * 2003-04-02 2008-03-11 Agency For Science, Technology And Research Promoter construct for gene expression in neuronal cells
CA2522805A1 (en) * 2003-04-21 2004-11-04 Elan Pharmaceuticals, Inc. Phenacyl 2-hydroxy-3-diaminoalkanes
EP1615892A2 (en) 2003-04-21 2006-01-18 Elan Pharmaceuticals, Inc. Benzamide 2-hydroxy-3-diaminoalkanes
IL155666A (en) * 2003-04-29 2013-12-31 Neurim Pharma 1991 Composition for treating insomnia
BRPI0410348A (en) 2003-05-14 2006-05-30 Torreypines Therapeutics Inc compounds and uses thereof in amyloid-beta modulation
US7358331B2 (en) 2003-05-19 2008-04-15 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in Lewy body disease
CA2526900C (en) 2003-05-19 2015-11-24 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in lewy body disease
TWI306458B (en) 2003-05-30 2009-02-21 Elan Pharma Int Ltd Humanized antibodies that recognize beta amyloid peptide
US20060014790A1 (en) * 2004-01-21 2006-01-19 Varghese John Methods of treatment of amyloidosis using spirocyclohexane aspartyl-protease inhibitors
US7262223B2 (en) * 2004-01-23 2007-08-28 Neurochem (International) Limited Amidine derivatives for treating amyloidosis
WO2005087215A1 (en) 2004-03-09 2005-09-22 Elan Pharmaceuticals, Inc. Substituted urea and carbamate, phenacyl-2-hydroxy-3-diaminoalkane, and benzamide-2-hydroxy-3-diaminoalkane aspartyl-protease inhibitors
CA2556826A1 (en) * 2004-03-09 2005-09-22 Elan Pharmaceuticals, Inc. Methods of treatment of amyloidosis using bi-cyclic aspartyl protease inhibitors
WO2005087751A2 (en) * 2004-03-09 2005-09-22 Elan Pharmaceuticals, Inc. Substituted hydroxyethylamine aspartyl protease inhibitors
JP2007528402A (en) * 2004-03-09 2007-10-11 エラン ファーマシューティカルズ,インコーポレイテッド Substituted hydroxyethylamine aspartic protease inhibitors
SE0400707D0 (en) 2004-03-22 2004-03-22 Bioarctic Neuroscience Ab Transgenic animal model
KR100574544B1 (en) * 2004-04-01 2006-04-27 주식회사 뉴로테크 Transgenic mice inducing Alzheimer's disease expressing mutant ?CTF99
US7544855B2 (en) * 2004-04-23 2009-06-09 Buck Institute Transgenic mouse whose genome comprises an APP having a mutation at amino acid 664
JP2007538024A (en) * 2004-05-19 2007-12-27 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Methods of treating diseases and conditions associated with altered levels of amyloid beta peptide and novel enol carboxamide compounds
JP4476050B2 (en) * 2004-06-30 2010-06-09 株式会社ニデック Perimeter
JP2008505929A (en) * 2004-07-09 2008-02-28 エラン ファーマシューティカルズ,インコーポレイテッド Oxime derivative hydroxyethylamine aspartic protease inhibitor
US7385085B2 (en) * 2004-07-09 2008-06-10 Elan Pharmaceuticals, Inc. Oxime derivative substituted hydroxyethylamine aspartyl protease inhibitors
US8383637B2 (en) * 2004-08-06 2013-02-26 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8436006B2 (en) * 2004-08-06 2013-05-07 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
US8426429B2 (en) * 2004-08-06 2013-04-23 Jansssen Pharmaceutica N.V. 2-amino-quinazoline derivatives useful as inhibitors of β-secretase (BACE)
GB0419124D0 (en) * 2004-08-27 2004-09-29 Proteome Sciences Plc Methods and compositions relating to Alzheimer's disease
CA2577392A1 (en) * 2004-08-27 2006-03-09 Elan Pharmaceuticals, Inc. Methods of treatment of amyloidosis using ethanol cyclicamine derivatives aspartyl protease inhibitors
US7833513B2 (en) 2004-12-03 2010-11-16 Rhode Island Hospital Treatment of Alzheimer's Disease
PE20061329A1 (en) 2004-12-15 2006-12-08 Neuralab Ltd HUMANIZED AB ANTIBODIES TO IMPROVE COGNITION
EP2332408B1 (en) 2005-02-17 2013-11-13 Biogen Idec MA Inc. Treating neurological disorders
EP1746092A1 (en) 2005-07-22 2007-01-24 Exonhit Therapeutics SA Compounds and methods for treatment of amyloid-B-peptide related disorders
US20080293680A1 (en) * 2005-08-03 2008-11-27 Stefan Peters Substituted Ethane-1,2-Diamines for the Treatment of Alzheimer's Disease II
CA2618019A1 (en) * 2005-08-11 2007-02-15 Boehringer Ingelheim International Gmbh Compounds for the treatment of alzheimer's disease
EP1915353A2 (en) * 2005-08-11 2008-04-30 Boehringer Ingelheim International GmbH Isophthalic acid diamides for treating alzheimer disease
US20100144681A1 (en) * 2005-08-11 2010-06-10 Klaus Fuchs Compounds for the treatment of alzheimer's disease
CA2618013A1 (en) * 2005-08-11 2007-02-15 Boehringer Ingelheim International Gmbh Beta-secretase inhibitors for use in the treatment of alzheimer's disease
EP1937638A1 (en) * 2005-10-12 2008-07-02 Elan Pharmaceuticals Inc. Methods of treating amyloidosis using aryl-cyclopropyl derivative aspartyl protease inhibitors
WO2007047305A1 (en) * 2005-10-12 2007-04-26 Elan Pharmaceuticals, Inc. Methods of treating amyloidosis using cyclopropyl derivative aspartyl protease inhibitors
WO2007088418A1 (en) 2006-01-31 2007-08-09 Multi Gene Vascular Systems, Inc. Drug-eluting intravascular prostheses and methods of use
US7776882B2 (en) * 2006-02-06 2010-08-17 Baxter Ellen W 2-amino-3,4-dihydro-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US7932261B2 (en) * 2006-02-06 2011-04-26 Janssen Pharmaceutica Nv Macrocycle derivatives useful as inhibitors of β-secretase (BACE)
US7868022B2 (en) * 2006-02-06 2011-01-11 Janssen Pharmaceutica Nv 2-amino-quinoline derivatives useful as inhibitors of β-secretase (BACE)
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2008019368A2 (en) * 2006-08-07 2008-02-14 Teva Biopharmaceuticals Usa, Inc. Albumin-insulin fusion proteins
US9127084B2 (en) 2006-09-14 2015-09-08 Medgenics Medical Israel Ltd. Long lasting drug formulations
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
EP2862581B1 (en) 2006-10-12 2021-08-11 BHI Limited Partnership Methods, compounds, compositions and vehicles for delivering 3-amino-1-propanesulfonic acid
PL381605A1 (en) 2007-01-25 2008-08-04 Instytut Biologii Doświadczalnej Im. M. Nenckiego Polskiej Akadmii Nauk Transgenic animal and the manner of obtaining it
EP2134863B1 (en) 2007-03-13 2014-05-21 The Children's Hospital Of Philadelphia Genetic alterations on chromosome 16 and methods of use thereof for the diagnosis of type 1 diabetes
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
EP2191001B1 (en) 2007-04-09 2016-06-08 University of Florida Research Foundation, Inc. Raav vector compositions having tyrosine-modified capsid proteins and methods for use
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
WO2009007300A2 (en) 2007-07-06 2009-01-15 Boehringer Ingelheim International Gmbh Substituted amino-quinazolinones, medicaments comprising said compound, their use and their method of manufacture
ES2498040T3 (en) 2007-07-27 2014-09-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases with humanized anti-beta antibodies
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
BRPI0906625A2 (en) * 2008-01-28 2015-07-14 Janssen Pharmaceutica Nv Thio-2-amino-quinoline-6-substituted derivatives useful as beta-secretase (bace) inhibitors
NZ586830A (en) * 2008-01-29 2012-02-24 Janssen Pharmaceutica Nv 2-amino-quinoline derivatives useful as inhibitors of beta-secretase (bace)
CA2714713C (en) 2008-02-19 2018-02-27 The Children's Hospital Of Philadelphia Identification of pediatric onset inflammatory bowel disease loci and methods of use thereof for the diagnosis and treatment of the same
EP2116260A1 (en) 2008-04-16 2009-11-11 Freie Universität Berlin Screening method for agents suitable for prophylaxis and therapy of Alzheimer's disease (AD)
WO2009140679A2 (en) 2008-05-16 2009-11-19 The Children's Hospital Of Philadelphia Genetic alterations on chromosomes 21q, 6q and 15q and methods of use thereof for the diagnosis and treatment of type i diabetes
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US20110023152A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of cognition related genes in animals
US8871460B2 (en) 2009-11-09 2014-10-28 Neurogenetic Pharmaceuticals, Inc. Gamma-secretase modulatory compounds, methods for identifying same, and uses therefor
KR20200047642A (en) 2010-04-23 2020-05-07 유니버시티 오브 플로리다 리서치 파운데이션, 인크. Raav-guanylate cyclase compositions and methods for treating leber's congenital amaurosis-1 (lca1)
MX353096B (en) 2010-08-19 2017-12-19 Buck Institute For Age Res Star Methods of treating miild cognitive impairment (mci) and related discorders.
AU2013235422B2 (en) 2012-03-19 2016-12-15 Buck Institute For Research On Aging APP specific BACE inhibitors (ASBIs) and uses thereof
JP6471100B2 (en) 2013-02-12 2019-02-13 バック・インスティテュート・フォー・リサーチ・オン・エイジング Hydantoins that regulate BACE-mediated APP processing
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
WO2016077078A1 (en) 2014-10-29 2016-05-19 The Children's Hospital Of Philadelphia Diagnosis and treatment of genetic alterations associated with eosinophilic esophagitis
US10683552B2 (en) 2014-11-25 2020-06-16 Presidents And Fellows Of Harvard College Clonal haematopoiesis
EP3626832A3 (en) 2014-11-25 2020-05-13 The Brigham and Women's Hospital, Inc. Method of identifying and treating a person having a predisposition to or afflicted with a cardiometabolic disease
WO2017035017A1 (en) 2015-08-21 2017-03-02 The Children's Hospital Of Philadelphia Methods of treating autoimmune conditions in patients with genetic variations in dcr3 or in a dcr3 network gene
AU2016311502B2 (en) 2015-08-27 2019-07-18 Nantneuro, Llc Compositions for app-selective bace inhibition and uses therfor
CN109563089B (en) 2016-05-12 2023-06-16 巴克老年研究所 Compounds that promote normal processing of APP
EP4286405A1 (en) * 2021-01-28 2023-12-06 Abrain Gene therapy for treating neurodegenerative diseases

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3636991A1 (en) * 1986-03-03 1987-09-24 Transgene Gmbh METHOD FOR TRANSMITTING ORGANIC AND / OR INORGANIC SUBSTANCES ON EGGS AND / OR SOMA CELLS OF ANIMALS AND CORRESPONDING COMPOSITIONS
AU7879987A (en) * 1986-08-28 1988-03-24 Immunex Corp. Expression of heterologous proteins by transgenic lactating mammals
US4954437A (en) * 1986-09-15 1990-09-04 Integrated Genetics, Inc. Cell encoding recombinant human erythropoietin
US5220013A (en) * 1986-11-17 1993-06-15 Scios Nova Inc. DNA sequence useful for the detection of Alzheimer's disease
EP0332640A1 (en) * 1986-11-17 1989-09-20 California Biotechnology, Inc. Recombinant alzheimer's amyloid protein
US4994384A (en) * 1986-12-31 1991-02-19 W. R. Grace & Co.-Conn. Multiplying bovine embryos
EP0832981A1 (en) * 1987-02-17 1998-04-01 Pharming B.V. DNA sequences to target proteins to the mammary gland for efficient secretion
AU3056289A (en) * 1988-01-13 1989-08-11 Mclean Hospital Corporation, The Genetic constructs containing the alzheimer brain amyloid gene
AU6275190A (en) * 1989-09-14 1991-04-18 Norina Bautechnik Gmbh Floor with electrical resistance heating
US5221607A (en) * 1989-09-18 1993-06-22 Scios Nova Inc. Assays and reagents for amyloid deposition
EP0451700A1 (en) * 1990-04-10 1991-10-16 Miles Inc. Recombinant APP minigenes for expression in transgenic mice as models for Alzheimers's disease
ATE260974T1 (en) * 1990-06-15 2004-03-15 Scios Inc TRANSGENIC NON-HUMAN MAMMAL EXHIBITING THE AMYLOID-FORMING PATHOLOGY OF ALZHEIMER'S DISEASE
EP0550675A1 (en) * 1990-09-28 1993-07-14 Cephalon, Inc. Transgenic animals with alzheimer's amyloid precursor gene
DE69233774D1 (en) * 1991-01-21 2009-12-10 Elan Pharm Inc Exam and model for Alzheimers disease
US5672805A (en) * 1991-07-18 1997-09-30 The Regents Of The University Of California Transgenic mice expressing the neurotoxic C-terminus of β-amyloid precursor protein
AU2765992A (en) * 1991-10-03 1993-05-03 Indiana University Foundation Method for screening for alzheimer's disease
PT620849E (en) * 1992-01-07 2003-11-28 Elan Pharm Inc TRANSGENIC ANIMAL MODELS FOR ALZHEIMER'S DISEASE
EP0561087B1 (en) * 1992-03-20 1999-08-04 N.V. Innogenetics S.A. Mutated form of the beta-amyloid precursor protein gene
US5604102A (en) * 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
EP0667959B1 (en) * 1992-10-26 2003-08-13 Elan Pharmaceuticals, Inc. Methods for identifying inhibitors of the production of beta-amyloid peptide
WO1994023049A2 (en) * 1993-04-02 1994-10-13 The Johns Hopkins University The introduction and expression of large genomic sequences in transgenic animals
ES2155099T3 (en) * 1993-10-27 2001-05-01 Elan Pharm Inc TRANSGENIC ANIMALS THAT HOST APP ALELOS THAT PRESENT A SWEDISH MUTATION.
WO1995014769A1 (en) * 1993-11-29 1995-06-01 University Of South Florida TRANSGENIC MOUSE PREPARED USING YEAST ARTIFICIAL CHROMOSOMES (YACs) AND HOMOLOGOUS RECOMBINATION

Also Published As

Publication number Publication date
JP2004089187A (en) 2004-03-25
JP2007267742A (en) 2007-10-18
EP0620849B1 (en) 2003-06-25
DE69233109D1 (en) 2003-07-31
PT620849E (en) 2003-11-28
US5811633A (en) 1998-09-22
AU671093B2 (en) 1996-08-15
JPH07506720A (en) 1995-07-27
CA2127450A1 (en) 1993-07-22
US5720936A (en) 1998-02-24
EP0620849A1 (en) 1994-10-26
ES2204899T3 (en) 2004-05-01
AU3336093A (en) 1993-08-03
WO1993014200A1 (en) 1993-07-22
DK0620849T3 (en) 2003-10-20
IL104304A (en) 2004-09-27
DE69233109T2 (en) 2004-05-19
IL104304A0 (en) 1993-05-13
ATE243746T1 (en) 2003-07-15

Similar Documents

Publication Publication Date Title
CA2127450C (en) Transgenic animal models for alzheimer's disease
US7186881B2 (en) Testing compounds for effects on synaptophysin in transgenic mice expressing an Alzheimer's disease FAD DNA sequence
EP1005542B1 (en) APOLIPOPROTEIN E TRANSGENIC mice AND ASSAY METHODS
US6187992B1 (en) Transgenic mouse having a disrupted amyloid precursor protein gene
JPH07132033A (en) Transgenic animal for alzheimer's disease model
EP0904349A1 (en) Transgenic non-human mammals with progressive neurologic disease
WO1997048792A9 (en) Transgenic non-human mammals with progressive neurologic disease
JP2001517065A (en) Methods for identifying therapeutic agents for Alzheimer's disease using transgenic animal models
US5604131A (en) cDNA-genomic DNA hybrid sequence encoding APP770 containing a genomic DNA insert of the KI and OX-2 regions
EP0981602A1 (en) Transgenic animal expressing non-native wild-type and familial alzheimer's disease mutant presenilin 1 protein on native presenilin 1 null background
WO1996040896A1 (en) Method for identifying alzheimer's disease therapeutics using transgenic animal models
EP0920495B1 (en) Transgenic animal model for alzheimer disease
US20070022483A1 (en) Transgenic animal model for Alzheimer's disease
Wadsworth et al. Transgenic mouse expressing APP 770
WO1994024266A1 (en) Transgenic animal models for alzheimer's disease

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20121231