WO1991004339A1 - Assays and reagents for amyloid deposition - Google Patents

Assays and reagents for amyloid deposition Download PDF

Info

Publication number
WO1991004339A1
WO1991004339A1 PCT/US1990/005155 US9005155W WO9104339A1 WO 1991004339 A1 WO1991004339 A1 WO 1991004339A1 US 9005155 W US9005155 W US 9005155W WO 9104339 A1 WO9104339 A1 WO 9104339A1
Authority
WO
WIPO (PCT)
Prior art keywords
amyloid
preamyloid
val
protein
beta
Prior art date
Application number
PCT/US1990/005155
Other languages
French (fr)
Inventor
Barbara Cordell
David Wolf
Original Assignee
California Biotechnology Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by California Biotechnology Inc. filed Critical California Biotechnology Inc.
Priority to CA002065404A priority Critical patent/CA2065404C/en
Publication of WO1991004339A1 publication Critical patent/WO1991004339A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • the present invention relates to assays and re ⁇ agents useful for the chemical intervention of amyloidosis in Alzheimer's disease.
  • AD Alzheimer's disease
  • NFT neurofibrillary tangles
  • the major protein component of amyloid is an ⁇ 4 kilodalton (kd) protein, designated the beta-protein or A4 protein due to a partial beta pleated structure or its molecular weight, respectively.
  • the A4 protein accumulates extracellularly, both in brain parenchyma and in the walls of blood vessels, generally as amyloid plaques which form aggregate fibril structures and are insoluble on SDS-polyacrylamide gels.
  • the fibrils are generally identified as amyloid based on their green birefringence after staining with Congo red and their 40- to 90-A diameter.
  • the second protein mentioned previously, ac ⁇ cumulates intracellularly in neurons of Alzheimer's brains (Castano and Frangione, (1988) Lab Invest 58:122-132) and forms tangles composed of structures resembling paired helical filaments (PHFs).
  • PHFs paired helical filaments
  • the primary structure and number of proteins comprising PHFs are unknown.
  • PHF-containing neurites are found in the periphery of the plaque, whereas deposits of beta-amyloid protein form the central core of mature plaques, surrounded by degenerated neurites and glial cells.
  • the present invention provides a method for determining the ability of a potential therapeutic agent to intervene in the amyloid deposition process associated with Alzheimer's disease in a cellular environment, which method utilizes a recombinantly produced amyloid substrate in a screening assay.
  • the present invention also allows for the development and use of immunological reagents to detect the formation of preamyloid protein aggregation in the cell lines provided by the invention.
  • a method of screening agents capable of intervention in Alzheimer's disease amyloidosis comprises: a) culturing a cell line capable of expressing a gene encoding beta-amyloid protein under conditions suit- able to produce the beta-amyloid protein as an insoluble, preamyloid aggregate; b) combining a known quantity of the agent to be tested to the cell culture; and c) monitoring the combination to determine whether preamyloid aggregate formation is reduced.
  • preamyloid formation can be induced through infection of a cell line with a recombinant virus capable of expressing the beta-amyloid protein as an insoluble preamyloid aggregate.
  • recombinant viruses carry expression vectors comprising DNA encoding the beta-amyloid protein.
  • Immunoassay kits employing the reagents useful to screen potential amyloid intervening agents are also provided by the present invention.
  • FIG. 1 is a schematic illustration of two amyloid expression constructs employing the vaccinia pUVl insertion vector.
  • FIG 2. illustrates the results of immunoprecipitation of 35S-methionine labeled W:A99 infected CV-1 cell lysates using APCP antibodies.
  • FIG 3. are fluorescent photomicrographs of infected CV-1 cells stained with APCP antibodies.
  • FIG. 3A is a Mock control;
  • FIG. 3B is a W:CONT control;
  • FIG. 3C is the W:99 construct; and
  • FIG. 3D is the W:42 construct.
  • the magnification is 200x with a 0.4 second exposure time for each photo.
  • FIG. 4 is a illustration of the modified beta- actin expression selection vector, pAX-neo, that was employed to express the beta-amyloid core constructs in mammalian cells. Detailed Description of the Preferred Embodiments
  • the invention involves a method of screening agents capable of intervention in Alzheimer's disease amyloidosis.
  • beta-amyloid core protein or "A4 protein” refers to an approximately 4 kd protein first identified by Glenner and Wong, (1984) Biochem Biophvs Res Comm 120:885, which is defined at the amino terminus by sequence analysis as a mixture of four peptides with slightly different amino termini, the amino termini of the three smaller peptides being completely encoded by that of the largest.
  • beta-amyloid precursor protein refers to either the amyloid precursor protein of 695 amino acids (Kang et al., (1987) supra) or the 751 amino acid protein (Ponte et al., (1988) supra) containing within their sequence, the beta-amyloid core protein sequence defined above.
  • the A4 core protein begins at amino acid 597 of the 695 amino acid protein and at amino acid 653 of the 751 amino acid sequence.
  • preamyloid aggregation refers to a morphological description — first discovered by Tagliavini et al., (1988) Neurosci Lett 93:191-196 — of spherical, granular deposits which are considerably smaller than pre-plaques and plaques found at a high frequency in the brains of Alzheimer's victims. These deposits can be occasionally detected with silver stain but not with Congo red, a stain to which amyloid proteins demonstrate high binding affinity.
  • insertion vector includes plasmids, cosmids or phages capable of mediating homologous recombination into a viral genome such that the DNA encoding the beta-amyloid protein is stably carried by the resulting recombinant virus.
  • plasmids constructed from vaccinia virus DNA are employed.
  • expression vector includes plasmids, cosmids or phages capable of synthesizing a protein encoded by the respective recombinant gene carried by said vector. Such vectors are independently replicated in or capable of integration into the chromosome of an appropri ⁇ ate host cell for expression of the amyloid protein.
  • a cell has been "transformed” by exogenous or heterologous DNA when such DNA has been introduced inside the cell.
  • the transforming DNA may or may not be integrated (covalently linked) into chromosomal DNA making up the genome of the cell.
  • the transforming DNA may be maintained on an episomal element such as a plasmid.
  • the cell has been stably transformed when the cell is able to establish cell lines or clones comprised of a population of daughter cells containing the transforming DNA.
  • a "clone” is a population of cells derived from a single cell or common ancestor by mitosis.
  • a "cell line” is a clone of a cell that is capable of stable growth in vitro for many generations.
  • Beta-Amyloid Coding Sequences The beta-amyloid genes may be synthetic or natural, or combinations thereof.
  • the gene encoding the natural 751 amino acid precursor protein is described in PCT WO88/03951, published 2 June 1988 and assigned to the same assignee of the present application, and the expres- sion of the protein in mammalian cells is provided in
  • Example 4 therein. The relevant portions of this publica ⁇ tion are specifically incorporated herein by reference.
  • the genes encode the A42 core protein or an amyloid protein, A99, which comprises the A42 core protein and the cytoplasmic domain.
  • A99 which comprises the A42 core protein and the cytoplasmic domain.
  • This latter protein consists of the 42 residue core protein and 57 amino acids of the cytoplasmic domain of the 751 precursor protein.
  • the sequence of A99 is as follows:
  • genes are provided for expression of the desired protein using recombinant DNA expression vectors.
  • these genes may be natural, synthetic or combinations thereof.
  • it may be desirable to modify the natural amyloid nucleic acid sequence. For example, it will often be preferred to use codons which are preferentially recognized by the desired host.
  • it may be desirable to further alter the nucleotide sequence, either synthetic or natural, to create or remove restriction sites to, for example, enhance insertion of the gene sequence into convenient expression vectors or to substitute one or more amino acids in the resulting polypeptide to increase stability.
  • a general method for site-specific mutagenesis is described in Noren et al., (1989) Science 244:182-188.
  • Peptides of this precursor protein are also provided herein for the generation of specific immunological re- agents and may also be synthetic or natural.
  • Synthetic oligonucleotides are prepared by either the phosphotriester method as described by Edge et al., (1981) Nature 292:756 and Duckworth et al., (1981) Nuc Acids Res ⁇ :1691 or the phosphoramidite method as described by Beaucage and Caruthers, (1981) Tet Lett 22:1859 and Matteucci and Caruthers, (1981) J Am Chem Soc 103:3185, and can be prepared using commercially available automated oligonucleotide synthesizers.
  • the coding sequences for the amyloid proteins can be inserted into vaccinia virus plasmid insertion vec ⁇ tors for the purpose of generating recombinant vaccinia viruses using the methods described in Moss et al., (1983) Methods in Gene Amplification, Vol. 3, Elsevier-North Hol ⁇ land, p. 202-213; and in Moss et al., (1984) J Virol 4jJ:857-864.
  • the amyloid-vaccinia recombinants can then be used for (1) expression of the respective amyloid protein and analysis of preamyloid formation, and (2) production of amyloid antibodies.
  • the two vaccinia virus insertion vectors pSCll (Chakrabarti et al., (1985) Mol Cell Biol 5:3403-3409 and pUVl (Falkner et al., (1987) Nuc Acids Res 15:7192) were used for the expression of the amyloid proteins and generation of amyloid-vaccinia recombinants. Both vectors are of the co-insertion variety and each contains two vaccinia virus promoters. One promoter (PI) is used to drive the expression of a selectable marker gene (in this case, beta-galactosidase) while the other promoter (P2) is used to drive expression of the heterologous amyloid DNA insert.
  • PI promoter
  • P2 is used to drive expression of the heterologous amyloid DNA insert.
  • the pSCll vector utilizes a vaccinia early-late promoter (P7.5) to drive heterologous gene expression and has a single S al cloning site.
  • the pUVl vector utilizes a vaccinia late promoter (Pll) to drive heterologous gene expression and is designed for the expression of fusion proteins behind the ATG of the Pll late gene. In all cases, amyloid-pUVl constructs were made using the most 5' (after the ATG) cloning site (EcoRI) in order to avoid introduction of additional amino terminal amino acids into the native amyloid protein sequence.
  • procaryotic and eucaryotic systems may be used to express the amyloid genes described herein.
  • Procaryotes most frequently are represented by various strains of E. coli; however, other microbial strains may also be used.
  • Plasmid vectors which contain replication sites, selectable markers and control sequences derived from a species compatible with the host are used; for example, E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species by Bolivar et al. , (1977) Gene 2:95.
  • pBR322 contains genes for ampicillin and tetracycline resistance, and thus provides multiple selectable markers which can be either retained or destroyed in constructing the desired vector.
  • Commonly used procaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, include such commonly used promoters as the beta-lactamase (penicillinase) and lactose (lac) promoter systems (Chang et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel et al. , (1980) Nucleic Acids Res 8:4057), the lambda-derived P-. promoter (Shimatake et al. , (1981)
  • eucaryotic microbes such as yeast
  • yeast may also be used as hosts.
  • Laboratory strains of Saccharomyces cerevisiae, Baker's yeast are most used although a number of other strains or species are commonly available.
  • Vectors employing, for example, the 2 micron origin of replication of Broach, (1983) Meth Enz 101:307, or other yeast compatible origins of replica- tion (see, for example, Stinchcomb et al., (1979) Nature 282:39; Tschumper et al., (1980) Gene 10:157 and Clarke et al., (1983) Meth Enz 101:300) may be used.
  • Control sequences for yeast vectors include promoters for the synthesis of glycolytic enzymes (Hess et al. , (1968) J Adv Enzyme Reg 7:149; Holland et al. , (1978) Biochemistry
  • promoters known in the art include the promoter for 3-phosphoglycerate kinase (Hitzeman et al., (1980) J Biol Chem 255:2073).
  • Other promoters which have the additional advantage of transcription controlled by growth conditions and/or genetic background are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, the alpha factor system and enzymes responsible for maltose and galactose utilization. It is also believed terminator sequences are desirable at the 3' end of the coding sequences. Such terminators are found in the 3' untranslated region fol ⁇ lowing the coding sequences in yeast-derived genes.
  • eucaryotic host cell cultures derived from multicellular organisms. See, for example, Axel et al. , U.S. Patent No. 4,399,216. These systems have the additional advantage of the ability to splice out introns and thus can be used directly to express genomic ragments.
  • Useful host cell lines include VERO, HeLa, baby hamster kidney (BHK), CV-1, COS, MDCK, NIH 3T3, L, and Chinese hamster ovary (CHO) cells. Expression vectors for such cells ordinarily include promoters and control sequences compatible with mammalian cells such as, for example, the commonly used early and late promoters from Simian Virus 40 (SV40) (Fiers et al., (1978) Nature
  • viral promoters such as those derived from polyoma, herpes virus, Adenovirus 2, bovine papilloma virus, or avian sarcoma viruses.
  • the controllable promoter, hMTII Kerin et al., (1987) Nature 299:797-802
  • hMTII Kerin et al., (1987) Nature 299:797-802
  • Insect expression systems may also be employed to express the amyloid genes.
  • the baculovirus polyhedrin gene has been employed for high- level expression of heterologous proteins (Smith et al., (1983) Mol Cell Biol 3(12) :2156-2165; Summers et al., "Genetic Engineering of the Genome of the Autographa Californica Nuclear Polyhedrosis Virus", Banbury Report: Genetically Altered Viruses in the Environment, 22:319- 339, Cold Spring Harbor Laboratory, 1985).
  • the amyloid DNA clones expressed in vaccinia can also be used to generate stably transfected cell lines expressing the amyloid proteins.
  • these cell lines are generated by first constructing one of two expression plasmids.
  • the selectable marker is provided by a G418 neomycin expres ⁇ sion cassette (neo) consisting of the SV40 early promoter, the bacterial kanamycin-resistance gene also containing its own promoter, the SV40 intervening sequence, and the SV40 polyadenylation site from the early region.
  • the amyloid DNA cloning site is flanked at the 5' end by the human metallothionein gene promoter, pMtlla, modified with an SV40 enhancer, and at the 3' end by the SV40 polyadenylation site from the early region.
  • the amyloid DNA cloning site is flanked at the 5' end by a beta-actin promoter, and at the 3' end by a sequence encoding a use ⁇ ful polyadenylation site, such as that of the SV40 early region or the beta-actin gene.
  • Each of the vectors described above can be transformed into a mammalian cell line such as, but not limited to, those described in the following examples by either calcium phosphate-DNA coprecipitation or electro- poration.
  • the cells are subjected to 1 mg/ml G418 to provide pools of G418-resistant colonies.
  • Suc ⁇ cessful transformants also having a stable inheritance of the DNA contained in the expression construct, are then plated at low density for purification of clonal isolates. Clonal isolates are then analyzed for maximum production of the amyloid protein of interest and high-producing clones are expanded to serve as stock.
  • the amyloid protein may need to be conjugated to a carrier protein such as bovine serum albumin or keyhole limpet hemacyanin using a chemical process which employs carbodiimide, glutaraldehyde, or other cross-linking agents.
  • a carrier protein such as bovine serum albumin or keyhole limpet hemacyanin
  • the protein may be administered without being conjugated to a carrier protein.
  • Vaccinia virus re ⁇ combinants which are expressing amyloid proteins may also be used to prepare antibodies. The vaccinia virus re ⁇ combinants are injected into an animal and then the animal is boosted several weeks after the initial immunization. Ten days to two weeks later the animals are bled and antiserum is collected and analyzed for titer.
  • Monoclonal antibodies are commonly prepared by fusing, under appropriate conditions, B-lymphocytes of an animal which is making polyclonal antibodies with an im ⁇ mortalizing myeloma cell line.
  • the B-lymphocytes can be spleen cells or peripheral blood lymphocytes.
  • Techniques for fusion are also well known in the art, and in general, involve mixing the cells with a fusing agent such as poly ⁇ ethylene glycol.
  • Successful hybrid ⁇ ma formation is as ⁇ sessed and selected by standard procedures such as, for example, HAT medium. From among successful hybridomas, those secreting the desired antibody are screened by as ⁇ saying the culture medium for their presence.
  • Standard immunological techniques such as ELISA (enzyme-linked immun ⁇ assay) , RIA (radioimmunoassay) , IFA (immunofluorescence assay) and Western blot analysis, which are well known in the art, can be employed for diagnostic screening for amyloid expression.
  • ELISA enzyme-linked immun ⁇ assay
  • RIA radioimmunoassay
  • IFA immunofluorescence assay
  • Western blot analysis which are well known in the art, can be employed for diagnostic screening for amyloid expression.
  • One typical arrangement utilizes competition, between labeled antigen (e.g. amyloid protein) and the analyte, for the antibody, followed by physical separation of bound and unbound fractions.
  • labeled antigen e.g. amyloid protein
  • Analyte competes for the binding of the labeled antigen; hence more label will remain in the unbound fraction when larger amounts of analyte are present.
  • this competitive-binding type assay the sample is incubated with a known titer of labeled amyloid protein and amyloid protein antibody.
  • Antibody-protein complex is then separated from uncomplexed reagents using known techniques and the amount of label in the co plexed material is measured, e.g.
  • the amount of amyloid protein in the sample is determined by comparing the measured amount of label with a standard curve.
  • the present assay provides one of the first steps in addressing the question whether preamyloid corti- cal deposits herald organic dementia.
  • Down's syndrome is the one known disease closely related to the proposed etiology of AD.
  • NFTs congophilic angiopathy
  • senile plaques As reported in Giacione et al.
  • the expression of the amyloid protein is initiated by culturing the transformed cell line under conditions which are suitable for cell growth and expression of the amyloid protein.
  • high level expression of the protein is preferred.
  • a CHO cell line transformed with a beta-actin vector comprising the DNA encoding the A42 or A99 amyloid protein is grown in a mammalian culture medium such as, for example, a 1:1 mixture of F12 medium and DME medium with 10% fetal calf serum for 5-72 hr at 37°C.
  • Transfected viral monolayers are selected and plaque purified, and stocks of amyloid-vaccinia recombinant viruses are prepared.
  • preamyloid aggregates can be monitored by standard immunocytochemical methods using, for example, beta-amyloid primary antibodies which are detected using a secondary, labeled anti-antibody. If one is interested in testing whether the compound of interest can inhibit preamyloid formation, the compound is introduced to the culture medium before monitoring for preamyloid aggregation. Alternatively, the compound is introduced to the culture medium after preamyloid forma ⁇ tion has been established and this reaction mixture is monitored to see whether the compound induces amyloid resorption.
  • Potential therapeutic compounds for use in the present invention include, for example, amyloid-fibril denaturing agents such as dimethyl sulfoxide, and cytotoxic agents such as colchicine and chlorambucil.
  • amyloid-fibril denaturing agents such as dimethyl sulfoxide
  • cytotoxic agents such as colchicine and chlorambucil.
  • the efficacy of these agents may be monitored through observa ⁇ tion of reduced antibody binding to the amyloid deposit. Reduction in such binding is indicative of reduced preamyloid deposition.
  • preamyloid formation in the host cell may trigger other cellular events which could be employed as markers unrelated to the etiology of Alzheimer's disease, but correlative with the presence of preamyloid deposits.
  • an increase in the level of certain enzymes, specifically proteases may be measured in lieu of the preamyloid deposition.
  • an increase in the concentration levels of these enzymes is observed when cultured cells are subjected to stress.
  • kits suitable for the above diagnostic or screening methods contain the appropriate reagents and are constructed by packaging the appropriate materials, including the preamyloid protein aggregates immobilized on a solid support with labeled antibodies in suitable containers, along with any other reagents (e.g., wash solutions, enzyme substrate, anti-amyloid antibodies) or other materials required for the conduct of the assay.
  • the reagents are usually premeasured for ease of use.
  • An optional component of the kit is a set of instructions describing any of the available immunoassay methods.
  • a kit for a direct assay can comprise preamyloid proteins aggregates immobilized on a solid immunoassay support and a container comprising labeled antibody to the amyloid protein, as well as the other reagents mentioned above.
  • the following examples describe the expression vectors containing the 42 amino acid plaque core region (A42), and the 42 amino acid plaque core region including the 57 amino acid adjacent carboxy-terminal region of the beta-amyloid precursor protein (A99).
  • Alternative constructs for the A42 and A99 constructs were prepared which included a 17 amino acid amyloid signal sequence. As these constructs did not express the amyloid protein well, further experimentation with these vectors was not performed.
  • FIG. 1 il ⁇ lustrates the various amyloid expression constructs, all of which were modified at the 5' end to satisfy the clon ⁇ ing constraints of the vaccinia Pll promoter in the pUVl vector. Specifics for each construct are as follows:
  • the A42-encoding sequence (nucleotides 2080 to 2205, numbered in accordance with the 751 amyloid pre- cursor sequence) was synthesized as a 145 basepair (bp) EcoRI-BamHI oligomer, provided below, containing the ap ⁇ intestinalte TGA stop codon and an amino-terminal Asn-Ser adaptor sequence:
  • the synthetic fragment was ligated to EcoRI- and BamHI-digested pGeml (Promega-Biotec) , deriving pGemA42.
  • the EcoRI-BamHI fragment of pGemA42 was subsequently isolated and ligated into the EcoRI-BamHI site of pUVl deriving pUVl:A42.
  • the 590 bp Ddel-PvuII fragment of plasmid 4T4B was isolated from the carboxy-terminal 1 kilobase (kb) EcoRI fragment of 4T4B and ligated with a 27 bp EcoRI-Ddel adaptor sequence and cloned into the EcoRI- and Smal- digested pUVl, deriving pUVl:A99.
  • the 761 bp Xbal-Sall fragment of pUVl:A99 was further subcloned into the Xbal-Sall vector fragment of mpl8 and pGem2. Sequence data confirmed the predicted sequence.
  • the vaccinia insertion vectors described in Example 1 were used to generate amyloid-vaccinia re- combinant viruses as follows.
  • Confluent monolayers of CV-1 cells in 60 mm dishes were infected with vaccinia virus (Wyeth strain) at a multiplicity of infection (moi) of 0.05 pfu/cell.
  • vaccinia virus Wild-type vaccinia virus DNA
  • m multiplicity of infection
  • the cells were transfected with a calcium phosphate precipitate of 10 ug insertion plasmid DNA and 0.5 ug wild-type vaccinia virus DNA.
  • Cells were fed with complete medium and incubated at 37°C for two days.
  • Monolayers were collected and TK- vaccinia viruses were selected on TK-143 cells in the presence of 5- bromodeoxyuridine (BudR) at 25 ug/ l.
  • BudR 5- bromodeoxyuridine
  • the beta-amyloid antibodies were generated from synthetic peptides.
  • the synthetic peptides were prepared using solid phase synthesis according to standard protocols. Purification of the crude peptides was ac ⁇ complished by desalting with gel filtration followed by ion-exchange chromatography and preparative reverse-phase liquid chromatography. Each peptide was fully character ⁇ ized by amino acid composition and sequence analysis. COOH-CORE corresponds to amino acids 653-
  • C00H-B2 and C00H-C2 correspond to amino acids 736-751(NGYENPTYKFFEQMQN)
  • C00H-B3 and C00H-C3 correspond to amino acids 705-719(KKKQYTSIHHGWEV)
  • C00H-C5 corresponds to amino acids 729- 742(HLSKMQQNGYENPT) .
  • Antibody titers against the appropriate peptide were determined by enzyme-linked immunosorbent assays coupled with horseradish peroxidase and found to be 7.4xl0 4 , 2.7xl0 5 , IxlO 5 , 9.1xl0 6 , 8.2xl0 5 , and 2.5xl0 5 for COOH-CORE, C00H-B2, C00H-C2, C00H-B3, C00H-C3, and C00H- C5, respectively.
  • Antibodies to 9523 correspond to amino acids 673-685(AEDVGSKNGAIIG) and 9524 correspond to amino acids 701-712(LVMLKKKQYTSI) .
  • Antibodies to these two peptides were generated by coinjecting New Zealand white rabbits each with 200 ug methylated bovine serum albumin (PBS) plus 200 ug of the respective synthetic peptide in PBS. Rabbits were boosted one, two and three weeks following primary inoculation with identical amounts of peptide.
  • PBS methylated bovine serum albumin
  • CV-1 cells were infected with W:99 at a multiplicity of infection of one.
  • 35S-methionine 250 uCi/ml was added at 20 hr post infection for 4 hr.
  • the expression product of W:A99 demonstrated high level expression of the 99 amino acid core protein and showed evidence of self-aggregation as well as ag ⁇ gregation with other proteins or self-aggregation combined with proteolysis since multi ers of A99 did not always occur in integers of 11.5-17 kd.
  • SK-N-MC ATCC # HTB10
  • IMR-32 ATCC # CCL127
  • the culture medium for each host was as follows:
  • CV-1 The medium was Eagle MEM supplemented with 10% FBS, penicillin, streptomycin and L- Gin-
  • SK-N-MC Eagle MEM supplemented with 10% FBS, non- essential amino acids, penicillin, streptomycin and L-Gln.
  • PC-12 DMEM21, 5% DHS, 5% DFBS and L-Gln;
  • IMR-32 Eagle MEM (Hank's BSS) and 10% deltaFBS plus nonessential amino acids, penicillin, streptomycin and L-Gln.
  • Each cell line was grown to confluency on a microscope slide divided into 4 individual chambers (Lab Tech) .
  • One chamber was mock infected, the second infected with a control recombinant virus lacking A4 sequences
  • FIG. 3 shows fluorescent photomicrographs of CV-
  • a number of constructs expressing the beta- amyloid core protein were constructed using a derivative of the beta-actin expression/selection vector designated pHbetaAPr-1-neo.
  • This vector illustrated in FIG. 4, is a combination of the following elements: a) bp 1-4300 is the 4.3 kb EcoRI-AluI fragment from the human beta-actin gene isolate pl4Tbeta-17 (Leavitt et al. , (1984) Mol Cell Biol 4:1961-1969). For sequencing details of the promoter see Ng et al. , (1985) Mol Cell Biol 5:2720-2732. The cap site, 5' untranslated region and IVS 1 positions are indicated in FIG. 4.
  • bp 4300-4320 is in part derived from pSP64 polylinker (Melton et al., (1984) Nuc Acids Res 12:7035- 7056); c) bp 4320-6600 is derived from pcDVl (Okayama & Berg, (1983) Mol Cell Biol 3:280-289); and d) bp 6600-10000 is the PvuII-EcoRI fragment from pSV-neo (Southern & Berg, (1982) J Mol App Genet 1:327-341) containing the bacterial neo ycin gene linked to the SV40 origin plus early promoter.
  • Beta-actin A42 was constructed by excising the EcoRI-BamHI 145 bp fragment from pGEM-A42, adding a Sall- EcoRI adaptor sequence (5'-TCG ACA TGG ATG CAC AAT TA-3') and cloning into the pAX-neo expression vector at the Sall-and BamHI sites.
  • the beta-actin A99 plasmid was constructed by excising the 670 bp EcoRI-Hindlll fragment of pGEM_-A99, adding the above-described Sall-EcoRI adap ⁇ tor sequence and cloning into the pAX-neo vector at the Sail and HindiII sites.
  • Each construct was introduced into CHO cells by the calcium phosphate precipitation method using 7 ug of each DNA per 10 cells, and a resistant population was selected with G418-neomycin. The efficiency of
  • transfection for the A99 or A42 constructs was over 10 for 10 cells and pools of cells transfected with either beta-actin A99 or with beta-actin A42 were selected using G418-neomycin resistance (500 ug/ml).
  • Cells infected with W:99 or W:42 which are capable of forming amyloid deposits are plated in a 96- well microtiter plate.
  • an automated pipetter is used to introduce the drug to be tested to the cells.
  • a range of concentrations of the drug is incubated in a tissue culture incubator (or preincubated) with the cells at 37 C for a predetermined time period, or alternatively, for 3 to 72 hours.
  • the culture media is removed, and the cells are prepared for preamyloid measurement as follows.
  • the cells are fixed for immunocytochemical staining with amyloid antibodies.
  • the primary antibodies are introduced followed by incubation with labeled, secondary anti-antibodies and the level of binding between the primary and secondary antibodies is measured using an ELISA plate reader to record the optical density of the labeled antibody.
  • a smaller optical density reading as compared to a control sample of cells grown in the absence of the test drug is indicative of that drug's ability to inhibit amyloid deposition.
  • This procedure may be modified to permit detection of preamyloid dissolution using a correlative enzyme marker.

Abstract

The present invention provides an in vitro tissue culture-based assay for amyloid deposition specific for Alzheimer's disease which is suitable for routine drug screening analysis. Immunological diagnostic reagents for Alzheimer's disease are also provided.

Description

ASSAYS AND REAGENTS FOR AMYLOID DEPOSITION
Field of the Invention
The present invention relates to assays and re¬ agents useful for the chemical intervention of amyloidosis in Alzheimer's disease.
Background of the Invention
Alzheimer's disease (AD) is an age-related brain degenerative disease that is the most common cause of intellectual failure in late life. Neuritic or senile plaques and neurofibrillary tangles (NFT) are the hallmark characteristic of the histopathology of Alzheimer's brains. These plaques and tangles are believed to result from deposits of two different proteins which share the properties of the amyloid class of proteins specific for AD. The major protein component of amyloid is an ~4 kilodalton (kd) protein, designated the beta-protein or A4 protein due to a partial beta pleated structure or its molecular weight, respectively. The amino acid sequence of A4 has been defined (Wong et al., (1985) Proc Natl Acad Sci USA 82:8729-8732) and full-length cDNA encoding a primary translation product of 695 residues has been cloned (Kang et al. , (1987) Nature 325;733-736) while other cDNAs have been identified which encode a 751- residue or 770-residue precursor form (Ponte et al. , (1988) Nature 331:525-527; Tanzi et al. , (1988) Nature _331:528-530; and Kitaguchi et al., (1988) Nature 331:530- 532) .
The A4 protein accumulates extracellularly, both in brain parenchyma and in the walls of blood vessels, generally as amyloid plaques which form aggregate fibril structures and are insoluble on SDS-polyacrylamide gels. The fibrils are generally identified as amyloid based on their green birefringence after staining with Congo red and their 40- to 90-A diameter. The second protein, mentioned previously, ac¬ cumulates intracellularly in neurons of Alzheimer's brains (Castano and Frangione, (1988) Lab Invest 58:122-132) and forms tangles composed of structures resembling paired helical filaments (PHFs). In contrast to the beta-amyloid protein, the primary structure and number of proteins comprising PHFs are unknown. PHF-containing neurites are found in the periphery of the plaque, whereas deposits of beta-amyloid protein form the central core of mature plaques, surrounded by degenerated neurites and glial cells.
Although the etiology of AD is unknown, it has been demonstrated that the frequency of neuritic plaques found in the cortex of AD patients correlates with the degree of dementia (Roth et al., (1966) Nature 209:109- 110; Wilcock and Esiri, (1982) J Neurol Sci 56:343-356). The therapeutic goals in amyloidosis are to prevent further deposition of amyloid material and to promote or accelerate its resorption. To date, there are no means available to treat the pathogenesis of AD and the paucity of understanding concerning the mechanism of amyloid formation in AD is a major obstacle in the development and design of therapeutic agents that can intervene in this process. Moreover, no animal models for brain amyloidosis with beta-amyloid protein deposits or PHFs exist, creating yet another obstacle to test such putative therapeutic agents. Logical therapeutic approaches are now, however, emerging for treating the particular amyloidosis associ¬ ated with AD. These approaches are attributable, in part, from the successes and failure gained in attempting to treat other forms of amyloidosis, such as the use of dimethyl sulfoxide which blocks amyloid formation from Bence Jones proteins in vitro (Coria et al. , (1988) Lab Invest 58:454-458) and use of colchicine to reduce the size of renal amyloid deposits and induce clinical remis- sions in several cases of familial Mediterranean fever and amyloid nephropathy (Ravid et al. , (1977) Ann Intern Med 87:568-570) .
Efforts directed to the design of in vitro models of age-related cerebral amyloidogenesis using A4- derived synthetic peptides are disclosed in Castano et al., (1986) Biochem Biophys Res Comm 141:782-789, and in Kirschner et al., (1987) Proc Natl Acad Sci USA 84:6953- 6957. Castano et al. demonstrated that amyloid fibrils could be formed in vitro when using a synthetic peptide corresponding to the amino-terminal 28 residues of the amyloid core protein. This 28 residue peptide, as well as a 17 residue sequence contained within the 28 amino acids, both formed fibrils which stain similarly to material isolated from Alzheimer's brains; however, the synthetic amyloid fibrils were soluble, unlike the naturally occur¬ ring insoluble amyloid isolated from Alzheimer's brains. Kirschner et al. demonstrated that the same 28 residue peptide could be produced as an insoluble aggregate; however, this particular in vitro model is not expected to correlate well to the cellular environment in which amyloid deposition occurs.
Dyrks et al., (1988) EMBO J 7:949-957 showed that a shortened cell-free translation product comprising the amyloid A42 part and the cytoplasmic domain of the 695-residue precursor can form multimers. While aggregation was observed employing an in vitro cell-free syste , this system fails to reveal whether aggregation of the translation product would naturally follow in vivo. Moreover, the in vitro cell-free system does not address protein stability issues, that is, whether adequate levels of the protein could be expressed, whether protein proteolysis exists, and other concerns generally associated with in vivo expression of recombinant proteins.
Therefore, there exists a need for a definitive cellular deposition model with which one may assay agents capable of chemically intervening in the process of amyloid deposition. Such a method should be relatively simple to perform and should be highly specific in distinguishing AD plaques from the plaques associated with other disorders. Furthermore, it is desirable that the assay be capable of being reduced to a standardized format. The present invention satisfies such needs and provides further advantages.
Summary of the Invention
The present invention provides a method for determining the ability of a potential therapeutic agent to intervene in the amyloid deposition process associated with Alzheimer's disease in a cellular environment, which method utilizes a recombinantly produced amyloid substrate in a screening assay. The present invention also allows for the development and use of immunological reagents to detect the formation of preamyloid protein aggregation in the cell lines provided by the invention. To achieve the objects and in accordance with the purpose of the invention, as embodied and broadly described herein, a method of screening agents capable of intervention in Alzheimer's disease amyloidosis comprises: a) culturing a cell line capable of expressing a gene encoding beta-amyloid protein under conditions suit- able to produce the beta-amyloid protein as an insoluble, preamyloid aggregate; b) combining a known quantity of the agent to be tested to the cell culture; and c) monitoring the combination to determine whether preamyloid aggregate formation is reduced.
In an alternative embodiment of the invention, preamyloid formation can be induced through infection of a cell line with a recombinant virus capable of expressing the beta-amyloid protein as an insoluble preamyloid aggregate. Such recombinant viruses carry expression vectors comprising DNA encoding the beta-amyloid protein.
Immunoassay kits employing the reagents useful to screen potential amyloid intervening agents are also provided by the present invention.
Brief Description of the Drawings
FIG. 1 is a schematic illustration of two amyloid expression constructs employing the vaccinia pUVl insertion vector.
FIG 2. illustrates the results of immunoprecipitation of 35S-methionine labeled W:A99 infected CV-1 cell lysates using APCP antibodies. The arrows mark A99 protein. FIG 3. are fluorescent photomicrographs of infected CV-1 cells stained with APCP antibodies. FIG. 3A is a Mock control; FIG. 3B is a W:CONT control; FIG. 3C is the W:99 construct; and FIG. 3D is the W:42 construct. The magnification is 200x with a 0.4 second exposure time for each photo.
FIG. 4 is a illustration of the modified beta- actin expression selection vector, pAX-neo, that was employed to express the beta-amyloid core constructs in mammalian cells. Detailed Description of the Preferred Embodiments
As indicated above, the invention involves a method of screening agents capable of intervention in Alzheimer's disease amyloidosis. As used herein, the term "beta-amyloid core protein" or "A4 protein" refers to an approximately 4 kd protein first identified by Glenner and Wong, (1984) Biochem Biophvs Res Comm 120:885, which is defined at the amino terminus by sequence analysis as a mixture of four peptides with slightly different amino termini, the amino termini of the three smaller peptides being completely encoded by that of the largest.
The term "beta-amyloid precursor protein" refers to either the amyloid precursor protein of 695 amino acids (Kang et al., (1987) supra) or the 751 amino acid protein (Ponte et al., (1988) supra) containing within their sequence, the beta-amyloid core protein sequence defined above. The A4 core protein begins at amino acid 597 of the 695 amino acid protein and at amino acid 653 of the 751 amino acid sequence.
The terms "preamyloid aggregation", "preamyloid formation", or "preamyloid deposits" refer to a morphological description — first discovered by Tagliavini et al., (1988) Neurosci Lett 93:191-196 — of spherical, granular deposits which are considerably smaller than pre-plaques and plaques found at a high frequency in the brains of Alzheimer's victims. These deposits can be occasionally detected with silver stain but not with Congo red, a stain to which amyloid proteins demonstrate high binding affinity.
As used herein, the term "insertion vector" includes plasmids, cosmids or phages capable of mediating homologous recombination into a viral genome such that the DNA encoding the beta-amyloid protein is stably carried by the resulting recombinant virus. In one embodiment of the invention plasmids constructed from vaccinia virus DNA are employed.
The term "expression vector" includes plasmids, cosmids or phages capable of synthesizing a protein encoded by the respective recombinant gene carried by said vector. Such vectors are independently replicated in or capable of integration into the chromosome of an appropri¬ ate host cell for expression of the amyloid protein.
A cell has been "transformed" by exogenous or heterologous DNA when such DNA has been introduced inside the cell. The transforming DNA may or may not be integrated (covalently linked) into chromosomal DNA making up the genome of the cell. In prokaryotes, yeast, and mammalian cells, for example, the transforming DNA may be maintained on an episomal element such as a plasmid. The cell has been stably transformed when the cell is able to establish cell lines or clones comprised of a population of daughter cells containing the transforming DNA. A "clone" is a population of cells derived from a single cell or common ancestor by mitosis. A "cell line" is a clone of a cell that is capable of stable growth in vitro for many generations.
A. Beta-Amyloid Coding Sequences The beta-amyloid genes may be synthetic or natural, or combinations thereof. The gene encoding the natural 751 amino acid precursor protein is described in PCT WO88/03951, published 2 June 1988 and assigned to the same assignee of the present application, and the expres- sion of the protein in mammalian cells is provided in
Example 4 therein. The relevant portions of this publica¬ tion are specifically incorporated herein by reference.
The genes encode the A42 core protein or an amyloid protein, A99, which comprises the A42 core protein and the cytoplasmic domain. This latter protein consists of the 42 residue core protein and 57 amino acids of the cytoplasmic domain of the 751 precursor protein. The sequence of A99 is as follows:
10 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin 20 30
Lys Leu Val Phe Phe Ala Glu Asp Val Gly Ser Asn Lys Gly Ala
40 (42) He He Gly Leu Met Val Gly Gly Val Val He Ala Thr Val He
50 60 Val He Thr Leu Val Met Leu Lys Lys Lys Gin Tyr Thr Ser He
70 His His Gly Val Val Glu Val Asp Ala Ala Val Thr Pro Glu Glu
80 90
Arg His Leu Ser Lys Met Gin Gin Asn Gly Tyr Glu Asn Pro Thr (99)
Tyr Lys Phe Phe Glu Gin Met Gin Asn.
These genes are provided for expression of the desired protein using recombinant DNA expression vectors. As mentioned above, these genes may be natural, synthetic or combinations thereof. When preparing a synthetic nucleotide sequence, it may be desirable to modify the natural amyloid nucleic acid sequence. For example, it will often be preferred to use codons which are preferentially recognized by the desired host. In some instances, it may be desirable to further alter the nucleotide sequence, either synthetic or natural, to create or remove restriction sites to, for example, enhance insertion of the gene sequence into convenient expression vectors or to substitute one or more amino acids in the resulting polypeptide to increase stability. A general method for site-specific mutagenesis is described in Noren et al., (1989) Science 244:182-188.
Peptides of this precursor protein, for example, those derived from the A4 core protein, are also provided herein for the generation of specific immunological re- agents and may also be synthetic or natural. Synthetic oligonucleotides are prepared by either the phosphotriester method as described by Edge et al., (1981) Nature 292:756 and Duckworth et al., (1981) Nuc Acids Res ^:1691 or the phosphoramidite method as described by Beaucage and Caruthers, (1981) Tet Lett 22:1859 and Matteucci and Caruthers, (1981) J Am Chem Soc 103:3185, and can be prepared using commercially available automated oligonucleotide synthesizers.
B. Vaccinia Viral Vectors
The coding sequences for the amyloid proteins can be inserted into vaccinia virus plasmid insertion vec¬ tors for the purpose of generating recombinant vaccinia viruses using the methods described in Moss et al., (1983) Methods in Gene Amplification, Vol. 3, Elsevier-North Hol¬ land, p. 202-213; and in Moss et al., (1984) J Virol 4jJ:857-864. The amyloid-vaccinia recombinants can then be used for (1) expression of the respective amyloid protein and analysis of preamyloid formation, and (2) production of amyloid antibodies.
The two vaccinia virus insertion vectors, pSCll (Chakrabarti et al., (1985) Mol Cell Biol 5:3403-3409 and pUVl (Falkner et al., (1987) Nuc Acids Res 15:7192) were used for the expression of the amyloid proteins and generation of amyloid-vaccinia recombinants. Both vectors are of the co-insertion variety and each contains two vaccinia virus promoters. One promoter (PI) is used to drive the expression of a selectable marker gene (in this case, beta-galactosidase) while the other promoter (P2) is used to drive expression of the heterologous amyloid DNA insert. Both are flanked by vaccinia virus DNA (an inter¬ rupted thymidine kinase [tk] gene) which facilitates homologous recombination into a wild-type vaccinia virus genome and provides a selection mechanism (generation of tk minus viruses). The pSCll vector utilizes a vaccinia early-late promoter (P7.5) to drive heterologous gene expression and has a single S al cloning site. The pUVl vector utilizes a vaccinia late promoter (Pll) to drive heterologous gene expression and is designed for the expression of fusion proteins behind the ATG of the Pll late gene. In all cases, amyloid-pUVl constructs were made using the most 5' (after the ATG) cloning site (EcoRI) in order to avoid introduction of additional amino terminal amino acids into the native amyloid protein sequence.
C. Recombinant Expression Vectors and Hosts
It will also be understood by those skilled in the art that both procaryotic and eucaryotic systems may be used to express the amyloid genes described herein. Procaryotes most frequently are represented by various strains of E. coli; however, other microbial strains may also be used. Plasmid vectors which contain replication sites, selectable markers and control sequences derived from a species compatible with the host are used; for example, E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species by Bolivar et al. , (1977) Gene 2:95. pBR322 contains genes for ampicillin and tetracycline resistance, and thus provides multiple selectable markers which can be either retained or destroyed in constructing the desired vector. Commonly used procaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, include such commonly used promoters as the beta-lactamase (penicillinase) and lactose (lac) promoter systems (Chang et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel et al. , (1980) Nucleic Acids Res 8:4057), the lambda-derived P-. promoter (Shimatake et al. , (1981)
Nature 292:128) and N-gene ribosome binding site, and the trp-lac (trc) promoter system (A ann and Brosius, (1985) Gene 40:183) .
In addition to bacteria, eucaryotic microbes, such as yeast, may also be used as hosts. Laboratory strains of Saccharomyces cerevisiae, Baker's yeast, are most used although a number of other strains or species are commonly available. Vectors employing, for example, the 2 micron origin of replication of Broach, (1983) Meth Enz 101:307, or other yeast compatible origins of replica- tion (see, for example, Stinchcomb et al., (1979) Nature 282:39; Tschumper et al., (1980) Gene 10:157 and Clarke et al., (1983) Meth Enz 101:300) may be used. Control sequences for yeast vectors include promoters for the synthesis of glycolytic enzymes (Hess et al. , (1968) J Adv Enzyme Reg 7:149; Holland et al. , (1978) Biochemistry
17:4900). Additional promoters known in the art include the promoter for 3-phosphoglycerate kinase (Hitzeman et al., (1980) J Biol Chem 255:2073). Other promoters, which have the additional advantage of transcription controlled by growth conditions and/or genetic background are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, the alpha factor system and enzymes responsible for maltose and galactose utilization. It is also believed terminator sequences are desirable at the 3' end of the coding sequences. Such terminators are found in the 3' untranslated region fol¬ lowing the coding sequences in yeast-derived genes.
It is also, of course, possible to express genes encoding polypeptides in eucaryotic host cell cultures derived from multicellular organisms. See, for example, Axel et al. , U.S. Patent No. 4,399,216. These systems have the additional advantage of the ability to splice out introns and thus can be used directly to express genomic ragments. Useful host cell lines include VERO, HeLa, baby hamster kidney (BHK), CV-1, COS, MDCK, NIH 3T3, L, and Chinese hamster ovary (CHO) cells. Expression vectors for such cells ordinarily include promoters and control sequences compatible with mammalian cells such as, for example, the commonly used early and late promoters from Simian Virus 40 (SV40) (Fiers et al., (1978) Nature
273:113) , or other viral promoters such as those derived from polyoma, herpes virus, Adenovirus 2, bovine papilloma virus, or avian sarcoma viruses. The controllable promoter, hMTII (Karin et al., (1987) Nature 299:797-802) may also be used. General aspects of mammalian cell host system transformations have been described by Axel, supra.
Insect expression systems may also be employed to express the amyloid genes. For example, the baculovirus polyhedrin gene has been employed for high- level expression of heterologous proteins (Smith et al., (1983) Mol Cell Biol 3(12) :2156-2165; Summers et al., "Genetic Engineering of the Genome of the Autographa Californica Nuclear Polyhedrosis Virus", Banbury Report: Genetically Altered Viruses in the Environment, 22:319- 339, Cold Spring Harbor Laboratory, 1985).
D. Generation of Stably Transfected Cell Lines
The amyloid DNA clones expressed in vaccinia can also be used to generate stably transfected cell lines expressing the amyloid proteins. In general, these cell lines are generated by first constructing one of two expression plasmids. In both expression plasmids, the selectable marker is provided by a G418 neomycin expres¬ sion cassette (neo) consisting of the SV40 early promoter, the bacterial kanamycin-resistance gene also containing its own promoter, the SV40 intervening sequence, and the SV40 polyadenylation site from the early region. In the first expression plasmid, the amyloid DNA cloning site is flanked at the 5' end by the human metallothionein gene promoter, pMtlla, modified with an SV40 enhancer, and at the 3' end by the SV40 polyadenylation site from the early region. In the second expression construct, the amyloid DNA cloning site is flanked at the 5' end by a beta-actin promoter, and at the 3' end by a sequence encoding a use¬ ful polyadenylation site, such as that of the SV40 early region or the beta-actin gene.
Each of the vectors described above can be transformed into a mammalian cell line such as, but not limited to, those described in the following examples by either calcium phosphate-DNA coprecipitation or electro- poration. A day later, the cells are subjected to 1 mg/ml G418 to provide pools of G418-resistant colonies. Suc¬ cessful transformants, also having a stable inheritance of the DNA contained in the expression construct, are then plated at low density for purification of clonal isolates. Clonal isolates are then analyzed for maximum production of the amyloid protein of interest and high-producing clones are expanded to serve as stock.
E. Detection Methods for Preamyloid Formation The diagnosis of amyloidosis is established by demonstration of the characteristic emerald-green birefringence of tissue specimens stained with Congo red and examined by polarization microscopy. Congo red stain¬ ing is generally carried out using commercially available diagnostic kits. The isolation and characterization of the A4 protein has allowed specific antibodies to be raised that recognized cerebral amyloid in Alzheimer's disease (Allsop et al (1986) Neurosci Lett 68:252-256). Moreover, Tagliavini et al., (1988) supra, have demonstrated that antibodies can be generated which detect in both Alzheimer's patients and to a lesser extent in non-demented individuals preamyloid deposits, which deposits lack the tinctorial and optical properties of amyloid and are, therefore, undetectable using conventional staining methods employing principally Congo red, but also thioflavin S or silver salts. Standard protocols can be employed for preparing antibodies directed against the amyloid proteins of the invention. Techniques for preparing both polyclonal and monoclonal antibodies are well known in the art. Briefly, polyclonal antibodies are prepared by injecting amyloid protein or synthetic amyloid peptides with an adjuvant into an animal such as rabbits or mice. The amyloid protein may need to be conjugated to a carrier protein such as bovine serum albumin or keyhole limpet hemacyanin using a chemical process which employs carbodiimide, glutaraldehyde, or other cross-linking agents. Alternatively, the protein may be administered without being conjugated to a carrier protein. Vaccinia virus re¬ combinants which are expressing amyloid proteins may also be used to prepare antibodies. The vaccinia virus re¬ combinants are injected into an animal and then the animal is boosted several weeks after the initial immunization. Ten days to two weeks later the animals are bled and antiserum is collected and analyzed for titer. Monoclonal antibodies are commonly prepared by fusing, under appropriate conditions, B-lymphocytes of an animal which is making polyclonal antibodies with an im¬ mortalizing myeloma cell line. The B-lymphocytes can be spleen cells or peripheral blood lymphocytes. Techniques for fusion are also well known in the art, and in general, involve mixing the cells with a fusing agent such as poly¬ ethylene glycol. Successful hybridσma formation is as¬ sessed and selected by standard procedures such as, for example, HAT medium. From among successful hybridomas, those secreting the desired antibody are screened by as¬ saying the culture medium for their presence.
Standard immunological techniques such as ELISA (enzyme-linked immunσassay) , RIA (radioimmunoassay) , IFA (immunofluorescence assay) and Western blot analysis, which are well known in the art, can be employed for diagnostic screening for amyloid expression. A vast literature now exists with respect to various modifica¬ tions of the basic assay principle, which is simply that there must be a specific association between target analyte and antibody, which association is detectable qualitatively and/or quantitatively. Fluorescent, enzymatic, or radioactive labels are generally used.
One typical arrangement utilizes competition, between labeled antigen (e.g. amyloid protein) and the analyte, for the antibody, followed by physical separation of bound and unbound fractions. Analyte competes for the binding of the labeled antigen; hence more label will remain in the unbound fraction when larger amounts of analyte are present. In this competitive-binding type assay, the sample is incubated with a known titer of labeled amyloid protein and amyloid protein antibody. Antibody-protein complex is then separated from uncomplexed reagents using known techniques and the amount of label in the co plexed material is measured, e.g. by gamma counting in the case of radioimmunoassay or photo- metrically in the case of enzyme immunoassay. The amount of amyloid protein in the sample, if any, is determined by comparing the measured amount of label with a standard curve.
Other embodiments of this basic principle include use of labeled antibodies per se, sandwich assays involving a three-way complex between analyte, anti- analyte antibody, and anti-antibody wherein one of the components contains a label, and separation of bound and unbound fractions using an immunosorbent. Agglutination assays which result in visible precipitates are also available (Limet et al. , (1982) J Clin Che Clin Biochem 2 :142-147) .
F. Screening Assay The present assay provides one of the first steps in addressing the question whether preamyloid corti- cal deposits herald organic dementia. The concomitant appearance of preamyloid deposits and senile plaques sug¬ gests that preamyloid deposits may evolve into senile plaques. Down's syndrome is the one known disease closely related to the proposed etiology of AD. As from their twenties onward, Down's patients develop the full spectrum of Alzheimer's changes, i.e., NFTs, congophilic angiopathy and senile plaques. As reported in Giacione et al. , (1989) Neurosci Letts 97:232-238, a time-related analysis of preamyloid deposits and senile plaque distribution showed an age-dependent, inverse correlation between extracellular preamyloid deposits and senile plaque in Down's patients. While a similar, time-dependent study with Alzheimer's patients cannot be conducted, it is expected that a corresponding pattern (preamyloid turning to senile plaque deposits) would be found. Therapeutic agents which interfere with this process promise the development of successful therapeutic regimens for Alzheimer's disease.
In the practice of the method of the invention, the expression of the amyloid protein is initiated by culturing the transformed cell line under conditions which are suitable for cell growth and expression of the amyloid protein. In this method, high level expression of the protein is preferred. In one embodiment of the invention, a CHO cell line transformed with a beta-actin vector comprising the DNA encoding the A42 or A99 amyloid protein is grown in a mammalian culture medium such as, for example, a 1:1 mixture of F12 medium and DME medium with 10% fetal calf serum for 5-72 hr at 37°C. Transfected viral monolayers are selected and plaque purified, and stocks of amyloid-vaccinia recombinant viruses are prepared. The formation of the preamyloid aggregates can be monitored by standard immunocytochemical methods using, for example, beta-amyloid primary antibodies which are detected using a secondary, labeled anti-antibody. If one is interested in testing whether the compound of interest can inhibit preamyloid formation, the compound is introduced to the culture medium before monitoring for preamyloid aggregation. Alternatively, the compound is introduced to the culture medium after preamyloid forma¬ tion has been established and this reaction mixture is monitored to see whether the compound induces amyloid resorption.
Potential therapeutic compounds for use in the present invention include, for example, amyloid-fibril denaturing agents such as dimethyl sulfoxide, and cytotoxic agents such as colchicine and chlorambucil. The efficacy of these agents may be monitored through observa¬ tion of reduced antibody binding to the amyloid deposit. Reduction in such binding is indicative of reduced preamyloid deposition. Alternatively, preamyloid formation in the host cell may trigger other cellular events which could be employed as markers unrelated to the etiology of Alzheimer's disease, but correlative with the presence of preamyloid deposits. For example, an increase in the level of certain enzymes, specifically proteases, may be measured in lieu of the preamyloid deposition. Typically, an increase in the concentration levels of these enzymes is observed when cultured cells are subjected to stress.
The present invention also encompasses kits suitable for the above diagnostic or screening methods. These kits contain the appropriate reagents and are constructed by packaging the appropriate materials, including the preamyloid protein aggregates immobilized on a solid support with labeled antibodies in suitable containers, along with any other reagents (e.g., wash solutions, enzyme substrate, anti-amyloid antibodies) or other materials required for the conduct of the assay. The reagents are usually premeasured for ease of use. An optional component of the kit is a set of instructions describing any of the available immunoassay methods. For example, a kit for a direct assay can comprise preamyloid proteins aggregates immobilized on a solid immunoassay support and a container comprising labeled antibody to the amyloid protein, as well as the other reagents mentioned above.
The following examples are designed to elucidate the teachings of the present invention, and in no way limit the scope of the invention. Most of the techniques which are used to transform cells, construct vectors perform immunoassays, and the like are widely practiced in the art, and most practitioners are familiar with the standard resource materials which describe specific condi¬ tions and procedures. The examples are written in observation of such knowledge and incorporate by reference procedures considered conventional in the art.
EXAMPLE 1
Description of Amyloid Plaque Core DNA Constructs
The following examples describe the expression vectors containing the 42 amino acid plaque core region (A42), and the 42 amino acid plaque core region including the 57 amino acid adjacent carboxy-terminal region of the beta-amyloid precursor protein (A99). Alternative constructs for the A42 and A99 constructs were prepared which included a 17 amino acid amyloid signal sequence. As these constructs did not express the amyloid protein well, further experimentation with these vectors was not performed.
Recombinant vaccinia viruses bearing amyloid DNAs encoding each of the two amyloid constructs (W:A42 and W:A99) were generated by standard methods as reviewed by Mackett and Smith in (1986) J Gen Virol 67:2067-2082, which is incorporated herein by reference. FIG. 1 il¬ lustrates the various amyloid expression constructs, all of which were modified at the 5' end to satisfy the clon¬ ing constraints of the vaccinia Pll promoter in the pUVl vector. Specifics for each construct are as follows:
A. W:A42:
The A42-encoding sequence (nucleotides 2080 to 2205, numbered in accordance with the 751 amyloid pre- cursor sequence) was synthesized as a 145 basepair (bp) EcoRI-BamHI oligomer, provided below, containing the ap¬ propriate TGA stop codon and an amino-terminal Asn-Ser adaptor sequence:
5' AAT TCC GAT GCA GAA TTC CGA CAT GAC TCA
GGA TAT GAA GTT CAT CAT CAA AAA TTG GTG TTC TTT GCA GAA GAT GTG GGT TCA AAC AAA GGT GCA ATC ATT GGA CTC ATC GTG GGC GGT GTT GTC ATA GCG TGA TCT AGA TGA G 3'
The synthetic fragment was ligated to EcoRI- and BamHI-digested pGeml (Promega-Biotec) , deriving pGemA42. The EcoRI-BamHI fragment of pGemA42 was subsequently isolated and ligated into the EcoRI-BamHI site of pUVl deriving pUVl:A42.
The Xbal-Sall fragment of pUVl:A42 (287bp) was further subcloned into mpl8 for sequence confirmation.
B. W:A99: The DNA encoding the amyloid protein for the pUVl-A99 constructs was derived from 4T4B, a plasmid en¬ coding the 751 amino acid precursor protein. The construction of plasmid 4T4B is described in Example 3 of PCT/US87/02953, owned by the same assignee. The relevant portions of this publication are incorporated herein by reference. The 590 bp Ddel-PvuII fragment of plasmid 4T4B was isolated from the carboxy-terminal 1 kilobase (kb) EcoRI fragment of 4T4B and ligated with a 27 bp EcoRI-Ddel adaptor sequence and cloned into the EcoRI- and Smal- digested pUVl, deriving pUVl:A99. The 761 bp Xbal-Sall fragment of pUVl:A99 was further subcloned into the Xbal-Sall vector fragment of mpl8 and pGem2. Sequence data confirmed the predicted sequence.
EXAMPLE 2
Expression of Amyloid Proteins
The vaccinia insertion vectors described in Example 1 were used to generate amyloid-vaccinia re- combinant viruses as follows.
A. Preparation of Amyloid-Vaccinia Virus Recombinants
Confluent monolayers of CV-1 cells in 60 mm dishes were infected with vaccinia virus (Wyeth strain) at a multiplicity of infection (moi) of 0.05 pfu/cell. At 0.5 hr post-infection, the cells were transfected with a calcium phosphate precipitate of 10 ug insertion plasmid DNA and 0.5 ug wild-type vaccinia virus DNA. Cells were fed with complete medium and incubated at 37°C for two days. Monolayers were collected and TK- vaccinia viruses were selected on TK-143 cells in the presence of 5- bromodeoxyuridine (BudR) at 25 ug/ l. At 48 hr after infection, monolayers were overlaid with 1% agarose containing 300 ug/ml 5-bromo-4-chloro-3-indolyl-B-D- galactopyranoside (Xgal). At 4-6 hr, blue plaques were picked and further purified by two additional rounds of plaque purification in the presence of BudR and Xgal. Stocks of the amyloid-vaccinia recombinant viruses were prepared in TK-142 or CV-1 cells. Recombinant viral DNA was prepared from each stock and was shown by Southern blot analysis to contain the appropriate amyloid DNA insert and to be free of contamination with wild-type or spontaneous TK- vaccinia.
B. Identification of Amyloid-specific Polypeptides Produced By Vaccinia Virus Recombinants
Characterization of the CV-1 expressed W:A42 and W:A99 amyloid proteins was carried out employing immunoprecipitation and polyacrylamide gel analysis of
35 S-methionine-labeled infected cell protein using anti- bodies directed against the carboxy-terminal region of the amyloid precursor.
The beta-amyloid antibodies were generated from synthetic peptides. The synthetic peptides were prepared using solid phase synthesis according to standard protocols. Purification of the crude peptides was ac¬ complished by desalting with gel filtration followed by ion-exchange chromatography and preparative reverse-phase liquid chromatography. Each peptide was fully character¬ ized by amino acid composition and sequence analysis. COOH-CORE corresponds to amino acids 653-
680(DAEFRHDSGYEVHHQKLVFFAEDVGSSA) (the carboxy-terminal two amino acids were taken from the amino acid sequence of Masters et al., (1985) Proc Natl Acad Sci 82:4245-4249 and are different in the deduced translation of the A4 cDNA of Ponte et al., supra. C00H-B2 and C00H-C2 correspond to amino acids 736-751(NGYENPTYKFFEQMQN) , C00H-B3 and C00H-C3 correspond to amino acids 705-719(KKKQYTSIHHGWEV) and C00H-C5 corresponds to amino acids 729- 742(HLSKMQQNGYENPT) . Reference for the numbering of peptides along the topology of the A4 precursor is from Ponte et al. , supra. New Zealand white rabbits were im¬ munized intradermally with 500 ug of peptide conjugated to keyhole limpet hemocyanin. The rabbits were first bled at 4 weeks and 1 week later the rabbits were boosted with 250 ug conjugated peptide. Subsequent bleeds were done at 3 week intervals with boosts following 1 week later. All animals were treated in accordance with institutional guidelines . Antibody titers against the appropriate peptide were determined by enzyme-linked immunosorbent assays coupled with horseradish peroxidase and found to be 7.4xl04, 2.7xl05, IxlO5, 9.1xl06, 8.2xl05, and 2.5xl05 for COOH-CORE, C00H-B2, C00H-C2, C00H-B3, C00H-C3, and C00H- C5, respectively.
Antibodies to 9523 correspond to amino acids 673-685(AEDVGSKNGAIIG) and 9524 correspond to amino acids 701-712(LVMLKKKQYTSI) . Antibodies to these two peptides were generated by coinjecting New Zealand white rabbits each with 200 ug methylated bovine serum albumin (PBS) plus 200 ug of the respective synthetic peptide in PBS. Rabbits were boosted one, two and three weeks following primary inoculation with identical amounts of peptide.
Serum samples were taken at week 6 and titered against
4 APCP synthetic peptide. Titers achieved were 1.5x10 for
9523 and 4xl05 for 9524.
CV-1 cells were infected with W:99 at a multiplicity of infection of one. 35S-methionine (250 uCi/ml) was added at 20 hr post infection for 4 hr. Cell
7 lysates were prepared and aliquots containing 10 cpm were immunoprecipitated with amyloid-specific antisera (C00H-
B3, COOH-C5 and COOH-CORE) or normal rabbit serum and protein A. Immunoprecipitates of 35S-methionine cell lysates were analyzed on denaturing 20% SDS-polyacrylamide gels. As shown in FIG. 2, high levels of expression and stability of the A99 protein generated by W:A99 was demonstrated. The control sera (normal, nonimmune rabbit sera) did not display reactivity with the W:A99 protein product. The W:A99 amyloid core protein migrated as a broad band spanning approximately 11.5-17 kd molecular weight. In addition, higher molecular weight forms of the A99 protein were clearly observed. The expression product of W:A99 demonstrated high level expression of the 99 amino acid core protein and showed evidence of self-aggregation as well as ag¬ gregation with other proteins or self-aggregation combined with proteolysis since multi ers of A99 did not always occur in integers of 11.5-17 kd.
EXAMPLE 3 Staining of A42 and A99 Expressing Cells
Two human, SK-N-MC (ATCC # HTB10) and IMR-32, (ATCC # CCL127) and one rat, PC-12 (Green and Tischler, (1976) Proc Natl Acad Sci USA 73:2424-2428) neuronal cell lines were examined for their ability to permit efficient infection with the W:A42 and W:A99 recombinant viruses. All cell lines were documented as permissive hosts for vaccinia virus replication by infecting cells with a given amount (moi=2) of vaccinia virus of known titer. The infected cells were harvested 20 hours after infection, disrupted by freeze-thaw, and then titered. The yield was compared to the input viral units and if 20-100 fold increase results, the host cell was considered permissible for vaccinia replication.
These neuronal lines and the CV-1 cell line were employed for amyloid staining studies. The culture medium for each host was as follows:
CV-1: The medium was Eagle MEM supplemented with 10% FBS, penicillin, streptomycin and L- Gin-
SK-N-MC: Eagle MEM supplemented with 10% FBS, non- essential amino acids, penicillin, streptomycin and L-Gln. PC-12: DMEM21, 5% DHS, 5% DFBS and L-Gln; and
IMR-32: Eagle MEM (Hank's BSS) and 10% deltaFBS plus nonessential amino acids, penicillin, streptomycin and L-Gln.
Each cell line was grown to confluency on a microscope slide divided into 4 individual chambers (Lab Tech) . One chamber was mock infected, the second infected with a control recombinant virus lacking A4 sequences
(W:C0NT), the third chamber infected with W:A99, and the fourth chamber infected with W:A42. This is an internally controlled method since each slide was manipulated as a single unit. Viral infections were carried out at a moi from
5 to 20 viral plaque forming units (pfu) per cell and were harvested for staining at approximately 20 hours post infection. Slides prepared for immunocytochemistry were fixed with 4% paraformaldehyde and permeabilized with 0.2% Triton X-100 prior to treatment with primary and rhodamine-conjugated second antibodies (Capell Labs). Briefly, after permeabilization, cells were washed with PBS containing 0.2% gelatin. 100 ul of primary amyloid antibody (diluted 1/200 with PBS plus 0.2% gelatin) was incubated on the cells at 37°C for 30 minutes. Cells were washed for 10 minutes in PBS and 0.2% gelatin, then incubated at 37°C for 20 minutes with a 1/200 dilution (in PBS and gelatin) of secondary antibody (goat-anti-rabbit) tagged with Rhodamine. Cells were washed for 10 min in PBS and gelatin, then mounted for visualization in a fluorescent microscope. Antibodies used with success included 9523, 9524, B3 and C5. CORE antibodies were not assessed. Alternatively, the slides were fixed in 4% paraformaldehyde then stained with Thioflavin S or Congo red, and counterstained with hematoxylin according to directions in commercial kits (Sigma) . IMR-32 and PC-12 cells presented some technical difficulties and thus further investigation with these cell lines was terminated. The IMR-32 cells did not adhere well to the microscope slides, which could be al- leviated by pretreatment with laminin, and, moreover, the IMR-32 cells did not tolerate the serum-free conditions during the infections. PC-12 cells showed high background immunostaining, hence, differences between experimental and control samples were not dramatic. FIG. 3 shows fluorescent photomicrographs of CV-
1 cells stained with 1/200 dilutions of the core domain antibodies 9523 antibodies. Specific and robust staining was seen in only the W:A99 and W:42 infected cells. W:99 specific staining, but not W:42 staining, was seen with the B3 antibody as would be anticipated since this region is not included in the W:A42 construct (results not shown) . Faint punctate staining was observed for both antibodies on all cells presumably due to endogenous A4 precursor expression. The W:A99 and W:A42 infected cells displayed strong reactivity in the form of large deposit-like structures which are cell associated. The deposit-like structures are probably not cell debris from the viral cytopathicity since they are not seen in the W:C0NT cells and their immunoreactivity could be eliminated by preadsorption of the antisera with the synthetic peptide used to raise the serum.
The possible potentiating effect of aluminum on deposit formation was investigated by pretreating the cells with 50 mM A1C1-.- Aluminum might be considered a "cofactor" in the pathology of amyloid formation since it is present in plaques. However, no obvious qualitative difference in the degree of deposit formation between cultures treated and untreated with aluminum was found. It seems relevant that several researchers investigating A4 core domain immunoreactivity in brains of Alzheimer's victims describe similar structures as those in FIG. 3. Each group reported finding significant amounts of specifically stained spherical, granular deposits which were considerably smaller than pre-plaques and plaques (Davies et al., (1988) Neuroloq 38:1688-1693; Ikeda et al., (1989) Lab Invest 60:113-122; Tagliavini et al., (1988) supra; Tate-Ostroff et al., (1989) Proc Natl Acad Sci 86:745-749). All research groups independently propose that the observed small granular deposits are the very early stages of amyloid plaque development. The structures observed in our cell culture system are analogous to those seen in the Alzheimer's diseased brain. It was noted by these investigators that the granular deposits could be occasionally detected with silver stain but not with Congo red. Because the Alzheimer's granular deposits were highly reactive with A4 antisera but were not easily reacted with stains capable of recognizing the tinctorial properties of amyloid, the structures were termed "preamyloid" deposits.
EXAMPLE 4
Establishment of Stable Cell Lines
A number of constructs expressing the beta- amyloid core protein were constructed using a derivative of the beta-actin expression/selection vector designated pHbetaAPr-1-neo. This vector, illustrated in FIG. 4, is a combination of the following elements: a) bp 1-4300 is the 4.3 kb EcoRI-AluI fragment from the human beta-actin gene isolate pl4Tbeta-17 (Leavitt et al. , (1984) Mol Cell Biol 4:1961-1969). For sequencing details of the promoter see Ng et al. , (1985) Mol Cell Biol 5:2720-2732. The cap site, 5' untranslated region and IVS 1 positions are indicated in FIG. 4. There is no ATG codon present in the 5' UT nor in the polylinker region from the 3' splice site to the BamHI site; b) bp 4300-4320 is in part derived from pSP64 polylinker (Melton et al., (1984) Nuc Acids Res 12:7035- 7056); c) bp 4320-6600 is derived from pcDVl (Okayama & Berg, (1983) Mol Cell Biol 3:280-289); and d) bp 6600-10000 is the PvuII-EcoRI fragment from pSV-neo (Southern & Berg, (1982) J Mol App Genet 1:327-341) containing the bacterial neo ycin gene linked to the SV40 origin plus early promoter. The direction of transcription is as indicated in FIG. 4. This vector was altered by deleting the EcoRI site and adding a new EcoRI site within the polylinker 3' to the Sail site and 5' to the HindiII site. This modified vector is designated pAX- neo. Beta-actin A42 was constructed by excising the EcoRI-BamHI 145 bp fragment from pGEM-A42, adding a Sall- EcoRI adaptor sequence (5'-TCG ACA TGG ATG CAC AAT TA-3') and cloning into the pAX-neo expression vector at the Sall-and BamHI sites. The beta-actin A99 plasmid was constructed by excising the 670 bp EcoRI-Hindlll fragment of pGEM_-A99, adding the above-described Sall-EcoRI adap¬ tor sequence and cloning into the pAX-neo vector at the Sail and HindiII sites.
Each construct was introduced into CHO cells by the calcium phosphate precipitation method using 7 ug of each DNA per 10 cells, and a resistant population was selected with G418-neomycin. The efficiency of
3 transfection for the A99 or A42 constructs was over 10 for 10 cells and pools of cells transfected with either beta-actin A99 or with beta-actin A42 were selected using G418-neomycin resistance (500 ug/ml).
Cell lysates from these pools are prepared and analyzed by immunoprecipitation of the A4 proteins as well as by Western blotting. High expressing clones are then selected and assayed for "preamyloid" deposits using the immunocyto-staining procedures described in Example 3. EXAMPLE 5 Assay for Preamyloid Deposition
Cells infected with W:99 or W:42 which are capable of forming amyloid deposits are plated in a 96- well microtiter plate. To make the appropriate dilutions and additions, an automated pipetter is used to introduce the drug to be tested to the cells. A range of concentrations of the drug is incubated in a tissue culture incubator (or preincubated) with the cells at 37 C for a predetermined time period, or alternatively, for 3 to 72 hours.
Following incubation, the culture media is removed, and the cells are prepared for preamyloid measurement as follows. The cells are fixed for immunocytochemical staining with amyloid antibodies. The primary antibodies are introduced followed by incubation with labeled, secondary anti-antibodies and the level of binding between the primary and secondary antibodies is measured using an ELISA plate reader to record the optical density of the labeled antibody. A smaller optical density reading as compared to a control sample of cells grown in the absence of the test drug is indicative of that drug's ability to inhibit amyloid deposition. This procedure may be modified to permit detection of preamyloid dissolution using a correlative enzyme marker.
It will be apparent to those skilled in the art that various modifications and variations can be made in the method of the present invention without departing from the scope or spirit of the invention. Thus, it is intended that the claims cover the modifications and variations of the invention.

Claims

What is claimed is:
1. A method of screening agents capable of intervention in Alzheimer's disease amyloidosis compris¬ ing: a) culturing a cell line capable of expressing a gene encoding beta-amyloid protein under conditions suitable to produce the beta-amyloid protein as an insoluble, preamyloid aggregate; b) combining a known quantity of the agent to be tested to the cell culture; and c) monitoring the combination to determine whether preamyloid aggregate formation is reduced.
2. The method of claim 1 wherein the beta- amyloid gene encodes a protein comprising the amyloid plaque core domain.
3. The method of claim 1 wherein the beta- amyloid gene encodes a protein comprising the amyloid plaque core and the carboxy-terminal domains.
4. The method of claim 3 wherein the beta- amyloid gene encodes the following polypeptide:
10 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin
20 30
Lys Leu Val Phe Phe Ala Glu Asp Val Gly Ser Asn Lys Gly Ala 40 (42)
He He Gly Leu Met Val Gly Gly Val Val He Ala Thr Val He
50 60
Val He Thr Leu Val Met Leu Lys Lys Lys Gin Tyr Thr Ser He
70 His His Gly Val Val Glu Val Asp Ala Ala Val Thr Pro Glu Glu
80 90
Arg His Leu Ser Lys Met Gin Gin Asn Gly Tyr Glu Asn Pro Thr
(99) Tyr Lys Phe Phe Glu Gin Met Gin Asn.
5. The method of claim 1 wherein the preamyloid is derived from stable cell lines infected with recombinant vaccinia virus comprising the beta-amyloid gene.
6. The method of claim 1 wherein the cell lines are derived from mammalian host cells.
7. The method of claim 1 wherein the agent to be tested is introduced during the growth phase of the cell culture to determine whether the agent inhibits preamyloid plaque formation.
8. The method of claim 1 wherein the agent to be tested is added to the amyloid aggregate to determine whether the agent dissolves preamyloid plaque formation.
9- An immunological reagent capable of detecting preamyloid aggregate formation.
10. The immunological reagent of claim 9 which is a monoclonal antibody. -Si¬
11. The immunological reagent of claim 9 which is a polyclonal antibody.
12. A kit for an immunoassay to screen compounds capable of chemical intervention in amyloidosis of
Alzheimer's disease comprising: a predetermined amount of preamyloid aggregate specific for Alzheimer's disease; and a predetermined amount of labeled antibody to said preamyloid aggregate.
13. The kit according to claim 12 wherein said label is a component of an enzymatic reaction.
14. The kit according to claim 12 wherein said preamyloid aggregate is immobilized on a solid immunoassay support.
PCT/US1990/005155 1989-09-18 1990-09-12 Assays and reagents for amyloid deposition WO1991004339A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA002065404A CA2065404C (en) 1989-09-18 1990-09-12 Assays and reagents for amyloid deposition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US40876789A 1989-09-18 1989-09-18
US408,767 1989-09-18

Publications (1)

Publication Number Publication Date
WO1991004339A1 true WO1991004339A1 (en) 1991-04-04

Family

ID=23617679

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/005155 WO1991004339A1 (en) 1989-09-18 1990-09-12 Assays and reagents for amyloid deposition

Country Status (5)

Country Link
EP (1) EP0493470A4 (en)
JP (1) JPH05502368A (en)
AU (1) AU641434B2 (en)
CA (1) CA2065404C (en)
WO (1) WO1991004339A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997048983A1 (en) * 1996-06-18 1997-12-24 Athena Neurosciences, Inc. SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)
US5714471A (en) * 1995-01-06 1998-02-03 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
US5837449A (en) * 1991-12-24 1998-11-17 Isis Pharmaceuticals, Inc. Compositions and methods for modulating β-amyloid
US5863902A (en) * 1995-01-06 1999-01-26 Sibia Neurosciences, Inc. Methods of treating neurodegenerative disorders using protease inhibitors
WO1999029891A1 (en) * 1997-12-09 1999-06-17 Arch Development Corporation Methods for identifying factors controlling amyloid protein aggregation
WO2002065136A2 (en) * 2001-02-15 2002-08-22 University Of Chicago Yeast screens for agents affecting protein folding
EP1793001A3 (en) * 2001-02-15 2007-06-20 The University of Chicago Yeast screens for agents affecting protein folding

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4264729A (en) * 1977-09-19 1981-04-28 Mirko Beljanski Method and reagent for detecting cancerigenic and anticancerous substances
US4666829A (en) * 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
EP0304013A2 (en) * 1987-08-15 1989-02-22 Asahi Kasei Kogyo Kabushiki Kaisha A novel senile amyloid precursor protein and an antibody specific for the same
WO1989007657A1 (en) * 1988-02-10 1989-08-24 The Children's Medical Center Corporation Amyloid protein precursors, genetic probes, antibodies, and methods of use
US4912206A (en) * 1987-02-26 1990-03-27 The United States Of America As Represented By The Department Of Health And Human Services CDNA clone encoding brain amyloid of alzheimer's disease
US4919915A (en) * 1987-03-03 1990-04-24 Paul Averback Method for detecting the ability to prevent red-to-green congophilic birefringence

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0332640A1 (en) * 1986-11-17 1989-09-20 California Biotechnology, Inc. Recombinant alzheimer's amyloid protein
CA1339014C (en) * 1987-10-08 1997-03-25 Ronald E. Majocha Antibodies to a4 amyloid peptide
AU3055889A (en) * 1988-01-13 1989-08-11 Mclean Hospital Corporation, The Genetic sequences coding for alzheimer amyloid from brain

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4264729A (en) * 1977-09-19 1981-04-28 Mirko Beljanski Method and reagent for detecting cancerigenic and anticancerous substances
US4666829A (en) * 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
US4912206A (en) * 1987-02-26 1990-03-27 The United States Of America As Represented By The Department Of Health And Human Services CDNA clone encoding brain amyloid of alzheimer's disease
US4919915A (en) * 1987-03-03 1990-04-24 Paul Averback Method for detecting the ability to prevent red-to-green congophilic birefringence
EP0304013A2 (en) * 1987-08-15 1989-02-22 Asahi Kasei Kogyo Kabushiki Kaisha A novel senile amyloid precursor protein and an antibody specific for the same
WO1989007657A1 (en) * 1988-02-10 1989-08-24 The Children's Medical Center Corporation Amyloid protein precursors, genetic probes, antibodies, and methods of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0493470A4 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5837449A (en) * 1991-12-24 1998-11-17 Isis Pharmaceuticals, Inc. Compositions and methods for modulating β-amyloid
US6177246B1 (en) 1991-12-24 2001-01-23 Isis Pharmaceuticals, Inc. Compositions and methods for modulating β-amyloid
US7993627B2 (en) 1992-07-10 2011-08-09 Elan Pharmaceuticals, Inc. Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal
US6015879A (en) * 1995-01-06 2000-01-18 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
US5863902A (en) * 1995-01-06 1999-01-26 Sibia Neurosciences, Inc. Methods of treating neurodegenerative disorders using protease inhibitors
US5872101A (en) * 1995-01-06 1999-02-16 Sibia Neurosciences, Inc. Methods of treating neurodegenerative disorders using protease inhibitors
US5962419A (en) * 1995-01-06 1999-10-05 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
US5969100A (en) * 1995-01-06 1999-10-19 Sibia Neurosciences, Inc. Peptide, peptide analog and amino acid analog protease inhibitors
US5804560A (en) * 1995-01-06 1998-09-08 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
US6017887A (en) * 1995-01-06 2000-01-25 Sibia Neurosciences, Inc. Peptide, peptide analog and amino acid analog protease inhibitors
US6051684A (en) * 1995-01-06 2000-04-18 Sibia Neurosciences Inc. Methods of treating neurodegenerative disorders using protease inhibitors
US6153171A (en) * 1995-01-06 2000-11-28 Sibia Neurosciences, Inc. Methods for identifying compounds effective for treating neurodegenerative disorders and for monitoring the therapeutic intervention therefor
US5714471A (en) * 1995-01-06 1998-02-03 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
WO1997048983A1 (en) * 1996-06-18 1997-12-24 Athena Neurosciences, Inc. SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)
WO1999029891A1 (en) * 1997-12-09 1999-06-17 Arch Development Corporation Methods for identifying factors controlling amyloid protein aggregation
US7799535B1 (en) 1997-12-09 2010-09-21 Arch Development Corporation Methods for identifying factors that control the folding of amyloid proteins of diverse origin
WO2002065136A3 (en) * 2001-02-15 2003-12-11 Univ Chicago Yeast screens for agents affecting protein folding
US7045290B2 (en) 2001-02-15 2006-05-16 The University Of Chicago Yeast screens for treatment of human disease
EP1793001A3 (en) * 2001-02-15 2007-06-20 The University of Chicago Yeast screens for agents affecting protein folding
AU2002250102B2 (en) * 2001-02-15 2007-07-12 University Of Chicago Yeast screens for agents affecting protein folding
JP2008259509A (en) * 2001-02-15 2008-10-30 Univ Of Chicago Yeast screening of agent affecting protein folding
AU2007221933B2 (en) * 2001-02-15 2010-12-16 University Of Chicago Yeast screens for agents affecting protein folding
WO2002065136A2 (en) * 2001-02-15 2002-08-22 University Of Chicago Yeast screens for agents affecting protein folding
US8039209B2 (en) 2001-02-15 2011-10-18 The University Of Chicago Yeast screens for treatment of human disease
AU2011200744B2 (en) * 2001-02-15 2013-04-04 University Of Chicago Yeast screens for agents affecting protein folding
US9518284B2 (en) 2001-02-15 2016-12-13 The University Of Chicago Yeast screens for treatment of human disease

Also Published As

Publication number Publication date
AU641434B2 (en) 1993-09-23
CA2065404C (en) 2002-12-24
EP0493470A1 (en) 1992-07-08
AU6431190A (en) 1991-04-18
EP0493470A4 (en) 1992-11-25
CA2065404A1 (en) 1991-03-19
JPH05502368A (en) 1993-04-28

Similar Documents

Publication Publication Date Title
US5221607A (en) Assays and reagents for amyloid deposition
Otvos Jr et al. Monoclonal antibody PHF‐1 recognizes tau protein phosphorylated at serine residues 396 and 404
US6610493B1 (en) Screening compounds for the ability to alter the production of amyloid-β peptide
EP0736106B2 (en) Methods of screening for beta-amyloid peptide production inhibitors
CA2205359C (en) Methods for aiding in the diagnosis of alzheimer's disease by measuring amyloid-beta peptide (x->=41) and tau
CA2118243C (en) Methods and compositions for monitoring cellular processing of .beta.-amyloid precursor protein
US5593846A (en) Methods for the detection of soluble β-amyloid peptide
US5837672A (en) Methods and compositions for the detection of soluble β-amyloid peptide
US7674599B2 (en) Methods of using antibodies to detect alpha-synuclein in fluid samples
US5605811A (en) Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US7479372B2 (en) Beta-secretase substrates and uses thereof
US5733734A (en) Method of screening for Alzheimer's disease or disease associated with the accumulation of paired helical filaments
WO1997048983A9 (en) SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)
AU641434B2 (en) Assays and reagents for amyloid deposition
WO1994013315A1 (en) A novel eukaryotic transcription protein: host cell factor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

WWE Wipo information: entry into national phase

Ref document number: 2065404

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1990914284

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1990914284

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1990914284

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1990914284

Country of ref document: EP