AU720446B2 - Apolipoprotein E2 and treatment of Alzheimer's disease - Google Patents

Apolipoprotein E2 and treatment of Alzheimer's disease Download PDF

Info

Publication number
AU720446B2
AU720446B2 AU75526/96A AU7552696A AU720446B2 AU 720446 B2 AU720446 B2 AU 720446B2 AU 75526/96 A AU75526/96 A AU 75526/96A AU 7552696 A AU7552696 A AU 7552696A AU 720446 B2 AU720446 B2 AU 720446B2
Authority
AU
Australia
Prior art keywords
apolipoprotein
apoe2
apoe
recombinant
apoe3
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU75526/96A
Other versions
AU7552696A (en
Inventor
Sergie B. Aleshkov
Vassilis I. Zannis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boston University
Kos Pharmaceuticals Inc
Original Assignee
Boston University
Kos Pharmaceuticals 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 Boston University, Kos Pharmaceuticals Inc filed Critical Boston University
Publication of AU7552696A publication Critical patent/AU7552696A/en
Application granted granted Critical
Publication of AU720446B2 publication Critical patent/AU720446B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • 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
    • C07K14/775Apolipopeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Description

I
WO 97/16458 PCT/US96/17629 1 APOLIPOPROTEIN E2 AND TREATMENT OF ALZHEIMER'S DISEASE Field of the Invention This invention relates to recombinant DNA, protein purification, and Alzheimer's disease.
Background of the Invention Alzheimer's disease has two major neuropathological characteristics. The first is the presence of intraneuronal neurofibrillary tangles, the major protein component of which is hyperphosphorylated tau, a microtubule-associated protein. Electron microscopy shows neurofibrillary tangles to comprise paired helical filaments (Selkoe (1991) Neuron 6:487-498).
The second major characteristic is the presence of extracellular amyloid deposits in the brain. Amyloid deposits consist of polymers of the amyloid peptide Af, which is proteolytically derived from amyloid precursor protein Apolipoprotein E ("apoE") is a 34.2 kDa protein synthesized by the liver and various peripheral tissues, including kidney, adrenal gland, astrocytes and reticuloendothelial cells. ApoE is synthesized as precursor ("preapoE") with an 18-amino acid signal peptide. After the intracellular cleavage of the signal peptide, apoE is glycosylated with carbohydrate chains containing sialic acid and secreted as sialo apoE. It is subsequently desialated in plasma. For a discussion of apoE biosynthesis processing and other properties, see: Zannis et al. (1993) Adv. Hum. Genet. 21:145-319; Zannis et al. (1984) J. Biol. Chem. 259:5495-5499; and Zannis et al. (1986) J. Biol. Chem. 261:13415-13421.
WO 97/16458 PCT/US96/17629 2 Following synthesis, apoE is incorporated into lipoproteins and directs their catabolism, following recognition and binding by cell surface receptors. ApoE can be isolated from plasma and culture media of HepG-2 cells as a component of lipoprotein particles.
There are three common alleles that encode apoE in humans. The three alleles designated E4, e3, and E2 give rise to three homozygous phenotypes E4/E4, E3/E3, and E2/E2) and three heterozygous phenotypes E4/E3, E3/E2, and E4/E2)(Zannis and Breslow (1981) Biochemistry 20:1033-1041; Zannis et al. (1981) Am. J.
Hum. Genet. 33:11-24). The three different human apoE isoproteins, apoE4, apoE3, and apoE2, result from mutations at amino residues 112 and 158. ApoE4 contains Arg at position 112 and Arg at position 158. ApoE3 contains Cys at position 112, and Arg at position 158.
ApoE2 contains Cys at positions 112 and 158.
Mutations in apoE which affect its recognition by the LDL receptor are associated with type III hyperlipoproteinemia and premature atherosclerosis. See, Goldstein et al. (1983) in: The Metabolic Basis of Inherited Disease (Stanbury et al., eds.), McGraw-Hill, New York, pp. 672-712. The apoE mutation which results in the apoE4 phenotype occurs with increased frequency in patients with late onset of Alzheimer's disease (Corder et al. (1993) Science 262:921-923).
ApoE and Alzheimer's Disease ApoE synthesis is increased following injury and is implicated in the growth and repair of the nervous system during development or after injury (Boyles et al.
(1989) J. Clin. Invest. 83:1015-1031). In the peripheral nervous system, the synthesis of apoE increases 250-350 fold within 3 weeks of sciatic nerve crush, and apoE constitutes 5% of the total soluble extracellular 3 protein. Recently, it has been shown that apoE is found in the brain lesions of AD patients and that the apoE4 phenotype predominates in patients with late onset familial AD (Corder et al. (1993) Science 262:921-923).
It has been suggested that apoE is a risk factor and contributes to the pathogenesis of the disease. Two hypotheses have been advanced to explain the involvement of apoE in AD. The first hypothesis implies extracellular interaction of apoE with the 42 amino acid long amyloid peptide P(AP). Initial observations indicated that apoE from cerebrospinal fluid binds with high affinity to AP (Wisniewski et al. (1993) Biochem. Biophys. Res. Commun.
192:359-365; Strittmatter et al. (1993) Proc. Natl. Acad.
Sci. USA 90:1977-1981). In vitro experiments have shown that apoE can form a complex with AP peptide which is stable following boiling for 5 min at 2% SDS. Formation of stable complexes of apoE4 with AP is fast, compared to formation of complexes of apoE3 with A3.
:I Electron microscopy studies showed that a 20 combination of apoE4 with AP resulted in the formation of monofibrils 7 nm in diameter. The matrix of monofibrils formed with apoE4 was much denser than that with apoE3 ::::(Sanan et al. (1994) J. Clin. Invest. 94:860-869).
A second hypothesis is that apoE3 binds to tau, 25 and protects it from phosphorylation in the microtubulebinding domain, while apoE4 does not bind to tau (Strittmatter et al. (1994) Exper. Neurol. 125:163-171).
S.As a result, tau gets hyperphosphorylated and can no longer bind efficiently and stabilize microtubules, leading to the formation of neurofibrillary tangles inside neurons.
Throughout the description and claims of this specification, the word "comprise" and variations of the word, such as "comprising" and "comprises", means "including but not limited to", and is not intended to exclude other additives, components, integers or steps.
H:\Bkrot\Keep\speci\75526-96.doc 20/03/00 3a Summary of the Invention We have discovered that glycosylated apolipoprotein E2 having a conformation which is the same as, or which closely approximates, its native 9**9 09 9 9* 9 9e 9 0e 9 9 9 9 .9 9 9 .s 9 9 9**9 9 9 9 9999 9. 9 9 9 9 9999 9 9 9 9999 9 *9999.
e .9 9 99 9 99 0 H:\Bkrot\Keep\speci\75526-96.doc 20/03/00 WO 97/16458 PCT/US96/17629 4 conformation, it is not denatured and then renatured, and exists in lipid-free as well as lipidbound forms, binds to synthetic peptide amyloid peptide AP with a higher affinity than do apolipoproteins E3 and E4.
Accordingly, the invention features an isolated apolipoprotein E2 capable of specifically binding to amyloid peptide AP with a higher affinity than apolipoproteins E3 or E4; the apolipoprotein E2 can be produced by transforming a eukaryotic (preferably mammalian) host cell with DNA encoding apolipoprotein E2; culturing the host cell; and isolating the apolipoprotein E2 under non-denaturing conditions. As used herein, "apolipoprotein E2" refers both to the naturally-occurring amino sequence of the protein, and to variants in which there are one or more amino acid substitutions which do not substantially alter the conformation or other properties of the molecule.
Examples of such mutant forms of E2 are given below in Table 1; these mutants not only retain native conformation when expressed in mammalian host cells, they exhibit superior binding properties compared to the native sequence.
Apolipoprotein E2 can be isolated under nondenaturing conditions by subjecting the growth medium from a culture of mammalian cells that express recombinant apolipoprotein E2 to cation exchange chromatography to produce an apolipoprotein E2-enriched cation exchange protein fraction, and then subjecting the resulting pool of apolipoprotein E2-enriched cation exchange fractions to anion exchange chromatography, thereby producing an anion exchange fraction highly enriched for apolipoprotein E2.
The invention also features a method for inhibiting the progression of Alzheimer's disease in a TABLE 1 DERIVATIVES OF APOE2 Helix 3 Repeats 3, 4 and %z of 5 V3 of Helices 6 and 7 Repeats 13, 14 and V2 of Leu97-VaL b (ApoE2 bjskmfund) Cys112-.Arg Asp11-Cys d I (ApoE2 background) Arg 1O3-.Cys Cys1 12-Arg (ApoE2 background) LeulO4-Valb (ApoE2 biwkgwund) Asp1O7-.Ala MetlO8-ALa a (ApoE2 background) Glu 1O9-Leu Asp11O-.Ata' (ApoE2 background) Cysi 12-Arg VaLil l-CYsd (ApoEZ background) Cys112-Arg Gtyll3-Cys (ApoE2 background) Cy12AgArg119-.Cysd (pE akrud GysL 2-A IapLug 5AIa (ApoE background)
C
Arg251-VaL GLu255-AL6C (ApoE2 background) Arg260-VaL 1ys262-ALa C(ApoE2 background) Asp270-Ala Gtu271-.Ata~ (A oE2 background) Met272-Ata Arg273-VaL (ApoE2 background) Thrl92-ALaf (ApoE2 background) Leu159-Cysf (ApoE3 background) Alal6O-.Cys.f (ApoE3 background) Val161-.Cys~ (ApoE3 background) Tyr162-Cysf, (ApoE3 background) Glu163-Cysf, (ApoE3 background) Ala164-Cys 4 (ApoE3 background) GLnl56-Cysf (pE akrud fsl3.y (ApoE3 background) As1eu4-Cy (ApoE3 background) Leu1448.Cysf (ApoE3 background) Aeal48-Cysf (ApoE3 background) G~l8~~ f (ApoE3 background) (ApoE3 background) 9 9 9 9* 9 *9 9 9 0*e 0 9* 99* .90 0 9*0* 0*0* 9 0 0 00** 0 9 0 0 9**0 9 Helix 4 Repeats a) Change charge interactions; b) Change Leucine zipper or hydrophobic interactions; c) Change of charge or hydrophobic residues of the carboxy terminal domain which may affect the interactions with helix 3 and AP, d) Retain Arq1lZ (E4 phenotype) but change other residues in helix 3 to Cys. e) Change Met residues that may be subiect to oxidation and the 0-gtycosylation site. f) Retain Arg-158 change other residues of helix 4 to Cys.
WO 97/16458 PCT/US96/17629 5 patient with early onset Alzheimer's disease, or inhibiting the onset of Alzheimer's disease in a patient genetically at risk of developing Alzheimer's disease, by administering to the brain of the patient an effective amount of apolipoprotein E2 which is glycosylated, exists in lipid-free or lipid-bound forms, and has native or close to the native conformation produced by brain cells.
Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims.
Brief Description of the Drawings Fig. 1 is a schematic representation of the steps leading to production of plasmids of the invention.
Fig. 2 is a series of gel electrophoresis and autoradiography panels of apoE2 secretion from cells of the invention.
Fig. 3 is a series of panels illustrating apoE2, E3, and E4 expression by various cell types using the Semliki Forest Virus expression system.
Fig. 4 is a pair of panels illustrating apoE expression in a bioreactor setting.
Fig. 5 is a set of panels showing elution profiles of apoE from ion exchange columns.
Fig. 6 is a set of panels showing binding to AP of apolipoprotein E obtained by expression in eukaryotic cells. The apoE experiment shows that the ability of apolipoprotein E produced by eukaryotic cells to bind to AP follows the order: apolipoprotein E2 apolipoprotein E3 apolipoprotein E4.
Fig. 7 is a set of panels showing binding to AP of apolipoprotein E3 and apolipoprotein E4 obtained by the baculovirus expression system. The ability of apoE3 and apoE4 forms thus obtained to bind to AP is different from that of apoE expressed by eukaryotic cells (Fig. 6).
WO 97/16458 PCT/US96/17629 6 Fig. 8 is a set of panels showing binding to AP of VLDL, apoE3 and apoE4. The ability of apoE3 and apoE4 forms thus obtained to bind to AP is different from that of apoE expressed by eukaryotic cells (Fig. 6).
Fig. 9 is a panel showing distribution of apolipoprotein E secreted by C127 cells to lipid-free and lipid-bound forms.
Fig. 10 is a diagram predicting binding of apolipoprotein E2 derivatives obtained from eukaryotic cells to AP which differ from the binding of naturally occurring apolipoproteins E3, E4 and native-sequence E2.
Detailed Description The present invention provides recombinant apolipoprotein E2 in its native conformation. Because of its native conformation, the apoE2 of the invention binds to the A/ peptide with greater affinity than apoE3 or apoE4.
Generation of cell lines of C127 cells expressing apoE2, apoE3 and apoE4.
We have generated a recombinant C127 cell line that secretes apoE2. This cell line has been generated as follows: The apoE cDNA we isolated previously (Zannis et al. (1984) J. Biol. Chem. 259:5495), amplified and mutagenized by PCR, was excised, with nucleases AatII and DraII, from the original plasmid, and subcloned into the SmaI site of the pUC19 plasmid to produce the pUC-E3 derivative. This was followed by excision of the apoE DNA with BamHI and BglII. The pUC-E3 plasmid was amplified and mutagenized by PCR as described in Fig. 1A, using a set of 5' and 3' amplification primers and a set of mutagenic primers. The mutagenic primers were designed to replace Cys-11 2 with Arg, and to replace Arg- 158 with Cys.
WO 97/16458 PCTIUS96/17629 7 The 5' external amplification primer extended from nucleotides 223 to 241 of the apoE gene (sequence numbering of Karathanassis, Haddad, Salmon and Zannis, Biochemistry and Biology of Plasma Lipoproteins 1985, pp.
475-493). The 3' external amplification primer extended between nucleotides 1102-1082 in the 3' region of the cDNA. One set of mutagenic sense and antisense oligonucleotides covered codons 154-161 (nucleotides 576- 595) for alteration of Arg-158 to Cys-158 in order to produce apoE2. Another set of mutagenic sense and antisense oligonucleotides covered codons 108-113 (nucleotides 438-457) for the change of Cys-112 to Arg-112 in order to produce apoE4.
For alteration of Arg-158-Cys, two separate amplifications were utilized. The first utilized the external primer and the antisense mutagenic primer covering codon 158. The second utilized the 3' external primer and the sense mutagenic primer covering codon 158.
An aliquot of 4% from each reaction was mixed and amplified with the 5' and 3' external primers. The amplified fragment containing the Arg-158-Cys mutation was used to replace the corresponding region in the pUC- E3 plasmid, as shown in Fig. 1A, giving rise to pUC-E2 plasmid. Similarly, the amplified fragment containing the Cys-112-Arg mutation was used to replace the corresponding region of the pUC- E3 plasmid, as shown in Fig. 1A, giving rise to pUC-E4 plasmid (Fig. 1A).
The DNA insert obtained from the pUC-E2 derivative was initially cloned into the BamHI site of the pcDNA under the control of the CMV pcDNA promoter or into the BamHI and BglII site of Semliki Forest Virus vector (pSFV) to generate the pcDNA-E2 derivative and the pSFV- E2 derivatives. The apoE2 insert was then excised from the pUC-E2 derivative, blunt-ended with the Klenow WO 97/16458 PCT/US96/17629 8 fragment of the DNA polymerase I, and ligated to XhoI linkers digested with XhoI and cloned into the XhoI site of pBMT3X vector to generate pBMT3X-E2, expression plasmid (Fig. 1B). A similar approach was utilized to generate the pBMT3X-E3 and E4 plasmids.
The pBMT3X is a bovine papilloma virus-based vector in which the gene of interest is transcribed under the control of the mouse metallothionein I promoter. The vector also contains the human metallothionein I gene, which confers resistance to 10 AM CdC1 2 (Cladaras et al.
(1987) J. Biol. Chem. 262:2310-2315).
To generate stable cell lines expressing apoE2, cells were transfected with the pBMT3X-apoE2 expression plasmid by the Calcium Phosphate DNA co-precipitation method (Graham and van der Eb (1973) Virology 52:456).
Permanent cell lines were selected for their resistance to 10gM CdCl 2 Resistant colonies were isolated with cloning cylinders and grown to 90% confluency.
To characterize the protein secreted by this cell line, the cell line was labelled with 35 S-methionine for 2 hours. Media and cell lysates were immunoprecipitated with rabbit anti-apoE, mixed with 15 gg HDL containing apoE3, and analyzed with two-dimensional gel electrophoresis (PAGE) and autoradiography. In this analysis, proteins are separated on the basis of their charge by isoelectric focusing and on the basis of their size by SDS-PAGE.
The gel obtained from this analysis showed the position of the marker plasma apoE3 (Fig. 2A,B,C), and the autoradiogram showed the position of the newly synthesized apoE2 protein (Fig. Superimposition of the gel on the autoradiogram established the charge and size differences between the plasma apoE3 and the newly synthesized protein (Fig.
This analysis, shown in Figs. 2A-C", WO 97/16458 PCT/US96/17629 9 established that the newly synthesized apoE2 is more acidic by one and two positive charges than apoE3 and apoE4, respectively. The analysis also shows that the secreted protein is extensively modified and resembles the protein synthesized by hepatic and other types of cells (Zannis et al. (1986) J. Biol. Chem. 261:13415- 13421).
We have also expressed the apoE2 in Cos-1 cells using the pcDNA vectors under the control of the CMV promoter.
Expression of ApoE cDNA Using the Semliki Forest Virus Expression System In addition to apoE2 expression in C127 cells, described above, we have also used other eukaryotic expression systems for expression of apoE2, apoE3 and apoE4 based on Semliki Forest Virus (SFV) replicon.
These systems consist of two plasmids pSFV1 and pSFVhelper-2. The first plasmid represents the cDNA copy of Semliki Forest Virus genome with a deleted subgenomic (26S) coding region for the structural proteins of the virus, followed by a cassette of polylinker (BamHI-XmaI- SmaI) and translational stop codons in all three reading frames. The cDNA of human apoE2 was cut with BglII and BamHI, present in the polylinker region of pSFV, and inserted via a BamHI BglII site to create vector pSFV apoE2. In this vector, apoE2 cDNA expression is driven by the 26S promoter in the context of SFV genome. The second plasmid is pSFV-helper 2, which retains the 5' and 3' signals needed for RNA replication, as well as the complete structural region, including its promoter, but which lacks a region required for packaging of SFV RNA into nucleocapsids.
The pivotal step in the development of the pSFV expression system is helper catalyzed packaging of WO 97/16458 PCT/US96/17629 10 pSFVcE2 RNA. The system allows rapid transient expression of apoE2 cDNA mutants, allowing fast testing of their functions, physicochemical properties, and neuronal toxicity.
Initially, plasmid pSFV apoE2, or pSFV-helper 2 was transcribed in vitro under the control of an SP6 promoter. Both SFV apoE2 and SFV-helper 2 RNAs were then cotransfected by electroporation into BHK cells. In the electroporated cells, the SFV apoE2 RNA encodes the enzymes for RNA replication (amplification) and transcription, whereas SFV helper 2 RNA supports the production of the structural proteins via its subgenomic region. Since the helper lacks the region required for RNA packaging, RNA derived from this vector will not be packaged. Thus, transfections with recombinant and helper RNA produce only virus particles that carry recombinant RNA.
The viruses produced provide a one-round virus stock. After infection of cells with the recombinant virus, the cells will only express the heterologous protein (apoE2). This virus stock was utilized to infect BHK cells or primary astrocytes obtained from mice deficient in apoE. For this analysis, confluent BHK cells or apoE deficient mouse primary astrocyte cultures were infected with recombinant SFV apoE2, virus at multiplicity of infection of approximately three infective units per cell for 12 hrs and grown in serum-free BHK medium for 24 h. The viral titer was estimated between 107 and 108 infectious units/ml. Aliquots of 50 pl medium were removed and analyzed by SDS-PAGE and immunoblotting (Fig.
The above description demonstrates that recombinant, glycosylated apoE2 can be efficiently produced in a variety of eukaryotic cells. The level of expression estimated from the intensity of the stained bands is approximately 5-10 g apoE/10 6 cells/1 ml/24 hrs.
WO 97/16458 PCT/US96/17629 11 Apolipoprotein E4 and Apolipoprotein E3 Were Expressed in a Similar Way Using the SFV Expression System.
Large-Scale Growth of ApoE2 Producing Cells in Bioreactors (Verax System The Verax bioreactors consist of a vertical column containing microspheres in suspension (Fig. 4A). The microspheres are made of collagen and are hollow with a pore diameter which allows cell attachment and growth.
Cell densities of 108 cells/ml are achieved. The microspheres are kept in suspension by the flow of incoming culture medium. A gas exchange unit provides the required 02 and removes excess CO 2 Temperature control keeps the medium at 37 0 C. The medium is circulated by a pump through the bioreactor and the gas exchange unit.
Using the Verax System-10 Bioreactors the culture medium contain 30-50 pg apoE/ml (Fig. 4B).
The medium of C127 cells expressing apoE2, apoE3 or apoE4 was purified by a scheme employing a dextran sulfate sepharose column and DEAE sepharose column fractionation.
Briefly, a 20 ml dextran sulfate-sepharose column was equilibrated with 20 mM Tris-HCl, 0.2 M NaCl, pH 7.4.
A total of 1 liter of apoE-containing culture medium was adjusted to 0.2 M NaCl and loaded on the column at a flow rate of -80 ml/hr. The column was washed with 200 ml of mM Tris-HCl, 0.2 M NaC1, pH 7.4 at the same flow rate.
The column was eluted with a 120 ml 0.2-1.0 M NaC1 gradient in 20 mM Tris-HCl, pH 7.4 at the same flow rate; fractions of 2 ml were collected. The apoE-containing samples eluted from the dextran sulfate sepharose column were adjusted to 50 mM NaC1 by dilution with 20 mM Tris- HCl pH 7.4 and were loaded on a 10 ml DEAE column. The column was eluted with 70 ml of 0.05-0.75 M NaCl gradient in 20 mM Tris-HCl pH 7.4.
WO 97/16458 PCT/US96/17629 12 Properties of Apolipoproteins E3. E4 and E2 Produced by Mammalian Cells and Other Sources.
Binding of ApoE2. ApoE3 and ApoE4 Produced by Cell Cultures to the Synthetic Amyloid Peptide AB.
For this analysis, 1 Ag of apoE2 or apoE3 or apoE4 was diluted with 9 Al H 2 0 containing either 0 or 4 M As amino acid residues in length) in 20 mM tris pH buffer and incubated at 25 0 C for 12 hrs. Incubation was terminated by adding equal volume 2X Laemmli buffer (4% SDS without P-mercaptoethanol) and boiling for 5 min. The mixture was analyzed by gel electrophoresis in two gels polyacrylamide 2% SDS) and Western blotting. After blotting one of the filters (Panel A) was treated with mouse monoclonal antibody against A3 (Fig. 6A) and the other two panels (Panels B&C) with goat polyclonal antibody against apoE.
This experiment shows that recombinant apoE2, apoE3 and apoE4 form stable diffused complexes with Af in solution (Fig. 6A). Each such complex is stable following boiling in 2% SDS. The diffused nature of the complex may result from the fact that apoE produced by these cell lines is highly modified with carbohydrate chains containing sialic acid. There is no formation of complexes with higher Mr with any of the isoforms. The formation of complexes can be seen also in the first three lanes of Fig. 6B in lane 4 which do not contain AP.
In this experiment equal quantities of apoE (1 Mg) and Ap (4 pg) were utilized for all the isoforms. The analyses of Fig. 6A-C show that the ability of apoE produced by these cells to bind to AP follows the order apoE2>apoE3>apoE4. Similar results are obtained with apoE produced by apoE produced by the different expression systems described before. This binding behavior of apoE is different from that of apoE produced by the baculovirus expression system (Fig. 7) or VLDL (Fig. 8) WO 97/16458 PCT/US96/17629 13 (Strittmatter et al. (1993) Proc. Natl. Acad. Sci. USA 90:8098-8102).
The experiments of Fig. 6A-C show that recombinant apoE2, apoE3 and apoE4 form stable complexes with AP.
These complexes are visible in the first three lanes of Fig. 6A,B, which contain AP but not in the corresponding lanes of Fig. 6C, which does not contain AP. This analysis establishes unequivocally that the binding of newly synthesized apoE secreted into the cell medium which consist mostly of modified forms of apoE with AP follows the order apoE2>apoE3>apoE4.
In other experiments we have analyzed the apoE secreted by cell cultures by density gradient ultracentrifugation and immunoblotting. This analysis showed that approximately 45% of apoE is distributed in lipoprotein particles which float in the HDL region in d=1.08-1.20 g/ml, 40% in lipid-poor particles which float in the d=1.22-1.28 g/ml region, and 15% in a lipid-free fraction d>1.33 g/ml (Fig. These different types of particles participate to different degrees in the formation of complexes with AP.
According to epidemiological data (Corder et al.
(1993) Science 262:921) and the findings of Fig. 6A-C, increased binding of apoE isoforms to AP correlates negatively with the risk of developing Alzheimer's disease. Derivatives of apoE2 generated by in vitro mutagenesis containing one or more amino acid substitutions which do not substantially alter the conformation or other properties of the molecule may surpass the ability of the naturally occurring apoE2 form to bind AP. These derivatives (examples are shown in Table 1) will provide a series of experimental points to assess the binding behavior of apoE (Fig. 10). Taking into account the reported progress in the generation of an animal model of AD (Gomes et al. (1995) Nature WO 97/16458 PCT/US96/17629 14 373:523), one approach to further establish the relationship between the binding of apoE to Ap and AD will be as follows: mice expressing natural or mutant apoE forms in an apoE deficient background (Plum et al.
(1992) Cell 71:343-353) will be mated with the AD-prone mouse generated by Athena Neuroscience (Gomes et al.
(1995) Nature 373:523). Analysis of the offspring will provide information as to whether a specific apoE variant protects or accelerates the development of the AD phenotype in relationship to the parent AD mouse. For instance, crossing of the AD prone mouse with the EX1 or EX6 mouse will confirm that strong binding of apoE to AP is beneficial and weak binding is detrimental. If strong binding is beneficial, we will anticipate that the EX6 form will protect from AD when it is introduced into the AD-prone mouse, whereas EX1 will accelerate the development of AD.
Formulation and Administration To be effective, the apoE2 of the invention should come in contact with neuronal cells of the brain, or with the physiological fluids in the immediate vicinity of such cells. The apoE2, in that environment, binds to the Apamyloid peptide, or to P-amyloid precursor protein; when it binds to precursor protein; it prevents the precursor from progressing to the next stage, which is cleavage to form the P-amyloid protein characteristic of Alzheimer's disease. When apoE2 binds to A#-amyloid peptide, such binding inhibits progression of Alzheimer's disease by preventing formation of the extracellular amyloid plaques.
Generally, there are several approaches to bring apoE2 into contact with brain cells or to remove A/ from the brain.
WO 97/16458 PCT/US96/17629 15 It has been shown that noncovalent association of apoE with lecithin phospholipids leads to the formation of discoidal particles (Innerarity et al. (1979) J. Biol.
Chem. 254:4186-4190). ApoE also associates with triglyceride-rich phospholipid emulsions (Derksen and Small (1989) Biochemistry 28:900-906).
In one method, apoE2 produced by eukaryotic cells will be administered into the ventricles of the brain and will enter into the cerebrospinal fluid where it will be bound to AP.
In another method, apoE2 phospholipid vesicles or triglyceride phospholipid emulsion will be administered into the ventricles of the brain. The AS will bind to the vesicles or the emulsions and will be removed from the brain.
In a third method, apoE2 phospholipid vesicles or the triglyceride phospholipid emulsions will be administered intravenously. The complexes will cross the blood/brain barrier by interactions with the apoE receptor present in the blood/brain barrier.
In a fourth method, biotinylated polyamides will be conjugated to streptavidin conjugated apoE2 according to Partridge et al. (1995) Proc. Natl. Acad. Sci. USA 92:5592-5596). Following intravenous administration, the complex will cross the blood/brain barrier by interaction with the apoE receptor present in blood brain barrier.
Free apoE2 or apoE2 associated with lipids can be administered to the ventricle of the brain by means of a stereotaxic instrument which is commercially available.
The stereotaxic coordinates of the insertion point on the skull is marked by a sterile pencil. A hole of 2-3 mm is made in the skull. The dura is punctured and is covered with saline soaked gel-foam using the stereotaxic holder, and a thin tube is lowered into the ventricle and is connected with the infusion pump that will deliver the WO 97/16458 PCT/US96/17629 16 apoE2 in lipid-free or lipid-bound form into the ventricle.
The apoE2 solution or apoE2 phospholipid vesicles or apoE2 triglyceride phospholipid emulsions are loaded into the intraventricular pump, which has been implanted into the patient by known techniques. Each apoE2 preparation is continuously infused into the ventricle of the patient's brain, at a rate sufficient to deliver 500 to 1000 ug of protein per 75 kg body weight of the patient per day.
Another mode of administration is to inject the protein, dissolved in a carrier liquid, into the spinal column of the patient so that it contacts the patient's cerebrospinal fluid.
Another mode of administration is intravenous injection of apoE2 phospholipid vesicles or apoE2 triglyceride phospholipid emulsions or conjugates of apoE2 with polyamides in physiological solutions containing 2 mg apoE2/ml. Ten mls of apoE2-containing solution will be injected twice per day.
Generally, the apoE2 of the invention can be administered to three classes of patients: 1. Patients who have been diagnosed with Alzheimer's disease, and who are exhibiting moderate to severe symptoms of the disease; 2. Patients who are exhibiting early-stage symptoms of Alzheimer's disease, and who are therefore suspected of having the disease; and 3. Asymptomatic patients who are over the age of 45 and who have a family history of Alzheimer's disease; one class of such patients are those carrying two copies of the apoE4 gene.
An alternative method of treatment is to administer DNA encoding apoE2 rather than apoE2 to protein. This gene therapy method is preferably carried 17 out using either a retroviral vector or an expression vector containing apoE2encoding DNA, suspended in a liquid carrier; the suspension is administered either to the cerebrospinal fluid or the brain ventricle in the same manner that the protein in solution is administered. Suitable plasmid and retroviral vectors are well known and publicly and commercially available.
Other embodiments are within the following claims.
e e a e *9.
a H:\Bkrot\Keep\speci\75526-96.doc 20/03/00

Claims (7)

1. Recombinant apolipoprotein E2, said apolipoprotein E2 being capable of biding to amyloid peptide or to amyloid 3 precursor protein with greater affinity than do apolipoproteins E3 and E4, wherein said apolipoprotein E2 is glycosylated and is produced by the process of transforming a mammalian host cell with DNA encoding apolipoprotein E2, culturing said host cell in a growth medium, and isolating said apolipoprotein E2 under non- denaturing conditions.
2. A apolipoprotein E2 according to claim 1, having one of the amino acid sequences shown in Table 1. S3. A recombinant apolipoprotein E2 according to **20 claim 1 or claim 2, wherein Cys 112 of the native molecule is replaced with Arg.
4. A recombinant mammalian cell that expresses a recombinant apolipoprotein E2 according to any one of 25 claims 1 to 3.
5. A cell according to claim 4, wherein said apolipoprotein E2 has a non-native amino acid sequence.
6. A transgenic mouse having an apoE-deficient background, wherein said mouse expresses an apolipoprotein E2 according to any one of claims 1 to 3 in neurons of its brain.
7. A transgenic mouse according to claim 6, wherein said apolipoprotein E2 is human. H:\Bkrot\Keep\speci\15526-96.doc 20/03/00 19
8. A recombinant apolipoprotein E2 according to claim 1 substantially as hereinbefore, described with reference to any one of the examples. Dated this 20th day of March 2000 KOS PHARMACEUTICALS, INC and TRUSTEES OF BOSTON UNIVERSITY By their Patent Attorneys GRIFFITH HACK Fellows Institute of Patent and Trade Mark Attorneys of Australia H\Ekrot\Keep\speci\75526-96.dec 20/03/00
AU75526/96A 1995-11-01 1996-10-31 Apolipoprotein E2 and treatment of Alzheimer's disease Ceased AU720446B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US55172295A 1995-11-01 1995-11-01
US08/551722 1995-11-01
PCT/US1996/017629 WO1997016458A1 (en) 1995-11-01 1996-10-31 Apolipoprotein e2 and treatment of alzheimer's disease

Publications (2)

Publication Number Publication Date
AU7552696A AU7552696A (en) 1997-05-22
AU720446B2 true AU720446B2 (en) 2000-06-01

Family

ID=24202413

Family Applications (1)

Application Number Title Priority Date Filing Date
AU75526/96A Ceased AU720446B2 (en) 1995-11-01 1996-10-31 Apolipoprotein E2 and treatment of Alzheimer's disease

Country Status (7)

Country Link
EP (1) EP0866799A4 (en)
JP (1) JP2000500332A (en)
AU (1) AU720446B2 (en)
BR (1) BR9611329A (en)
CA (1) CA2236274A1 (en)
MX (1) MX9803422A (en)
WO (1) WO1997016458A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1121429A2 (en) * 1998-10-16 2001-08-08 Introgene B.V. Gene therapy of alzheimer's disease by delivery of an encoded apolipoprotein e
AU1742200A (en) * 1998-11-25 2000-06-13 Eli Lilly And Company Prevention and treatment of amyloid-associated disorders
CN1315402A (en) * 2000-03-28 2001-10-03 上海博德基因开发有限公司 Polypeptide-human amyloid precursor protein bindin 14 and polynucleotide for coding it
CN1315439A (en) * 2000-03-29 2001-10-03 上海博德基因开发有限公司 Polypeptide-human amyloid precursor protein bindin 9 and polynucleotide for coding it
US20070036761A1 (en) * 2003-09-26 2007-02-15 Bales Kelly R Lentiviral apoe2 gene therapy
JP2009531299A (en) * 2006-02-21 2009-09-03 オクラホマ メディカル リサーチ ファウンデーション Treatment of Alzheimer's disease with inhibitors of ApoE binding to the ApoE receptor
WO2010020822A1 (en) * 2008-08-19 2010-02-25 University Of Patras Therapy for apolipoprotein-induced hypertriglyceridemia

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116739A (en) * 1984-10-16 1992-05-26 Mitsubishi Chemical Industries Limited Process for the production of human apolipoprotein e, and transformed hosts and products thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9414624D0 (en) * 1994-07-20 1994-09-07 Smithkline Beecham Plc Novel method

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116739A (en) * 1984-10-16 1992-05-26 Mitsubishi Chemical Industries Limited Process for the production of human apolipoprotein e, and transformed hosts and products thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CELL, VOL. 71 PAGES 343-353 *
EXPERIMENTAL NEUROLOGY 1994, VOL. 125 PAGES 163-171 *

Also Published As

Publication number Publication date
EP0866799A1 (en) 1998-09-30
MX9803422A (en) 1998-11-29
JP2000500332A (en) 2000-01-18
AU7552696A (en) 1997-05-22
BR9611329A (en) 1999-12-28
WO1997016458A1 (en) 1997-05-09
EP0866799A4 (en) 2000-08-23
CA2236274A1 (en) 1997-05-09

Similar Documents

Publication Publication Date Title
CALERO et al. Functional and structural properties of lipid-associated apolipoprotein J (clusterin)
EP2767546B1 (en) Lipoprotein complexes and manufacturing and uses therof
US5750367A (en) Human and mouse very low density lipoprotein receptors and methods for use of such receptors
EP0446017A1 (en) New diagnostic and treatment methods involving the cystic fibrosis transmembrane regulator
CA1316132C (en) Cdna and gene for human angiogenin (angiogenesis factor) and method of expression
EP0239631A1 (en) Recombinant apolipoproteins and methods
EP0275305A1 (en) Dna sequences coding for modified factor viii:c and modified factor viii:c-like polypeptides and processes for producing these polypeptides in high yields
WO1993000443A1 (en) Purification of recombinant apolipoprotein e from bacteria
TW434260B (en) New variants of apolipoprotein A-I
Li et al. Domains of apoE required for binding to apoE receptor 2 and to phospholipids: implications for the functions of apoE in the brain
JP2757987B2 (en) Human anti-inflammatory phospholipase inhibitor protein
AU720446B2 (en) Apolipoprotein E2 and treatment of Alzheimer's disease
US4943527A (en) Mature apoai protein production under serum free culturing conditions
RU2128705C1 (en) Protein inhibiting collagen-caused aggregation of mammalian platelets, recombinant protein inhibiting collagen-caused aggregation of platelets, dna fragment encoding recombinant protein, expression vector (variants), strain of cultured mammalian cells bhk - producer of recombinant protein (variants), method of preparing recombinant protein, pharmaceutical composition
US5622843A (en) Phospholipid transfer proteins and DNA encoding them
JP2009171980A (en) Compound and method for lowering cholesterol level without inducing hypertriglyceridemia
Mehta et al. The low density lipoprotein receptor in Xenopus laevis. II. Feedback repression mediated by conserved sterol regulatory element
US20060142200A1 (en) Compounds and methods for lowering cholesterol levels without inducing hypertrigylceridemia
CA2042605A1 (en) Cytolysis inhibitor proteins (cli), dna sequences coding for said proteins, and plasmids, host organisms, and processes for obtaining said proteins
CN1206421A (en) Apolipoprotein Ez and treatment of alzheimer's disease
US20070053879A1 (en) Diagnostic and treatment methods involving the cystic fibrosis transmembrane regulator
AU2018203258B9 (en) Lipoprotein complexes and manufacturing and uses thereof
AU8418691A (en) Purification of recombinant apolipoprotein e from bacteria
Laccotripe Structural and functional analysis of human apolipoprotein AI
Bogari A study of the relationship between ABCA I gene polymorphisms, plasma lipid levels and risk of atherosclerosis

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)