EP0239631A1 - Apolipoproteines recombinantes et procedes - Google Patents

Apolipoproteines recombinantes et procedes

Info

Publication number
EP0239631A1
EP0239631A1 EP86906545A EP86906545A EP0239631A1 EP 0239631 A1 EP0239631 A1 EP 0239631A1 EP 86906545 A EP86906545 A EP 86906545A EP 86906545 A EP86906545 A EP 86906545A EP 0239631 A1 EP0239631 A1 EP 0239631A1
Authority
EP
European Patent Office
Prior art keywords
lipid
gene
peptide
apoai
binding
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.)
Withdrawn
Application number
EP86906545A
Other languages
German (de)
English (en)
Other versions
EP0239631A4 (fr
Inventor
Andrew A. Protter
Jean-Louis Vigne
Joanne B. Mallory
Karen D. Talmadge
John P. Kane
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.)
Biotechnology Research Partners Ltd
Original Assignee
Biotechnology Research Partners Ltd
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 Biotechnology Research Partners Ltd filed Critical Biotechnology Research Partners Ltd
Publication of EP0239631A1 publication Critical patent/EP0239631A1/fr
Publication of EP0239631A4 publication Critical patent/EP0239631A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • C07K2319/75Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor containing a fusion for activation of a cell surface receptor, e.g. thrombopoeitin, NPY and other peptide hormones

Definitions

  • the present invention relates to lipid-binding peptides, methods of producing, purifying and formulating the peptides, and novel lipopeptide compositions.
  • Lipid-binding proteins and their peptide fragments are potentially important in a number of therapeutic applications.
  • One major class of lipoproteins are the serum lipoproteins which are present in a variety of macromolecular protein/lipid complexes which transport and regulate serum levels of cholesterol, triglycerides, phospholipids, and other serum lipids. Since many cardiovascular diseases are related to lipid imbalances, it has been proposed to treat such diseases by administering one or more apolipoproteins or apolipoprotein/lipid complexes which can exert a counter effect on an undesired pattern of lipid accumulation in the tissues of the patient.
  • Apolipoproteins share a common design with respect to the arrangement of lipid-binding regions.
  • the proteins have a segment or segments which contain a series of amphipatic ⁇ -helical lipid binding regions. Some of the proteins also have a relatively polar region which may be a receptor-binding region or play some other protein-identification role.
  • the apolipoprotein is designed to perform a protein-specific function involving the more polar region.
  • Applications of this type are expected to require the polar regions of the complete mature apolipoproteins.
  • lipid-binding proteins In order to bring such therapeutic application of lipoproteins to a practical stage, it is necessary to produce the lipid-binding proteins in high-levels and usually, in relatively pure form.
  • the apolipoproteins may be isolated from human serum, the purification schemes are time consuming, and the yields of some are relatively poor.
  • difficulties arise in attempting to separate selected apolipoprotein types from one another. These difficulties are illustrated by current methods for purifying serum apolipoprotein A-4 (Weinberg, L.B, et al, J Lipid Res 26:26 (1985); and Weinberg, R.B., et al, J Biol Chem 260:4914 (1985)).
  • proteins purified by these methods can be contaminated with infectious viruses, such as hepatitis.
  • apolipoprotein Al (abbreviated apoAI) (Seilhamer, J.J., et al. DNA 3:309 (1984)), apoAII (Moore, et al, Biochera Biophys Res Commun 123:1 (1984), apoAIV (Bogouski, D., et al, Proc Natl Acad Sci (USA) 81:5021 (1984)).
  • apoCI Knott. et al.
  • a related object of the invention is to provide a high-producing expression system capable of producing and secreting recombinant apolipoprotein in processed, or mature, form.
  • Another related object of the invention is to provide a method for use in an expression system in which a heterologous lipid-binding peptide is present in a mixture of proteins produced by the system, for separating the lipid-binding peptide by simple floatation and/or size-related fractionation procedures.
  • Yet another object of the invention is to provide a stabilized nutritional emulsion which can be formulated in large scale using methods of the invention. Producing a nascent high-density lipoprotein complex by simple recombinant DNA methods is still another object of the invention.
  • the invention includes a method for producing, a purified lipid-binding peptide which can bind to phospholipids at one or more amphipatic alpha-helical peptide regions.
  • a gene coding for the peptide is introduced in expressible, heterologous form in a suitable expression system capable of synthesizing a mixture of peptides which includes the lipid-binding peptide.
  • Addition of either endogenous or exogenous lipids to the peptide mixture forms a particulate Hpopeptide complex composed of lipid and the lipid-binding peptide, and this complex can be separated easily from nonlipid-binding peptides in the peptide mixture on the basis of its size or, preferably, its density.
  • the expression system includes Chinese hamster ovary (CHO) cells, and the gene is introduced in an expression vector having a regulatable promoter derived from the human metallothionein II gene. Synthesis of the lipid-binding protein in the system is under the control of the promoter, which is responsive to the concentration of divalent metals in the cell medium.
  • the cells are effective to produce the apolipoprotein in mature, secreted form, and can also form, from endogenous lipid, the lipoprotein complexes which can be readily separated from nonlipid-binding proteins according to the method of the invention.
  • High-producer CHO cells can be selected by clonal selection.
  • the lipid which is added to the mixture of proteins produced in the expression system may be endogenous lipid.
  • Figure 1 is a flow diagram showing peptide synthesis, separation, purification, and formulation processes involved in the invention
  • Figure 2 shows the construction of two bacterial vectors, designated ⁇ Ap85 and pApoF, designed for expression of human apoAI C-terminal genomic fragment in E. coli;
  • Figure 3 is an autoradiographic analysis for the expression of cloned human apoAI C-terminal genoraic fragment in E. coli;
  • Figure 4 shows the construction of a bacterial vector, designated pFLAl-10. designed for expression of mature human apoAI in E. coli;
  • Figure 5 is an autoradiographic analysis of the expression of cloned human apoAI in E. coli;
  • Figure 6 shows the construction of a yeast vector, designated YEp-a-aA-11, designed for expression of mature human apoAI in S. cerevisiae:
  • Figure 7 is an autoradiographic analysis of the expression of cloned human apoAI in S. cerevisiae
  • Figure 8 shows the construction of a mammalian-cell vector, designated pMTAIR, designed for expression of mature human apoAI in Chinese hamster ovary (CHO) cells;
  • Figure 9 is an autoradiographic analysis of the expression of cloned human apoAI in CHO cells
  • Figure 10 shows gel electrophoretic patterns of proteins expressed by CHO cells transformed with control and pMTAIR vectors, and the proteins, including apoAI, which float with lipid emulsion particles.
  • Figure 11 is an autoradiographic analysis of the expression of cloned human apoAII in CHO cells
  • Figure 12 shows the construction of a mammalian-cell vector, designed for expression of cloned human apoAI C-terminal genomic fragment in CHO cells;
  • Figure 13 is a negative-stain electron micrograph of lipoprotein complexes formed in accordance with the invention.
  • Figure 14 is an ultraviolet scan of the elution profile from an HPLC column used to purify recombinant mature human apoAI;
  • Figure 15 shows the gel electrophoretic pattern of mature human apoAI produced by roller bottle culture and purified in accordance with the invention
  • Figure 16 is a negative-stain electron micrograph of phospholipid vesicles before lipoprotein complex formation by addition of recombinant apoAI
  • Figure 17 is a negative-stain electron micrograph of lipoprotein complexes formed by addition of recombinant apoAI to the lipid vesicles of Figure 16;
  • Figure 18 shows the DNA sequence of the cDNA insert containing the signal sequence and amino acid codons of mature apoB and the corresponding amino acid sequence
  • Figure 19 illustrates vector constructs used in isolating apoB cDNAs.
  • Figure 1 illustrates the general schema of the invention.
  • the upper portion of the flow diagram indicates the steps of (1) selecting of a suitable lipid-binding peptide, (2) constructing or isolating a gene coding for the peptide, and (3) placing the gene in an expression vector.
  • the expression vector may be one designed for use in either a bacterial, yeast, or mammalian-cell expression system. These aspects of the invention are discussed in Section I below. In Section II, methods of expressing the selected LPB in a variety of hosts are described.
  • the recombinantly produced LBP can be combined with lipids to produce lipid-protein complexes which may take the form of protein-associated triglyceride emulsions (LBP/emulsions) or LBP/phospholipid vesicle/(LBP/PL). Lipid emulsions or phospholipid vesicles may be added exogenously to the LBP, to form the lipoproteins. It has also been discovered, according to one aspect of the invention, that a mammalian-cell expression system, such as Chinese hamster ovary (CHO) cells, can supply endogenous lipids which combine with the expressed LBP to form the desired lipoprotein. These aspects of the invention are considered in Section III.
  • CHO Chinese hamster ovary
  • the lipoprotein complexes can be separated readily from nonlipid binding proteins on the basis of increased size or density, to yield lipoproteins which are largely purified from nonlipid binding proteins also produced in the expression system.
  • the lipoprotein separation provides a simple, rapid method for purifying lipid-binding peptides such as apolipoproteins in large scale. This approach will be detailed in Section IV.
  • the purified lipoprotein formed by LBP in association with lipid emulsion may be suitable, without further purification, as a stabilized lipid emulsion for nutritional purposes.
  • the LBP in the emulsion may be further purified and recombined with emulsion lipids to form an emulsion which is generally suitable for parenteral use.
  • the increased stability of the lipoprotein. emulsion is discussed in Section V.
  • the lipoprotein formed by association with phospholipids is useful directly as a nascent high-density lipoprotein (HDL), or the protein may be further purified and reassociated with phospholipid to form a nascent HDL composition.
  • HDL high-density lipoprotein
  • the utility of nascent HDL in reverse cholesterol uptake is also considered in Section V.
  • lipid-binding peptide refers to a protein or protein segment which has, or in the presence of phospholipids assumes, an amphipatic alpha-helical structure characterized by a polar side or face that can interact with the polar head groups of phospholipids, and a nonpolar side which can interact with lipid acyl chain groups.
  • the polar face contains charged amino acid residues, typically with acidic groups toward the center of the helix paired with basic residues at the edge, and it has been postulated that these charge pairs are able to form favorable ionic interactions with zwitterionic phospholipid head groups.
  • the nonpolar face contains largely hydrophobic residues.
  • apolipoproteins are one general source of lipid-binding peptides.
  • Apolipoproteins refer to the delipidated proteins which, when combined with lipids, such as triglycerides, cholesterol, cholesterol esters, and phospholipids (Breslow. J.L., Ann Rev Biochem 54:699 (1984)). form various types of lipoprotein complexes.
  • Alpha-helical, amphipatic lipid-binding regions are common to all human apolipoproteins, including apoAI, apoAII, apoAIV, apoB, apoCI, apoCII, apoCIII, apoD, and apoE.
  • the lipid-binding peptide may include the entire apolipoprotein. or one or more lipid-binding regions thereof.
  • the amino acid sequence and composition of the LBP may precisely match that of an apolipoprotein or lipid-binding region(s), or it may contain one or more amino acid substitutions which do not interfere with the LBP's ability to form an alpha-helical structure necessary for lipid binding (neutral substitutions).
  • the gene coding for selected LBP peptide is defined herein to include any natural or synthetic DNA sequence which codes for the selected LBP, and can be expressed in heterologous form in a suitable expression system.
  • the gene coding for the selected LBP may be derived either from genomic DNA or cDNA (copied from an isolated mRNA).
  • the gene is said to be derived from genomic or cDNA if it includes a partial or full-length DNA sequence coding for: (a) a human apolipoprotein, (b) one or more alpha-helical binding regions thereof, or (c) proteins or peptides containing neutral substitutions in (a) or (b).
  • the gene may also be a synthetic polynucleotide.
  • the coding sequence in the gene is constructed to correspond to the LBP amino acid sequence, or more preferably, to match the known gene sequence of a selected apolipoprotein lipid-binding region, as reported, for example, in the references given above on the full-length cDNA and/or genomic sequence of a variety of apolipoproteins and in Example XVIII.
  • the oligonucleotide can be synthesized, typically as a series of overlapping segments, by known methods.
  • a partial or full-length genomic DNA or cDNA for a selected apolipoprotein can be obtained according to published methods or by known cloning and selection procedures. The procedures described by the inventors (Seilhamer.
  • the cDNA inserts were subcloned into a bacterial plasmid, and the resulting plasmid library screened with a 15-mer representing the anti-codons for the last five amino acids of apoAI.
  • One of the clones, containing a 300 bp insert encompassing the 3'-terminal end of apoAI mRNA was selected, and from this an Alul fragment was obtained for use as a probe in screening for full-length cDNA.
  • the largest cDNA probe identified contained a 5' untranslated region 39 nucleotides long, and an open reading frame of 801 nucleotides coding for the complete 267 amino acid prepro-apoAI. Following the TGA termination codon was a 3' untranslated region of 55 nucleotides containing the polyadenylation signal AATAAA and a short polyA tail.
  • genomic apoAI gene was obtained by screening the cloned genomic fragment with the above Alul cDNA probe. One 2.2 kb PstI fragment was identified as containing the entire apoAI gene, including 5' and 3' untranslated regions. A comparison of the genomic and cDNA sequences revealed the presence in the genomic gene of three introns which interrupt the coding sequence in (a) the 5' untranslated region, (b) an 18 amino acid signal sequence, and (c) the region coding for the mature peptide.
  • Example XI describes similar methods for obtaining a full-length genomic clone coding for apoAII. References noted above also give details for obtaining genomic and/or cDNA genes for apo AIV, CII, CHI, and E.
  • Example XVIII describes similar methods for obtaining a cDNA that encodes the signal sequence and first 430 amino acids of apoB, which includes the receptor-binding domain and two amphipatic helices. This cDNA is described in detail in co-owned patent application for "Novel Lipoprotein-Based Drug-Delivery Systems", Serial No. 783,787, filed 3 October 1985.
  • the LBP gene from above is placed into a suitable expression vector for expression in a bacterial, yeast, or mammalian-cell system.
  • the vector used in the construction contains the necessary control elements for gene transcription and translation in a selected host system, and one or more restrictions sites at which the heterologous gene can be inserted in expressible form.
  • Vectors suitable for expressing protein in bacteria typically E. coli, and yeast, typically S. cerevisiae. are available, and methods for introducing genes in expressible forms into the vectors are known.
  • the Methods section below outlines general procedure for inserting heterologous gene sequences into expression vectors, selecting successful recombinants, and verifying the constructions.
  • the methods described in Example I for construction of a bacterial vector having an apoAI genomic fragment insert, and in Examples III and V, for construction of a bacterial vector and yeast vector, respectively, with a full-length apoAI cDNA insert are generally applicable.
  • Full-length cDNA was used in the vectors designed for production of complete apolipoproteins, to avoid limitations in bacterial and yeast systems in transcribing intron-containing genomic genes.
  • the gene in the yeast vector contained codons for the mature apoAI minus the first seven N-terminal codons.
  • a preferred mammalian-cell expression vector has been described in co-owned patent application for "A Superior Mammalian Expression System". Serial No. 701,296, filed 13 February 1985.
  • the vector contains the human metallothionein-II (hMT-II) promoter and is designed to transform Chinese hamster ovary (CHO) cells.
  • the vector/cell system is capable of high levels of expression of the heterologous gene, and gene expression is inducible, under the control of the hMT-II promoter, by addition of divalent metals, such as divalent zinc and iron.
  • genomic (intron-containing) genes and cDNA copies thereof, encoding a variety of protein and protein segments, including apolipoproteins and apolipoprotein ⁇ -helical segments, are expressed efficiently and at high levels in the CHO system.
  • the CHO cells are also capable of processing expressed apolipoproteins to mature proteins, and of forming nascent lipoprotein complexes with endogenous producing lipid.
  • hMT-II vector construction methods are generally applicable to other apoproteins and lipid-binding segments thereof.
  • An exemplary CHO expression vector containing the hMT-II promoter and a full-length genomic apoAI coding region under the control of the promoter in a high-producing CHO cell line was deposited with the American Type Culture Collection ATCC Patent Depository on 3 October 1985 and has deposit # CRL 8911.
  • the vector in the deposited CHO cells can be readily manipulated, by known techniques, to substitute other apolipoprotein or protein fragment coding regions for the apoAI region.
  • a gene coding for an LBP is joined to the gene of a second protein or peptide in the expression vector, to produce a fused-protein gene composed of the LBP gene and the coding sequence for a second, typically hydrophilic, protein.
  • the LBP moiety in the expressed fused protein can be used to facilitate protein purification, particularly in a scaled-up production method, according to procedures detailed in Section IV below. Methods for fusing an LBP gene to a second coding sequence would follow known cloning procedures.
  • the LBP gene would be attached at the C-terminus of the second-protein's coding region.
  • neither peptide moiety contains a methionine residue
  • the two genes can be fused at a methionine codon, to allow the fused protein to be cleaved into its unfused components, after purification, by cyanogen bromide cleavage, according to known methods.
  • Expression of the selected LBP gene is achieved by transforming a suitable bacterial, yeast, or mammalian-cell host with the expression vector constructed as above.
  • Methods for transforming bacterial and yeast cells with suitable expression vectors are known, and generally follow the methods outlined in Examples II and IV, for transforming E. coli. and Example VI, for transforming S. cerevisiae. Also described in these examples are typical reaction conditions under which protein synthesis in the expression system takes place.
  • the proteins produced in the system are analyzed for the presence of a peptide that can be distinguished on the basis of its unique size or reactivity with anti-LBP-antibody.
  • the cells For proteins formed in a bacterial expression system, which are often produced as intracellular forms, the cells must be ruptured or lysed, such as by detergent treatment, and the cell debris removed, such as by centrifugation or filtration.
  • the released cellular proteins when fractionated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), show a series of fractionated proteins which migrate on the gel according to their molecular weight.
  • Comparison of the fractionated proteins from cells transformed either with the LBP-gene vector, or a control vector may show the presence in the LBP cell proteins of a unique molecular weight species corresponding to the expected molecular weight of the LBP.
  • the identity of the LBP can be further confirmed by precipitating the protein mixture with anti-LBP antibody, and fractionating the precipitated material by SDS-PAGE, as described generally in Example II.
  • a bacterial system E. coli
  • E. coli is capable of producing both a apoAI fragment containing three amphipatic, alpha-helical protein regions and full-sequence apoAI itself.
  • the apolipoproteins have strong sequence similarities in their alpha-helical binding regions.
  • the finding herein that the bacterial system is capable of expressing both apoAI and a lipid-binding region thereof indicates that the system is capable of expressing genes which encode apolipoprotein alpha-helical lipid binding regions generally, whether the lipid binding regions are portions of an apolipoprotein, or the full-length apolipoprotein.
  • the bacterial system of Examples II and IV does not carry out post-translation processing of the expressed LBP because the gene was tailored to exclude the N-terminus signal sequence and propeptide codon(s). Similar methods were used to show that a yeast system (S. cerevisiae) is capable of expressing apoAI (Example VI). In the yeast system, the LBP are secreted into the extracellular medium, and therefore can be harvested simply by removing cells, such as by centrifugation. from the cell medium.
  • a preferred system for expression of LBP is the mammalian-cell system composed of CHO cells transformed with the hMT-II promoter vector. For a variety of reasons, this system is well suited to scaled-up production and purification of apolipoproteins and other LBPs.
  • One advantage of the system is its inducibility for LBP expression, by addition of divalent metals to the cell medium.
  • Example X below demonstrates the increasing levels of apoAI production which are produced at increasingly greater zinc concentrations. Expression levels may be increased substantially in clonal strains of CHO cells which are selected for high production of LBP.
  • Example IX illustrates the selection of clonal variants of the CHO cells which are high producers of mature apoAI.
  • the selection procedure involves culturing the cells under low cell concentration conditions which lead to individual-cell (clonal) colonies in the culture. These colonies are picked and screened for high levels of LBP expression, for example, by dot-blot Western blotting (Jahn et al, Proc Natl Acad Sci (USA) 81:1684-1687 (1984)). In the selection procedure described in Example IX, about 200 clonal cell colonies were screened. Of these, two high-producing strains produced apoAI at a level about 30 times that of the original CHO cells as a whole. In the high-producer cells, the proportion of apoAI in the secreted protein mixture is between about 30-50%.
  • LBP production by the CHO cells can be readily scaled up by culturing the cells in large-volume roller bottles, as described in Example XIX.
  • the CHO strain used here was a high-producing strain, and liquid volume in each bottle was 250 ml. As seen in Table I of the example. apoAI levels of up to 30 mg/liter/day can be achieved.
  • the apolipoproteins are processed and secreted in mature form.
  • Cellular processing of full-length (prepro) apoAI involves cleavage of an 18 amino acid leader sequence, to form the proapoAI, and further cleavage of 6 amino N-terminal residues to form the mature protein.
  • the bacterial, yeast, and mammalian-cell expression systems described above produce a mixture of proteins (peptides) which include a relatively small proportion of the desired heterologous LBP.
  • the LBP-containing mixture is in the intracellular protein fraction and produced by lysing the cells, and removing cellular debris.
  • yeast and mammalian-cell systems where the LBP is secreted, the protein mixture is in the extracellular protein fraction remaining after removing the cells from the reaction medium.
  • the number of heterogeneous sized proteins contained in the protein mixtures from the various expression systems can be seen from the SDS gel electrophoretic patterns of expressed proteins in Figures 3 and 5 (for E. coli).
  • Figure 7 for S.
  • the heterologous LBP gene product produced in an expression system can be purified conveniently and in large volume from the protein mixture by (a) adding lipid to the mixture, to form particulate lipoprotein complexes composed of lipid particles and associated LBP, and (b) separating the complex from nonlipid binding peptides in the mixture on the basis of the lower density or greater size of the complex.
  • the present section examines methods for forming such lipoprotein complexes in mixtures of peptides.
  • the lipid added to an LBP-containing protein mixture may be an oil emulsion, and typically a triglyceride oil emulsion containing a phospholipid emulsifier.
  • oil emulsions are commercially available for use in parenteral nutrition or may be prepared by emulsifying oil/phospholipid mixtures by homogenization or sonication, according to known procedures.
  • One standard oil emulsion is INTRALIPID, a commercially available triglyceride/phosphatidylcholine emulsion used for parenteral nutrition.
  • the emulsion typically has particle sizes in the 0.1 to 0.5 micron range.
  • the lipid emulsion is added to the LBP-containing protein mixture, and the suspension allowed to incubate for up to one or more hours with shaking.
  • the protein mixture is preferably concentrated to about 5-10 mg/ml before addition of lipids.
  • Lipid is added in a weight ratio of at least about 10:1 lipid/protein and preferably at a ratio of 50:1 or more.
  • the added lipid is phospholipid, usually taking the form of an aqueous suspension of uni- or multilamellar bilayer structures commonly known as liposomes.
  • a preferred liposome composition includes phosphatidylcholine (PC), such as egg PC, or a PC/cholesterol mixture.
  • PC phosphatidylcholine
  • the vesicle forming lipids are dried to a thin film, and hydrated slowly with an aqueous medium, forming a suspension of lipids.
  • the suspension can be further processed, for example, by homogenization or sonication, to reduce the size heterogeneity of the particles.
  • Example XVI below describes the preparation of a PC liposome suspension whose uni- and multilamellar structures are seen in the negative stain photomicrograph of Figure 16. Liposome particle sizes typically range from about 0.01 to 0.1 microns.
  • the liposome suspension is added at a weight ratio of preferably about 10:1 or more, and incubated for up to one or more hours.
  • the morphology of the lipoprotein. (LBP/phospholipid) complexes which form will depend on the nature of the apolipoprotein. Where the apolipoprotein is apoAI, the lipoprotein complexes include many disc-like structures, such as those shown in the photomicrograph of Figure 17. These structure strongly resemble the disc-like nascent high-density lipoprotein (HDL) particles found in liver perfusate and composed predominantly of phospholipid and apoAI and apoE.
  • HDL high-density lipoprotein
  • a third lipid source is endogenous lipids which are supplied by the expression system itself.
  • an expression system can supply lipid in an amount and form that is useful in producing particulate lipoprotein complexes with heterologous expressed lipid-binding proteins.
  • the lipids supplied by the system in the case of CHO cells, are presumably phospholipids which can combine with the expressed LBP to form characteristic LBP/phospholipid structures.
  • a comparison of the photomicrographs of Figures 13 and 17 indicates that the lipoprotein particles formed in the CHO system reported in Example XIII, involving apoAI expression, resemble the disc-like structures seen when purified apoAI is mixed with PC liposomes.
  • the particulate lipoprotein complex from above can be separated readily from nonlipid binding proteins in the expression system on the basis of either size or density.
  • the LBP/lipid complex is a particulate complex composed of lipid particles -- either emulsion, liposomal. or lipid-disc particles -- with associated LBP.
  • the lipoprotein particles are in the general size range of between about 0.01 to 0.55 ⁇ , and as such, can be readily separated from smaller, soluble proteins by particle exclusion in molecular sieve chromatography.
  • the molecular-sieve material is preferably selected to exclude material in the range 500,000 daltons or greater, insuring that essentially all of noncomplexed proteins will be included (retarded) by passage through the chromatography column bed.
  • the larger lipoprotein complex, which is excluded, will elute in the void volume of the column. The eluted fractions can be monitored, conventionally, by UV spectroscopy or the like.
  • Preferred molecular-sieve material includes Agarose 10%, whose exclusion size is about 1,000,000 daltons.
  • the lipoprotein complex is preferably separated from nonlipid binding peptides by flotation. Typically this is done by adjusting the density of the peptide-mixture medium to between about 1.06-1.21 by addition of a mono- or disaccharide, such as sucrose, or a salt, such as KCl. The medium is then placed in a centrifuge tube, overlayered with a lower density medium, and centrifuged until the complex has concentrated at the top of the tube. The lipoprotein material removed from the gradient may be resuspended in the same density medium and separated a second time, to further remove nonlipid-binding proteins.
  • Example XIV for the purification of apoAI. As seen in Figure 15, and discussed in Example XIV, the purification method produced about a 95% purification of apoAI, as judged by a gel density scan of the fractionated protein.
  • the lipoprotein complex will separate on its own by floatation if allowed to stand undisturbed in a medium of sufficient density.
  • This floatation procedure is, of course, advantageous in large-scale preparation, since both chromatography and centrifugation steps are eliminated.
  • the LBP associated with the separated lipoprotein complex may be further purified, as necessary, by delipidating the complex and purifying the peptide by conventional purification methods, such as described in Example XV.
  • the purification method just described is part of a general LBP-producing system in which recombinantly produced LBP is associated to particulate lipid and isolated from nonbinding lipids by simple floatation or molecular sizing procedures.
  • the method is more broadly applicable to purifying other proteins which have lipid-binding regions that promote protein association with particulate lipids.
  • proteins include lung surfactant protein, C-reactive protein, and fused LBP proteins of the type described above.
  • particulate lipid is added to a mixture of proteins produced in the expression system, to produce lipid-particle complexes with the heterologous lipophilic protein. The particulate lipoprotein complexes are then separated on the basis of size or density as described.
  • apoprotein components of the high-density lipoproteins (HDL) -- apoAI, apoAII, apoCI, apoCII, and apoCIII -- when reconstituted with phospholipid and then incubated in the presence of tissue culture cells, remove cholesterol from the cell membrane in vitro (Jackson, et al, J Biol Chem 250:7204 (1975)) and in vivo (Miller. N.E., et al. Nature 314:109 (1985)).
  • the complex formed by apoAI and phospholipid appears to be particularly effective in reverse cholesterol transport.
  • the lipoprotein complex formed by recombinant apoAI and PC liposomes resembles the disc-like structures of nascent HDL. This finding indicates that apolipoproteins formed in accordance with the invention retain their lipid-binding properties and that lipoproteins formed with the recombinant proteins would therefore have the same therapeutic uses, for reverse cholesterol transport, as HDL particles or nascent HDL particles formed with isolated serum apolipoproteins.
  • LCAT lecithin cholesterol acyl transferase
  • C-reactive protein synthesized at high levels in people stressed by inflammation, tissue injury, or infection (this group includes premature infants), may be enhancing the fusion of INTRALIPID lipid particles to particles of a larger size that are filtered out of the bloodstream by the lungs, sometimes leading to fat embolism and death.
  • Example XVII Studies conducted in support of the present invention, and reported in Example XVII show that serum from stressed animals causes a significant size increase in a major portion of INTRALIPID particles, whereas emulsion particle size is substantially stable in serum from unstressed animals (Table IV).
  • apoAI stabilized the emulsion particles against fusion and size growth in the presence of serum factor (s) (presumably including C-reactive protein) associated with stress.
  • the invention thus includes a stabilized lipid emulsion for parenteral nutritional use, composed of lipid emulsion particles and associated recombinant apolipoprotein, such as apoAI. From the foregoing it can be appreciated how various objects and features of the invention are met.
  • the invention provides a method for producing recombinant apolipoproteins, and lipid-binding peptides thereof, in a variety of expression systems, including E. coli, S. cerevisiae, and CHO cells.
  • apolipoproteins retain their native lipid-binding properties, as judged both by (a) their ability to bind to added lipid, either in the form of an oil/phospholipid emulsion or phospholipid vesicle suspension, (b) their ability to associate with and stabilize oil/phospholipid emulsion particles against fusion in the presence of fusogenic serum factor(s) and (c) their ability to interact with phospholipid vesicles, producing disc-like structures similar in appearance to nascent HDL particles.
  • the CHO cell expression system described herein in particular provides several advantages for large-scale production of apolipoproteins and their peptide fragments.
  • the system is inducible for heterologous gene expression, and high-producer clonal strains which are selected readily can increase heterologous gene expression by more than an order of magnitude and to a level in which the recombinant LBP constitutes 30%-50% of total secreted protein.
  • This cell system also has the ability to process full-sequence apolipoproteins to mature proteins, an ability that may require two separate peptide cleavages for apoAI and apoAII.
  • the lipid-binding capacity of the recombinant proteins is exploited in a simple, efficient method for producing purified apolipoprotein by recombinant methods.
  • the purity of the purified Al may be as high as 95% using only centrifugation steps.
  • the method is especially useful for producing apolipoproteins in large scale, in combination with the high-level CHO expression system.
  • the method of producing pure or nearly pure recombinant protein by lipid addition to recombinantly produced proteins is applicable to any lipid-binding recombinant protein which can associate stably with lipid particles.
  • Plasmids containing the desired coding and control sequences employs standard ligation and restriction techniques which are well understood in the art. Isolated plasmids, DNA sequences, or synthesized oligonucleotides are cleaved, tailored, and religated in the form desired.
  • Site specific DNA cleavage is performed by treating with the suitable restriction enzyme (or enzymes) under conditions specified by the manufacturer of these commercially available restriction enzymes.
  • plasmid or DNA sequence is cleaved by one unit of enzyme in about 20 ⁇ l of buffer solution; in the examples herein, typically, an excess of restriction enzyme is used to insure complete digestion of the DNA substrate. Incubation times of about one hour to two hours at about 37°C are workable, although variations can be tolerated. After each incubation, protein is removed by extraction with phenol/chloroform, and may be followed by ether extraction, and the nucleic acid recovered from aqueous fractions by precipitation with ethanol. If desired, size separation of the cleaved fragments may be performed by polyacrylamide gel or agarose gel electrophoresis using standard techniques.
  • Restriction cleaved fragments may be blunt ended by treating with the large fragment of E. coli DNA polymerase I (Klenow) in the presence of the four deoxynucleotide triphosphates (dNTPs) using incubation times of about 15 to 25 min at 20 to 25°C in 50 mM Tris pH 7.6, 50 mM NaCl, 6 mM MgCl 2 , 6 mM DTT and 5-10 ⁇ M dNTPs.
  • the Klenow fragment fills in at 5' sticky ends but chews back protruding 3' single strands, even though the four dNTPs are present.
  • selective repair can be performed by supplying only one of the, or selected, dNTPs within the limitations dictated by the nature of the sticky ends. After treatment with Klenow, the mixture is extracted with phenol/chloroform and ethanol precipitated. Treatment under appropriate conditions with S1 nuclease or exonuclease Bal-31 results in hydrolysis of any single-stranded portion.
  • Ligations are performed in 15-50 ⁇ l volumes under the following standard conditions and temperatures: 20 mM Tris-HCl pH 7.5, 10 mM MgCl 2 , 10 mM DTT, 33 ⁇ g/ml BSA, 10 mM-50 mM NaCl, and either 40 ⁇ M ATP, 0.01-0.02 (Weiss) units T4 DNA ligase at 0°C (for "sticky end” ligation) or 1 mM ATP, 0.3-0.6 (Weiss) units T4 DNA ligase at 14°C (for "blunt end” ligation).
  • Intermolecular "sticky end” ligations are usually performed at 33-100 ⁇ g/ml total DNA concentrations (5-100 nM total end concentration). Intermolecular blunt end ligations (usually employing a 10-30 fold molar excess of linkers) are performed at 1 ⁇ M total ends concentration.
  • vector construction employing "vector fragments” the vector fragment is commonly treated with bacterial alkaline phosphatase (BAP) or calf intestinal alkaline phosphatase (CIP) in order to remove the 5' phosphate and prevent religation of the vector.
  • BAP bacterial alkaline phosphatase
  • CIP calf intestinal alkaline phosphatase
  • Digestions are conducted at pH 8 in approximately 150 mM Tris, in the presence of Na and Mg using about 1 unit of BAP or CIP per ⁇ g of vector at 60° for about one hour.
  • the preparation is extracted with phenol/chloroform and ethanol precipitated.
  • religation can be prevented in vectors which have been double digested by additional restriction enzyme digestion of the unwanted fragments.
  • the desired sequences are thus recovered from colonies responding to probe.
  • chloramphenicol amplification optionally following chloramphenicol amplification (Clewell, D.B., J Bacteriol 110:667 (1972)).
  • the isolated DNA is analyzed by restriction and/or sequenced by the dideoxy method of Sanger. F., et al, Proc Natl Acad Sci (USA) 74:5463 (1977) as further described by Messing, et al, Nucleie Acids Res 9:309 (1981), or by the method of Maxam, et al. Methods in Enzymology 65:499 (1980).
  • Plasmid pPSAI.2 was constructed by isolating the 2.2 kb PstI fragment and inserting it into the PstI site of pBR322 (Maniatis, T., et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, NY (1982)). The pPSAI.2 insert was removed by PstI digestion and isolated by gel electrophoresis.
  • the isolated insert (10 ⁇ g) was digested with 5 units of StuI, and the 1065 base pair StuI fragment, which includes the part of the apoAI gene region that encodes amino acids 110-187 was isolated and digested with Narl, as shown in Figure 2.
  • the 5' overhang resulting from Narl digestion was blunt-ended by incubating the DNA with the Klenow fragment of DNA polymerase I in K.L. Buffer (10 mM Tris-HCl, pH 8.0. 5 mM MgCl 2 ), and 0.8 mM each dATP, dTTP, dCTP, and dGTP for 5 hours at 17°C.
  • the self-complementary oligonucleotide #2A-14 (5'-dCCTGAAGCTTCAGG) which contains translation stop signal (TGA) and Hindlll site (AAGCTT) was ligated to both ends of the blunt-ended fragment by incubating with 50 units T4 DNA ligase in K.L. Buffer with 10 mM ATP for 12 hours at 12°C. The linkers were cut back with Hindi II and the fragment was subcloned into the HindIII site of pKT19 (Talmadge. K., et al, Gene 12:235 (1980)) to give the plasmid pPSAI.4 ( Figure 2).
  • the pPSAI.4 plasmid (10 ⁇ g) was cut with 2 units of MboII for 2 hours at 37°C, and the resulting 3' overhang was removed by treatment with T4 DNA polymerase (Maniatis, et al, supra).
  • the self-complementary oligonucleotide #1A--10 (CATGGCCATG) which contains a translation initiation signal (TGA) was ligated to the MboII cut, blunt-ended DNA fragments, cut with Ncol and Hindlll and ligated into pBR329 (catalogued in the National Institutes of Health, Bethesda, MD data base of vectors and gene sequences) which had been cut with Ncol and Hindlll (see Figure 2).
  • the ligation mixtures were used to transform E. coli HB101 cells as described above. Plasmids with the correct insert were identified by restriction mapping.
  • the resulting plasmid, pPSAI.6 includes an Ncol/Hindlll fragment with the apoAI gene fragment encoding amino acids 110-187 in frame with an ATG translation initiation signal at the 5' end and a TGA translation stop signal at the 3' end.
  • the Ncol/Hindlll insert of pPSAI.6 was isolated and ligated into (a) the expression vector pKK233-2 (Brosius. J., et al.
  • trc contains the upstream portions of the trp promoter and the downstream, operator-containing, regions of the lac promoter and was originally prepared from two readily available plasmids containing these promoters.
  • an intermediate plasmid pKK10-0 was prepared containing the hybrid promoter.
  • pEA300 (Amman. E., et al, Gene (1983) 25 :167-178) was digested with PvuII and Clal, filled in using dCTP only in the presence of DNA polymerase (Klenow), followed by digestion with mungbean nuclease, and the large vector fragment isolated.
  • This vector fragment contains the upstream portions of the trp promoter.
  • the fragment was ligated with a 55 bp blunt-ended Hpall/PvuII digest excised from pGL101 (Lauer.
  • the resulting plasmid, pKK10-1 was digested with PvuII, and ligated to the Ncol linker, 5'-ACCATGGT-3'. digested with Ncol, filled in, and then ligated to a double-stranded linker containing PstI and Hindlll sites provided as two complementary oligonucleotides, 5'-GCTGCAGCCAAGCTTGG-3' and its complement. The ligation mixture was used to transform E. coli to Amp R .
  • the isolated plasmid DNA was digested with BamHI and Hindlll, and the small BamHI/HindlII fragment obtained on electrophoresis contains the trc promoter. To complete pKT52.
  • the BamHI/HindlII fragment containing the trc promoter was ligated into the large fragment obtained from BamHI/HindlH digestion of pKK10-2 (Brosius. J., Gene (1984) 27:161-172) which contains the Amp R gene and the origin of replication.
  • the resulting plasmid, pKK233-1 was digested to completion with Pvul and then partially with Bgll and ligated with the 360 bp Pvul/Bgll fragment containing the corresponding portion of the ampicillin resistance gene but lacking a PstI site from pUC8.
  • the ligation mixture was used to transform E. coli and transformants were screened for the presence of only one PstI site next to the trc promoter.
  • pKK233-2 The correct construction, pKK233-2. was digested with EcoRI and PvuII, filled in with dATP and dTTP, and religated to obtain the correct construction pKT52.
  • pKT52 contains the desired trc promoter, a downstream ATG start codon. and downstream Ncol, PstI and Hindlll sites.
  • E. coli JA221 (Nakamura. K., et al, J Mol Appl Genet 1:289 (1982)) (lpp-/F'lacI Q ) containing plasmid pKK233-2, pAP85 or pApoF were grown in M9 medium (Miller, J., Experiments in Molecular Genetics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1972) supplemented with glucose (2 mg/ml). thiamine (10 ⁇ g/ml). MgSO 4 -7H 2 O (250 ⁇ g/ml).
  • the dried TCA pellet was resuspended in 40 ⁇ l of 50 mM Tris-HCl. pH 6.8, 1 mM EDTA, and 1% (w/v) SDS. Ten ⁇ l of this resuspension was added to 10 ⁇ l of 2 x SDS gel sample buffer (125 mM Tris-HCl, pH 6.8. 20% glycerol. 2% SDS, 2% ß-mercaptoethanol. and 0.1% bromophenol blue), and incubated at 100°C for 2 minutes.
  • 2 x SDS gel sample buffer 125 mM Tris-HCl, pH 6.8. 20% glycerol. 2% SDS, 2% ß-mercaptoethanol. and 0.1% bromophenol blue
  • a unique polypeptide migrating at a molecular weight of approximately 4500 daltons appears in lanes 2 and 3 which are protein patterns from cells containing plasmids pApoF and pAP85. respectively.
  • Cells containing plasmid pKK233-2 (lane 1) do not have this unique peptide.
  • the estimated molecular weight of the apoAI fragment peptide based on amino acid composition is 8,700 daltons. The discrepancy between measured and expected molecular weight may be due either to the lipophilic nature of the protein, which may effect its binding to SDS, or to a processing event which is taking place in the cell.
  • This unique polypeptide is specifically iramunoreactive to serum raised against apoAI ( Figure 3, lanes 5 and 6). ApoAI antiserum does not react with any unique polypeptides extracted from cells containing plasmid pKK233-2 (lane 4).
  • Example III Bacterial Vector with Al cDNA Plasmid pBL13AI containing a full-length Al cDNA was prepared as described in Seilhamer, J.J., et al, supra. The plasmid (100 ⁇ g) was digested to completion with EcoRI for 3 hr at 37°C. The 965 base pair EcoRI fragment containing the apoAI cDNA was isolated on a 4% (w/v) nondenaturing polyacrylamide gel (Maniatis, et al, supra). The apoAI cDNA fragment was excised from the gel, electroeluted, and concentrated by ethanol precipitation. The resulting DNA pellet was dried in vacu and resuspended in H 2 O.
  • This EcoRI fragment was further digested with Sau3A for 30 min at 37°C with a DNA-to-enzyme ratio of 1 ⁇ g : 2 units.
  • the resulting Sau3A fragments were separated as described above.
  • the 783 base pair partial fragment was isolated and concentrated as described above.
  • the oligonucleotides were kinased with 32 P-ATP using T4 polynucleotide kinase (Maniatis, et al, supra). The two oligonucleotides (1 ng each) were mixed, boiled for 2 min, and allowed to hybridize at RT for 60 min. The hybridized oligonucleotides were ligated to 25 ng of Sau3A partial apoAI cDNA fragment with the addition of T4 DNA ligase by the method of Maniatis. et al, supra. After ligation. the mixture was digested to completion with EcoRI and Ncol. Products were separated on a 5% nondenaturing polyacrylamide gel.
  • the 811 base pair fragment corresponding to full-length coding sequence of mature apoAI protein was excised.
  • the DNA was eluted from the gel slice in 10 mM Tris-HCl, pH 8, 1 mM EDTA, 0.4 M NaCl with agitation at 37°C overnight and concentrated by ethanol precipitation.
  • the NcoI-EcoRI fragment was then ligated into pBR329 vector.
  • the resulting plasmid. shown in Figure 4, is designated pFLAI-2.
  • an Rsal fragment of pFLAI-2 encompassing the first 56 base pairs of the coding region for mature apoAI, as well as 112 base pairs downstream into pBR329 was sequenced by the dideoxy method (Messing, J., et a,. Gene 19:259 (1982) and Sanger. et al. Proc Natl Acad Sci (USA) 74:5463 (1977)).
  • the NcoI-EcoRI construct was the ligated into the bacterial expression vector pKK233-2 of Figure 2.
  • Plasmid pFLAI-2 (10 ⁇ g) was digested to completion with EcoRI and the ends filled in the method of Maniatis et al, supra, with E. coli DNA Polymerase I, Klenow fragment, and the addition of 0.5 mM dATP and dTTP. The blunted DNA was then phenol/chloroform extracted, and concentrated by ethanol precipitation. The blunted DNA was further digested to competition with Ncol. The 811 base pair fragment containing the coding region of mature apoAI was purified on a 5% nondenaturing acrylamide gel and concentrated as described above.
  • Ncol blunt fragment was then ligated using T4 DNA ligase (Maniatis et al, supra) into pKK233-2 which was cut with Hindlll, blunted, phenol extracted, and cut with Ncol.
  • the resulting plasmid is designated pFLAI-10 ( Figure 4).
  • Example IV Expression of Full-Lenqth ApoAI in E. coli E. coli JA221 (lpp-/F'lacI Q ) containing plasmids pKK233-2 or pFLAI-10 (Example III) were grown and labelled in M9 medium (Miller. J., Experiments in Molecular Genetics, Cold Spring Harbor Laboratory, Cold
  • the pellet was resuspended in 50 ⁇ l of 5 mM
  • Example II and incubated 100°C for 2 min. A second aliquot equal to 2 x 10 6 cpm was brought up to 1 ml total volume by the addition of 5 mM Tris-HCl, pH 6.8, 0.15 M NaCl. 0.1 mM EDTA, and 2% (v/v) Triton X-100. 2 ⁇ l of rabbit antiserum raised against human apoAI was added to the suspension. The suspension was incubated at room temperature for 30 min and further incubated on ice overnight. Following incubation, 50 ⁇ l of a 10% (w/v) Protein A Sepharose CL-4B (obtained from Sigma, St. Louis.
  • FIG. 5 A comparison of polypeptides extracted from cells containing plasmids pKK233-2 and pFLAI-10 is shown in Figure 5.
  • Commercially prepared mature apoAI protein (Calbiochem-Behring. La Jolla. CA) also migrates at 25.000 daltons, identically to the bacterial product. It should be noted that the calculated molecular weight of mature apoAI protein is 27,000 daltons. but the hydrophobic nature of this protein causes it to migrate at an apparent molecular weight of 25,000 daltons in this gel system.
  • This unique polypeptide is specifi- cally immunoreactive to serum raised against apoAI (lane 5). ApoAI antiserum does not react with any unique proteins from cells harboring plasmid pKK233-2 (
  • Example V Yeast Vector with ApoAI cDNA
  • E. coli-yeast shuttle vector YEp13 Nasmyth. K., et al, Cell 19:753 (1980)
  • YEp13 a yeast library in the E. coli-yeast shuttle vector YEp13 (Nasmyth. K., et al, Cell 19:753 (1980)) was screened using a 5'- 32 P end-labeled oligodeoxynucleotide 5'-dCCTGGCCAACCAATG-3' (Maniatis et al, at pp. 324-325). Plasmids containing inserts of yeast DNA hybridizing to this oligonucleotide were subsequently isolated.
  • One of these plasmids contained an insert of approximately 15 kb of yeast DNA, and was shown to contain the 1.7 kb EcoRI fragment containing the alpha-factor gene as described by Kurjan and Herskowitz (Kurjan. J., et al. Cell 30:933 (1982)).
  • the end of the 1.7 kb EcoRI fragment were made blunt by incubation with DNA Polymerase I (Klenow fragment) and BamHI linkers using T4-DNA ligase (Maniatis, et al, at pp. 113-114, 116, 392-394).
  • the BamHI ends were made cohesive by digestions with Bam HI restriction endonuclease, subsequently ligated into the BamHI site of the yeast-E.
  • Hindlll ends were made cohesive by digestion with Hindlll endonuclease and ligated into Hindlll-cleaved YEp- ⁇ -8 which had been treated with alkaline phosphatase to remove its 5' phosphate moieties (Maniatis, et al, supra, pp. 366-369). With insertion into the yeast vector, amino acid number eight of mature apoAI is restored by the Hindlll linker, allowing for coding of amino acids 8-243 of mature apoAI.
  • Expression vector YEp- ⁇ -aA-11 ( Figure 6) with the apoAI insert in the correct orientation encodes a chimeric protein consisting of factor signal/leader peptide and almost the entire mature apoAI sequence (amino acids 8-243).
  • Expression vector YEp- ⁇ -aA-5 with the insert in the reverse orientation encodes a chimeric protein with the same signal/leader peptide and a protein unrelated to apoAI.
  • Example VI Expression of ApoAI Full-Length cDNA in S. cerevisiae DNA was prepared from E. coli cultures containing the plasmids YEp- ⁇ -8.
  • YEp- ⁇ -aA-11, and YEp- ⁇ -aA-5 (Example V) and was used to transform yeast strain W301-18A ( ⁇ ade 2-1. trp 1-1, leu 2-3, -112, can 1-100, ura 3-1, his 3-11, -15) (Kramer, et al, Proc Natl Acad Sci (USA) 81:367 (1984)) to Leu 2 prototrophy.
  • Yeast strains were grown on standard media (Sherman et al. Methods in Yeast Genetics. Cold Spring Harbor Press. Cold Spring Harbor, New York). Plasmid DNA from E.
  • the supernatant was incubated on ice for 30 min, centrifuged for 15 min at 4°C in an Eppendorf centrifuge at 15,000 x g, and the pellet was washed with 1 ml of ice cold acetone. The final is dried in vacu. resuspended in 50 mM Tris-HCl, pH 6.8. 1 mM EDTA, 1% (w/v) SDS, and incubated at 100°C for 3 min. 10 ⁇ l of this resuspension was added to 10 ⁇ l of 2x SDS gel sample buffer.
  • FIG. 7 A comparison of polypeptides extracted from the media of S. cerevisiae cultures carrying the expression constructs is shown in Figure 7.
  • a unique polypeptide migrating at a molecular weight of approximately 23,000 daltons appears in cultures of yeast carrying the alpha factor expression vector with the apoAI cDNA insert in the correct orientation (Y9-2, lane 3).
  • This unique protein is not present in yeast carrying the ⁇ factor expression vector only (Y9-1) or the apoAI cDNA insert in the wrong orientation (Y9-3) (lanes 1 and 5, respectively).
  • This unique protein is specifically immunoreactive with rabbit antiserum raised against human apoAI (lane 4). Proteins from the other yeast cultures Y9-1 and Y9-3 have no specific reaction with apoAI antiserum (lanes 2 and 6.
  • the recombinant ⁇ factor-apoAI cDNA expression vector (YEp- ⁇ -aA-11) encodes for an apoAI protein 7 amino acids shorter from the N-terminus than mature apoAI (only amino acids 8-243 are encoded). This accounts for the fact that the apoAI product synthesized in yeast cultures migrates ahead of mature apoAI produced by bacterial expression (Example IV and Figure 5) and by mammalian-cell expression (Example VIII and Figure 9 below).
  • a hybrid gene was constructed in which the coding segment for apoAI was fused to a powerful regulated promoter derived from the human metallothionein II (hMT-II) gene. This was performed in two steps, which are illustrated in Figure 8.
  • the expression vector, pHSI carried 840 nucleotide base pairs of hMT-II sequence (Karin, M., et al. Nature 299:797 (1982)) from a naturally occurring Hindlll restriction site at base -765 from the start of transcription to base +70, located in the 5' untranslated region adjacent to the coding region.
  • pHSI also carries a region into which coding sequences may be inserted .
  • the plasmid p84H (Karin, et al, supra), which carries the hMT-II gene, was digested to completion with restriction endonuclease BamHI followed by treatment with exonuclease Bal-31 to remove terminal nucleotides. Following digestion with Hindlll. the products of this reaction were ligated into plasmid pUC8 (Messing, J., et al, supra) which had been opened with Hindlll and Hindi digestion.
  • One of the resulting plasmid recombinants had the composition of pHSI as determined by nucleotide sequencing.
  • the PstI fragment spanning the apoAI gene was isolated from plasmid pPSAI.2 (Example I) by digestion with PstI followed by polyacrylamide gel purification.
  • the PstI fragment extends from a point in the 5' untranslated region through the entire coding sequence (with introns) and terminates beyond the poly A addition site.
  • the purified fragment was blunt ended by digesting away the single-stranded 3' termini with T4 DNA polymerase in the presence of dCTP.
  • the flush-ended molecules were then ligated to the expression vector plasmid pHSI, which had been opened by restriction with Sraal. followed by treatment with bacterial alkaline phosphatase.
  • pMTAIR The products of the reaction were introduced into E. coli MC1061, and the recombinant. pMTAIR was identified. In the resulting plasmid. pMTAIR, the apolipoprotein Al coding sequences are in position to be expressed by hMT-II promoter. The plasmid is contained in the deposited CHO cells, CRL #8911.
  • Example VIII Al Synthesis in CHO Cells Plasmid pMTAIR from Example VII was introduced into the Chinese hamster ovary (CHO:KI) line of cultured cells (growing in McCoy's 5A medium with 10% fetal bovine serum) by co-transformation with pSV2:NEO (Southern, P., et al, J Mol Appl Genet 1:327 (1982)). a plasmid carrying a functional gene conferring resistance to the neomycin analog G418. Five hundred ng of pSV2:NEO and 5 ⁇ g of pMTAIR were applied to a 60 mm dish of cells in a calcium phosphate-DNA co-precipitate according to standard protocols (Wigler. M., et al.
  • CHO:KI cells transformed with either plasmid pMT401 (control, hMT-II plasmid with no foreign insert) or pMTAIR were grown to 70% confluency in 9.6 cm 2 wells in standard medium (RPMI plus 10% dialyzed fetal bovine serum). Cells were preinduced with 1.5 x 10 -4
  • FIG. 9 A comparison of polypeptides extracted from medium of CHO cells transformed with the recombinant plasmids pMT401 and pMTAIR is shown in Figure 9.
  • a unique polypeptide migrating at molecular weight of 25,000 daltons appears in the medium from cells transformed with pMTAIR (lane 4). This unique protein is specifically immunoreactive to serum raised against apoAI (lane 5).
  • lane 5 There are no unique polypeptides present in lane 2, proteins extracted from cells transformed with pMT401, nor do any proteins immunoreact with apoAI antiserum, lane 3.
  • Commercially prepared mature apoAI co-migrates with the mammalian expression product, and both migrate an an apparent molecular weight of 25,000 daltons due to the lipophilic nature of apoAI and the gel system used.
  • Protein in Figure 9 represents apoAI being made from a transformed pool of CHO:KI cells (CHO/pMTAIR).
  • Example VIII was plated at low density (100-200 cells/ml) in standard medium (DMEM 21/Coon's F-12 plus 10% FBS), producing individual clonal colonies after 4-7 days growth at 37°C. The colonies were separately picked and grown in the above medium to a cell density of about 10 6 cell/ml.
  • standard medium DMEM 21/Coon's F-12 plus 10% FBS
  • the clonal cultures were individually assayed for Al expression by dot-blot Western, using the method described in Jahn, et al, Proc Natl Acad Sci (USA) 81:1684 (1984). Individual clones were seeded at 25% confluency in 12 well dishes in 1.5 ml DMEM21/Coons F12 plus 10% FBS. After 24 hr, the cells were washed once with 1 ml PBS and fresh medium containing 1 x 10 -4 M
  • Zn sulfate was added to begin preinduction.
  • the cells were washed twice with 1 ml PBS and refed with 0.65 ml serum-free medium containing 3 x 10 -5 M Zn sulfate and 3 x 10 -5 M Fe sulfate.
  • the media was harvested and centrifuged at 1000 rpm for 5 min to remove cell debris.
  • 0.5 ml serum-free conditioned medium was applied to each well onto a nitrocellulose filter, (Schleicher & Schuell, Keene, NH) using a dot-blot microfiltration apparatus (Bio-Rad
  • Lanes 1 and 3 are total S 35 -methionine-labeled secreted proteins from
  • CHO pMTAIR cells of clone 104.
  • apoAI secreted from clone 104 is easily identifiable in the total secreted medium proteins without immunoprecipi- tation. This clone is producing apoAI at approximately a 30-fold higher level than the pool cells.
  • Example IX producing the same level of apoAI as clone 104 and designated clone 143, was used for large-scale Al production in roller bottles.
  • the clone 104 is the high-producing CHO cell line identified as CRL 8911.
  • a 10 cm plate of clone 143 was grown to confluence in 10 ml DMEM-21/Coon's F-12 (Gibco, Irvine, CA) plus 10% fetal bovine serum (Gibco) medium.
  • the cells in the plate were washed once with 2 ml phosphate buffered saline (PBS), trypsinized.
  • PBS phosphate buffered saline
  • DMEM-21/Coon's F-12 medium containing 10% FBS and 15 mM HEPES buffer in an 850 cm 2 roller bottle.
  • zinc sulfate was added to a final concentration of 1 x 10 -4 M.
  • the cells were switched to serum-free DMEM-21/Coon's F-12 medium containing 6 x 10 -5 M zinc sulfate and 3 x 10 -5 M iron sulfate, marking the beginning of the production period.
  • the cells were cultured for 2 days following the medium switch (day 2 in Table I), and the conditioned medium harvested by low-speed centrifugation to separate cell debris from spent medium.
  • the cell medium was fractionated by SDS gel electrophoresis, as in Example VIII. and the gel. after staining with Coomassie Brilliant Blue, was scanned optically to quantitate the amount of apoAI protein in the cell-free medium, using a purified Al standard obtained from
  • ⁇ AII had a 440 base EcoRI insert corresponding to the full-sequence AH cDNA (Sharpe. et al, supra) plus ⁇ 20 bases of 5' untranslated region.
  • the EcoRI insert was isolated from the phage and cloned into pHSI.
  • Lanes 1, 2, and 3 are cell-free proteins from CHO cells transformed with pHSI (control), pHSI/AII, and pHSI/incorrect, respectively.
  • pHSI/AII transformed cells produce two low-molecular peptides of about 6,000-8,000 dalton molecular weight which are not seen in the two controls.
  • the larger of the two pHSI/AII bands may be pro AH (78 amino acids), the other being mature AH (73 amino acids) or the different molecular weights may represent different degrees of glycosylation.
  • Example XII Cloning and Expression of Al Fragment in CHO Cells An expression system capable of producing apoAI (110-187) in CHO cells is constructed as described here.
  • the plasmid pAP85 containing the apoAI (110-187) fragment is cut with Ncol and Hindlll and the insert containing the DNA coding for Met-apoAI (110-187)-Stop is isolated.
  • the insert is blunted and ligated to the BamHI-NcoI adapter 5'-CATGGGATCC and the BamHI-Hindlll adapter 5'-AGCTGGATCC at the 5' Ncol site and the 3' Hindlll site, respectively.
  • the DNA adapters are cut back with BamHI and the modified insert is purified by gel electrophoresis.
  • the BamHI fragment is then ligated to the BamHI site of pMT apoAI(BS), the metallothionein expression plasmid which contains the 3' untranslated region of apoAI which includes a transcription termination signal, as described in detail in co-owned patent application Serial No. 680,358, and incorporated herein by reference.
  • the plasmid is formed by digesting the pMTAIR vector from Example VII (on deposit) first with BamHI. and after blunt-ending with Klenow fragment, further digesting with StuI and treating with CIP. The ligation produces the desired pMT apoAI (BS) vector.
  • the plasmid containing the insert in the correct orientation can be determined by restriction mapping.
  • the plasmid containing the insert in the correct orientation is used to transfect CHO cells and high-producing clones selected as described.
  • Example XIII ApoAI Separation/Endogenous Lipid Culture medium from the CHO:pMTAIR clone 143 cells in Example X was used.
  • the medium was adjusted to a density of 1.125 g/ml by addition of solid potassium bromide, then centrifuged at 38K rpm for 18 hours in a swinging-bucket rotor.
  • the top fraction was then removed by a conventional slicing method, and dialyzed extensively against saline.
  • the material when examined by SDS gel electrophoresis as in Example VIII, gave a prominent band in the 25,000 molecular weight range, corresponding to apoAI. Between about 10%-20% of the total apoAI produced by the CHO:pMTAIR cells was in the upper fraction, as judged by the relative staining intensities of the 25,000 molecular weight bands on SDS gels from the upper and lower centrifugation bands.
  • Electron micrograph of the top-fraction material shows the presence of numerous disc-shape structures characteristic serum of apoAI/phospholipid complexes, as shown in Fig. 13 (described in Hamilton, R.L., et al, J Clin Invest 58:667 (1976)).
  • Example XIV ApoAI Separation/Added Lipid Emulsion INTRALIPID obtained from Cutter Labs (Berkeley, CA) was used as the substrate to bind the apolipoprotein ligand.
  • INTRALIPID is an artificial lipid emulsion composed of soybean triacylglycerol and egg lecithins.
  • the mesophase, or phospholipid-rich portion of the emulsion was removed by ultracentrifugal floatation in a discontinuous sucrose gradient.
  • the bottom layer contained 2 ml of emulsion, 0.6 g sucrose and saline to give a final volume of 4 ml and a density of 1.06 g/ml.
  • the triglyceride rich emulsion on the top of the gradient was separated from the infranatant solution by the tube slicing technique using a Beckman slicer.
  • High-producing CHO:pMTAIR clone 143 from Example IX and control CHO:pMT401 cells were cultured in roller bottles, as described in Example X.
  • the culture medium was concentrated 100 times by ultrafiltration using an Amicon YM 10 membrane.
  • the concentrated medium was incubated with purified INTRALIPID and centrifuged as described for INTRALIPID except that only one step centrifugation was used. Lipid-to-protein ratios of either 0.1, 0.3, or 0.6 ml of concentrated medium per ml INTRALIPID were used.
  • the unbound fraction contains some apoAI and other contaminating proteins secreted from the CHO cells.
  • Example XV Al Purification The Al enriched, delipidated protein precipitate from Example XIV was dissolved in 0.01 M Tris-HCl, pH 8.3, buffer made in 6 M urea. A 50 ⁇ l aliquot was injected in a high-performance liquid chromatography system (HPLC) with a C 1 8 column equilibrated in a 20% acetonitrile, 0.1% trifluoroacetic acid (TFA) solution. After injection the sample was eluted with a gradient of acetonitrile from 20 to 70% containing 0.1% TFA at 2 ml/min flow rate. The elution profile, seen in Figure 14, shows a single major peak.
  • HPLC high-performance liquid chromatography system
  • TFA trifluoroacetic acid
  • the first 39 amino acids of the purified protein was sequenced by conventional methods. Table III below gives the results to the first 8 amino acids of the mature protein.
  • the major sequence, about 95% of the protein, is that of mature apoAI.
  • the minor (5%) sequence corresponds to the proapoAI having six additional N-terminal amino acids.
  • the sequence shows that the apoAI secreted into the medium is secreted as a processed mature apoAI and not as a pro Al.
  • the first 39 amino acids were sequenced and found to be all correct for mature native apoAI.
  • Figure 16 which is a negative-stain electron micrograph of the liposome preparation shows a mixture of unilamellar and multilamellar structures.
  • apoAI was incubated with the PC liposomes in a weight ratio of 1:5 at 37°C for 1 hour. After incubation, apoAI bound to liposomes was separated from free apoAI by gel filtration on 10% Agarose column. This material was then analyzed by negative-stain electron microscopy.
  • Figure 17 shows (a) the many disc-like structures which have formed in the presence of Al, and (b) the disappearance of more of the clearly identifiable lamellar vesicle structure seen in Figure 16.
  • the disc-like structures are similar in appearance to nascent HDL-like particles isolated from liver perfusate (see Hamilton. R.L., et al, supra).
  • Example XVII Stabilized Lipid Emulsion The emulsion was mixed with purified Al protein from Example XVI, at a wt ratio of 100:2 mg emulsion lipid/mg Al. The mixture was incubated with shaking at 37°C for 1 hour, to bind apoAI to the emulsion. Unbound apoAI was removed by centrifugation. The stability in serum of the lipid emulsion with and without apoAI was compared. Serum from rats treated with turpentine (5 ml/kg) or from control rats treated the same way but with saline were used. In each test, 180 ⁇ l of serum was incubated with 20 ⁇ l of lipid emulsion for 2 hours at 37°C with gentle shaking. After incubation, lipid-particle diameters were determined by laser light scattering using a sub-micron particle analyzer with optional size distribution processor analysis and multiple scattering angle detection (Coulter Model N4, Hialeah. FL).
  • the results are shown in Table IV below.
  • the lipid emulsion without apoAI was unstable in serum from rats treated with turpentine.
  • the emulsion being characterized by a bimodal distribution of sizes centered around 200 and 500 nm.
  • the emulsion containing apoAI showed no significant size change on exposure to serum obtained from turpentine-treated animals. Both emulsions were size stable in serum from control rats.
  • the filters were prewashed for 2 hr in 3 x NaCl/Cit (1 x NaCl/Cit is 150 mM NaCl/15 mM sodium citrate, pH 7.0), 0.1% SDS at 55°C. and then prehybridized in 6 x NaCl/Cit, 200 ⁇ g/ml denatured salmon sperm DNA, 5 x Denhardt's, 0.05% sodium pyrophosphate for 1 hr at 50°C.
  • a 192-fold degenerate 23 base oligonucleotide probe which encodes, taking account of codon redundancy, the first 8 amino acids of the previously determined sequence of apoB-26 was used as a probe.
  • the probe was
  • LB25-1 One positive plaque, designated LB25-1, was purified and the cDNA insert was subcloned in both orientations into M13/mp8 for sequencing.
  • the nucleotide sequence of this 970 bp insert is shown in
  • FIG 18 along with the deduced amino acid sequence of that reading frame which agrees with the amino terminal sequence determined from the B-26 peptide.
  • the sequence of LB25-1 contains an open reading frame extending 800 nucleotides downstream encoding 294 amino acids, and analysis of the predicted protein sequence directly upstream suggests the presence of a hydrophobic signal sequence preceded by a methionine residue.
  • the EcoRI insert was subcloned into pBR322 to obtain pLB25-1 for amplification. pLB25-1 thus contains some 5' untranslated region, the 28 amino acids of the signal sequence, and the first 266 amino acids of the mature protein.
  • Additional portions of the apoB encoding sequence were obtained by preparing a 2 x 10 5 member human adult intestine cDNA library in ⁇ gt10, as described above.
  • the approximately 1 kb insert of pLB25 was denatured and used as probe to isolate a cDNA fragment designated IB7, containing an approximately 1.3 kb insert, about 800 bp of which extended beyond the 3' end of clone pLB25.
  • Isolated, denatured IB7 insert was subcloned into pBR322 for amplification, creating pIB7.
  • the purified pIB7 insert was denatured and used to screen the intestine library.
  • I10 contained an approximately 3 kb insert, about 2.5 kb of which extended beyond the 3' end of IB7.
  • the cDNA insert was subcloned into the EcoRI site of pBR322, creating pB10.
  • Linearized, denatured pB10 insert was used as a probe to obtain a fourth cDNA fragment designated IB-(2)1, containing an approximately 2 kb insert, about 1 kb of which extends beyond the IB-10 sequence.
  • the EcoRI cDNA insert was also subcloned into the EcoRI site of pBR322, creating pB(2)1.
  • the 4 overlapping clones are digested to produce a single cDNA, as follows.
  • a 479 bp EcoRI-SacI fragment from the pBL25 insert, and a 1162 bp SacI-EcoRI fragment from the pBI7 insert, are ligated into the EcoRI site of pBR322.
  • the resulting plasmid is called pB25-7.
  • the larger fragment contains 1342 bp encoding the signal sequence and first 427 amino acids of apoB.
  • TWO synthetic oligonucleoties oligo 1 with the sequence 5'-AATTCTGAATGATTGAG-3' and oligo 2 with the sequence 5'-TCGACTCAATCATTCAG-3', are synthesized by standard methods using commercially available reagents, and hybridized by the method of Rossi et al, J Biol Chem 257:9226-9229 (1982). The resulting fragment has the sequence: oligo 1 5'-AATTCTGAATGATTGAG oligo 2 GACTTACTAACTCAGCT-5'.
  • This fragment has an EcoRI overhang, stop codons in all 3 frames, and Sail overhangs.
  • the EcoRI insert isolated from pBI10, is ligated to the synthetic fragment. The insert now has stop codons and Sail overhangs at both ends.
  • This insert is cloned into the Sail site of pBR322 for amplification, creating p10/stop.
  • the larger Sall-Aval fragment contains 2733 bp encoding the carboxy-terminus of the cloned apoB protein portion.
  • the expressed protein terminates (with one extra amino acid due to the insertion of the synthetic fragment) in frame after amino acid 1299.
  • the amino-terminal EcoRI-AvaI fragment isolated from pB25-7, described above, and the carboxy-terminal Aval-Sail fragment from p10/stop, also described above, are ligated into EcoRI-Sall digested pBR322.
  • the resulting vector encodes the signal sequence and the first 1299 amino acids of the mature apoB.
  • the amino acid sequence is identical to the first 1299 apoB amino acids shown in Figure 18 and includes an additional C-terminal aspartic acid residue due to the synthetic fragment.
  • the EcoRI/Sall fragment is then excised and placed in a suitable expression vector, like those described above, for expression of the apoB N-terminal portion.
  • the insert for the intestine cDNA clone 1(2)1 was made radioactive and used as a probe to screen the human intestine library as described previously (Protter, et al. PNAS 83, 5678-5682 (1986)).
  • a clone 1(3)13 was plaque purified and the nucleotide sequence of the insert DNA obtained as described previously.
  • a clone 1(7)1 was plaque purified and the sequence of the insert determined.
  • FIG. 19 The insert for the intestine cDNA clone 1(2)1 ( Figure 19A) was isolated by agarose gel electrophoresis. cut with BstXI and the large fragment purified.
  • the resultant plasmid pBI (2)1/(3)13 has the restriction map shown in Figure 19C.
  • the plasmid pBI (2)1/(3)13 was partially cut with EcoRI and the insert of the intestine cDNA clone 1(7)1 was isolated by cutting with EcoRI and preparative agarose gel electrophoresis. The two pieces of DNA were ligated and used to transfect competent HB101 bacteria. The plasmid with the correct orientation of EcoRI fragment can be identified by restriction mapping. The resultant clone pBI(2)1/(3)13/(7)1 is shown in Figure 19E.
  • oligo 3 (5'-AATTCTGAATG-3') and Oligo 4 (5'-TCGACATTCAG-3') in the construct.
  • the plasmid pBI (2)1/(3)13/(7)1 was partially digested with EcoRI, partially digested with Sail, and then ligated to the oligos oligo 3 and oligo 4.
  • the reaction was then transfected into competent HB101 bacteria and the resultant colonies were screened for the correct construct with the oligos, oligo 5 (5'-ATTCCAGAATTCTGAATGTCGA-3') and oligo 6
  • the plasmid above contains the EcoRI-Aval fragment isolated from pB25-7 and the carboxy-terminal Aval-Sail fragment from p10/stop in the EcoRI-Sall site of the pBR322.
  • This plasmid was digested with EcoRI and Xhol and the large DNA fragment containing the amino-terminal region was isolated by agarose gel electrophoresis.
  • the insert of the plasmid- pBI(2)1/(3)13/(7)1/stop was purified by complete digestion with Xhol, partial digestion with Sail, and preparative gel electrophoresis. These two DNA fragments were ligated into the EcoRI/Sall fragment of pBR322.
  • the resulting correct construct can be described by restriction fragment mapping.
  • the EcoRI/Sall insert can be excised and placed in a suitable expression vector.
  • the nucleotide sequence and the corresponding amino acid sequence is shown in Figure 18.
  • the coding sequence, or suitable LBP coding portions thereof, can be placed in a suitable expression vector, by techniques like those described above, to achieve apoB expression in the corresponding expression system, such the high-producer CHO cells.
  • the recombinantly produced apoBl can be purified according to the method of the invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Procédé de production d'un peptide à liaison lipide purifié qui peut se lier à des phospholipides au niveau d'une ou de plusieurs régions peptides alpha-hélicoïdales amphipatiques. Le procédé consiste à établir un codage génétique du peptide et à introduire le gène sous forme hétérologue exprimable dans un système d'expression approprié capable de synthétiser un mélange de peptides qui comprend le peptide à liaison lipide. L'addition de lipides endogènes ou exogènes au mélange de peptides forme un complexe lipopeptide de faible densité composé du lipide et du peptide à liaison lipide, et ce complexe peut être séparé aisément des peptides à liaison non lipide dans le peptide d'après sa taille et/ou sa densité. Le procédé est destiné en particulier à la production à grande échelle d'apolipoprotéines humaines purifiées et de leurs régions à liaison lipide alpha-hélicoïdale. Sont également décrits des procédés apparentés pour a production d'apolipoprotéines recombinantes, des compositions lipopeptides thérapeutiques, et une émulsion lipide stabilisée pour la thérapie nutritionelle. Sont également décrits des procédés d'expression des apolipoprotéines ou de leurs segments à liaison lipide dans des systèmes d'expression cellulaire mammifère, bactérienne, et de levure, ainsi que des procédés de purification de protéines à liaison lipide, y compris des protéines recombinantes fusionnées.
EP19860906545 1985-10-04 1986-10-02 Apolipoproteines recombinantes et procedes. Withdrawn EP0239631A4 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US78441885A 1985-10-04 1985-10-04
US784418 1985-10-04
US80469285A 1985-12-04 1985-12-04
US804692 1985-12-04

Publications (2)

Publication Number Publication Date
EP0239631A1 true EP0239631A1 (fr) 1987-10-07
EP0239631A4 EP0239631A4 (fr) 1989-01-12

Family

ID=27120281

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19860906545 Withdrawn EP0239631A4 (fr) 1985-10-04 1986-10-02 Apolipoproteines recombinantes et procedes.

Country Status (3)

Country Link
EP (1) EP0239631A4 (fr)
AU (1) AU6522486A (fr)
WO (1) WO1987002062A1 (fr)

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU587989B2 (en) * 1984-10-16 1989-09-07 Mitsubishi Chemical Corporation DMA fragments, expression vectors, proteins, hosts, and process for production of the proteins
GB8712540D0 (en) 1987-05-28 1987-07-01 Ucb Sa Expression of human proapolipoprotein a-i
EP0347915A3 (fr) * 1988-06-23 1991-03-20 The Rockefeller University Méthode et agents inhibiteurs de la synthèse des lipoprotéines de basse densité
WO1990002762A1 (fr) 1988-09-02 1990-03-22 The Rockefeller University Mediateur inflammatoire derive de macrophages (mip-2)
IT1229996B (it) * 1989-04-20 1991-09-20 Cesare Sirtori Espressione di apolipoproteina ai e apolipoproteina ai-milano in lievito e composizioni farmaceutiche che contengono dette apolipoproteine.
FR2666813A1 (fr) * 1990-09-18 1992-03-20 Rhone Poulenc Sante Procede microbiologique de preparation de l'apolipoproteine aiv humaine ou de derives de celle-ci.
ES2283007T3 (es) 1995-11-09 2007-10-16 The Government Of The Usa As Represented By The Secretary Of Department Of Health And Human Services Utilizacion de la lecitina colesterol aciltransferasa (lcat) en el tratamiento de la aterosclerosis.
US6306433B1 (en) 1997-08-12 2001-10-23 Pharmacia Ab Method of preparing pharmaceutical compositions
JP2003525883A (ja) * 2000-03-03 2003-09-02 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム ワクチン
RU2003136090A (ru) 2001-05-15 2005-04-20 Джэпэн Иммьюнорисерч Лэборэтериз Ко., Лтд (Jp) Пептид, взаимодействующий с липопротеином высокой плотности
US7759315B2 (en) 2005-03-09 2010-07-20 Csl Behring Ag Treatment of inflammatory conditions of the intestine
PL2465519T3 (pl) 2007-03-01 2014-05-30 Csl Ltd Leczenie dysfunkcji śródbłonka u pacjentów z cukrzycą
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
MX2010013759A (es) 2008-06-13 2011-05-25 Proyecto Biomedicina Cima Sl Conjugados para la administracion de compuestos biologicamente activos.
KR102344392B1 (ko) 2008-11-10 2021-12-28 알닐람 파마슈티칼스 인코포레이티드 치료제 운반용 신규 지질 및 조성물
AU2010208035B2 (en) 2009-01-29 2016-06-23 Arbutus Biopharma Corporation Improved lipid formulation for the delivery of nucleic acids
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
NZ594995A (en) 2009-03-12 2013-06-28 Alnylam Pharmaceuticals Inc LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF HUMAN KINESIN FAMILY MEMBER 11 (Eg5) AND VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) GENES
NZ621981A (en) 2009-05-05 2015-09-25 Tekmira Pharmaceuticals Corp Lipid compositions
EA201791744A3 (ru) 2009-06-10 2018-07-31 Арбутус Биофарма Корпорэйшн Улучшенная липидная композиция
WO2011020023A2 (fr) 2009-08-14 2011-02-17 Alnylam Pharmaceuticals, Inc. Compositions formulées dans des lipides et procédés pour inhiber l’expression d’un gène du virus ebola
US9687550B2 (en) 2009-12-07 2017-06-27 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
ES2749426T3 (es) 2009-12-18 2020-03-20 Univ British Columbia Métodos y composiciones para administración de ácidos nucleicos
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
US20130260460A1 (en) 2010-04-22 2013-10-03 Isis Pharmaceuticals Inc Conformationally restricted dinucleotide monomers and oligonucleotides
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011133871A2 (fr) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Dérivés d'extrémité 5'
WO2011139911A2 (fr) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Arn simple brin à formulation lipidique
WO2011143362A1 (fr) 2010-05-11 2011-11-17 Esperion Therapeutics, Inc. Variants d'apolipoprotéine a1 résistants à l'oxydation dimères
DK2575764T3 (en) 2010-06-03 2017-08-07 Alnylam Pharmaceuticals Inc BIODEGRADABLE LIPIDS FOR THE ACTIVATION OF ACTIVE AGENTS
WO2012016188A2 (fr) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Procédés et compositions pour l'administration d'agents actifs
WO2012016184A2 (fr) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Procédés et compositions pour la délivrance d'agents actifs
CN103370054A (zh) 2010-11-09 2013-10-23 阿尔尼拉姆医药品有限公司 用于抑制Eg5和VEGF基因的表达的脂质配制的组合物和方法
EP2663548B1 (fr) 2011-01-11 2017-04-05 Alnylam Pharmaceuticals, Inc. Lipides pégylés et leur utilisation pour une administration de médicament
EP2760477B1 (fr) 2011-09-27 2018-08-08 Alnylam Pharmaceuticals, Inc. Lipides di-aliphatiques pegylés substitués
US10894098B2 (en) 2012-04-09 2021-01-19 Signablok, Inc. Methods and compositions for targeted imaging
US9610324B2 (en) 2012-07-11 2017-04-04 Esperion Therapeutics, Inc. Apolipoprotein mixtures
JP2018504380A (ja) 2014-12-18 2018-02-15 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Reversir(商標)化合物

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0173280A1 (fr) * 1984-08-27 1986-03-05 Bio-Technology General Corporation Plasmides contenant des promoteurs lambda PL et site de restriction préparé pour le remplacement facile d'un site de liaison ribosomique, hôtes contenant les plasmides et méthodes correspondantes
WO1986004920A1 (fr) * 1985-02-13 1986-08-28 Biotechnology Research Partners, Limited Promoteur de la metallothioneine-ii humaine dans un systeme mammifere d'expression
WO1987002061A1 (fr) * 1985-10-03 1987-04-09 Biotechnology Research Partners, Ltd. Nouveaux systemes d'administration de medicaments a base de lipoproteines

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU542264B2 (en) * 1979-06-01 1985-02-14 G.D. Searle & Co. Plasmid vectors
US4399216A (en) * 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4579821A (en) * 1981-11-23 1986-04-01 University Patents, Inc. Control of DNA sequence transcription
US4546082A (en) * 1982-06-17 1985-10-08 Regents Of The Univ. Of California E. coli/Saccharomyces cerevisiae plasmid cloning vector containing the alpha-factor gene for secretion and processing of hybrid proteins

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0173280A1 (fr) * 1984-08-27 1986-03-05 Bio-Technology General Corporation Plasmides contenant des promoteurs lambda PL et site de restriction préparé pour le remplacement facile d'un site de liaison ribosomique, hôtes contenant les plasmides et méthodes correspondantes
WO1986004920A1 (fr) * 1985-02-13 1986-08-28 Biotechnology Research Partners, Limited Promoteur de la metallothioneine-ii humaine dans un systeme mammifere d'expression
WO1987002061A1 (fr) * 1985-10-03 1987-04-09 Biotechnology Research Partners, Ltd. Nouveaux systemes d'administration de medicaments a base de lipoproteines

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 262, no. 9, March 1987, pages 4241-4247, The American Society of Biological Chemists, Inc., Washington, DC, US; J. BEDNARZ MALLORY et al.: "Expression and characterization of human apolipoprotein A-I in Chinese hamster ovary cells" *
JOURNAL OF MEDICINAL CHEMISTRY, vol. 25, no. 10, October 1982, pages 1115-1120, American Chemical Society, Washington, DC, US; R.E. COUNSELL et al.: "Lipoproteins as potential site-specific delivery systems for diagnostic and therapeutic agents" *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCE, USA, vol. 83, March 1986, pages 1467-1471, Washington, DC, US; A.A. PROTTER et al.: "Isolation of a cDNA clone encoding the amino-terminal region of human apolipoprotein B" *
See also references of WO8702062A1 *

Also Published As

Publication number Publication date
EP0239631A4 (fr) 1989-01-12
WO1987002062A1 (fr) 1987-04-09
AU6522486A (en) 1987-04-24

Similar Documents

Publication Publication Date Title
EP0239631A1 (fr) Apolipoproteines recombinantes et procedes
EP0217822B1 (fr) Promoteur de la metallothioneine-ii humaine dans un systeme mammifere d'expression
US4943527A (en) Mature apoai protein production under serum free culturing conditions
CA1316132C (fr) Adnc et gene de l'angiogenine humaine (facteur d'angiogenese) et methode d'expression
JP2525438B2 (ja) ファクタ―8:cの製造方法
EP0361991A2 (fr) Méthode de préparation microbiologique du sérum d'albumine humaine et d'autres protéines hétérologues à partir d'une levure
Chong et al. Interactions of normal and mutant vesicular stomatitis virus matrix proteins with the plasma membrane and nucleocapsids
EP0446017A1 (fr) Nouvelles méthodes de diagnose et de traitement par utilisation de kystique fibrosis transmembrane régulateur
WO1987002061A1 (fr) Nouveaux systemes d'administration de medicaments a base de lipoproteines
AU641389B2 (en) Expression of human apolipoproteins AI-milano in yeast
PT85349B (pt) Processo para a preparacao de proteinas com actividade de factor viii por celulas microbianas hospedeiras, de vectores de expressao bem como de composicoes farmaceuticas contendo estas proteinas e de anticorpos uteis para a purificacao de factor viii e para diagnostico
WO1986003408A1 (fr) Proteine tensio-active alveolaire recombinante
US5935815A (en) Process for micro biological production of proteins
JP2757987B2 (ja) ヒト抗‐炎症ホスホリパーゼ阻害蛋白
US5993827A (en) Binding domains from plasmodium vivax and plasmodium falciparum erythrocyte binding proteins
JPH11505712A (ja) 新規のアポリポタンパク質a−i変異体
JPH08503858A (ja) 生物活性ポリペプチド融合ダイマー
FI105348B (fi) Menetelmä proteiinien käsittelemiseksi endoproteolyyttisesti sekä menetelmä proteiinien valmistamiseksi (mikro)biologisesti
WO1991017178A1 (fr) Procede permettant de purifier des proteines produites par recombinaison
AU720446B2 (en) Apolipoprotein E2 and treatment of Alzheimer's disease
WO1989009818A1 (fr) Procede de purification de phospholipase a2 et de production de polypeptides semblables a la phospholipase a2
CA2042605A1 (fr) Proteines inhibitrices de la cytolyse, sequences d'adn codant pour ces proteines, et plasmides, organismes hotes et procedes permettant d'obtenir ces proteines
JPS63501764A (ja) 組換えアポリポタンパクおよび方法
de Zulueta Pascual et al. Retion of human apolipoprotein A-II by a mutant of Saccharomyces cerevisiæ

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19870609

AK Designated contracting states

Kind code of ref document: A1

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

A4 Supplementary search report drawn up and despatched

Effective date: 19890112

17Q First examination report despatched

Effective date: 19910425

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 19941001

RIN1 Information on inventor provided before grant (corrected)

Inventor name: KANE, JOHN, P.

Inventor name: PROTTER, ANDREW, A.

Inventor name: TALMADGE, KAREN, D.

Inventor name: MALLORY, JOANNE, B.

Inventor name: VIGNE, JEAN-LOUIS