US20090053790A1 - Polypeptide Having Affinity for Envelope Virus Constituent and Use Thereof in Transferring Substance Into Cell - Google Patents

Polypeptide Having Affinity for Envelope Virus Constituent and Use Thereof in Transferring Substance Into Cell Download PDF

Info

Publication number
US20090053790A1
US20090053790A1 US12/223,464 US22346407A US2009053790A1 US 20090053790 A1 US20090053790 A1 US 20090053790A1 US 22346407 A US22346407 A US 22346407A US 2009053790 A1 US2009053790 A1 US 2009053790A1
Authority
US
United States
Prior art keywords
polypeptide
envelope
amino acid
protein
acid sequence
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.)
Abandoned
Application number
US12/223,464
Other languages
English (en)
Inventor
Kuni Fushikida
Yoshitaka Kondo
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.)
Ishihara Sangyo Kaisha Ltd
Original Assignee
Ishihara Sangyo Kaisha 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 Ishihara Sangyo Kaisha Ltd filed Critical Ishihara Sangyo Kaisha Ltd
Assigned to ISHIHARA SANGYO KAISHA, LTD. reassignment ISHIHARA SANGYO KAISHA, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FUSHIKIDA, KUNI, KONDO, YOSHITAKA
Publication of US20090053790A1 publication Critical patent/US20090053790A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18811Sendai virus
    • C12N2760/18822New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18811Sendai virus
    • C12N2760/18841Use of virus, viral particle or viral elements as a vector
    • C12N2760/18845Special targeting system for viral vectors
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6072Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses

Definitions

  • the present invention relates to proteins which allow foreign substances to be enclosed within the envelope of envelope viruses or inactivated envelope viruses, and to methods for the introduction of foreign substances into target cells by means of envelope viruses or inactivated envelope viruses into which the foreign substance is incorporated with said protein.
  • the known technique for introducing proteins into cells includes methods utilizing cationic liposomes and membrane-permeable peptides. Since the cationic liposome-based delivery performs transfection into cells using charges, it is frequently hard to make preparations because the charge varies from protein to protein, and it is often difficult to perform introduction due to protein charges.
  • the known membrane-permeable peptide-based delivery techniques are those utilizing HIV-derived basic TAT peptides.
  • the reported method of introducing antibodies into cells is a method utilizing a fusion protein consisting of TAT which functions as the membrane-permeable peptide and an antibody-binding region such as Protein A (Patent Document 1).
  • the basic membrane-permeable peptide-based introduction of proteins still have problems in scenes wherein intracytoplasmic localization is demanded since basic peptides have nuclear import activity and TAT peptides are known to be transported into the nucleus.
  • microinjection-based introduction of proteins and antibodies into cells needs special machinery and tools and also requires extremely complicated operations.
  • Non-Patent Document 2 a nucleic acid such as a gene
  • Non-Patent Documents 1 & 2 a protein such as RNase Tl and ⁇ -Galactosidase
  • Patent Document 1 JP 2005-052083, A
  • Non-Patent Document 1 European Journal Biochemistry vol. 271, No. 17, 3567-3572, 2004
  • Non-Patent Document 4 Current Gene Therapy, vol. 4, 183-194, 2004
  • the present invention provides proteins for introducing one or more proteins, antibodies or other foreign substances (non-self or exogenous substances) into one or more cells without impairing the function, property or structure thereof; and methods of introducing the foreign substance into the cell in a time- and quantity-controllable manner with high efficiency by using the above-described delivery protein or an envelope virus or inactivated envelope virus in combination with said delivery protein.
  • the present inventors have conducted an extensive research on methods of enclosing foreign substances within the envelope of envelope viruses. As a result, the present inventors have succeeded in finding that a protein containing a polypeptide having an affinity for (or compatible to) a constituent of the envelope virus contributes to the efficient enclosure of the foreign substance in the envelope.
  • the present inventors have succeeded in finding that use of the above-described proteins enables foreign substances to be included in an envelope virus or inactivated envelope virus and the resultant foreign substance-containing envelope viruses or inactivated envelope viruses make it possible to efficiently transfer the substance into cells without damaging the physiological function thereof. Therefore, the present invention has been achieved.
  • the present invention provides the following:
  • a protein comprising (a) a polypeptide having an affinity for one or more envelope viral components in combination with (b) a polypeptide capable of binding to one or more foreign substances to be introduced into a cell.
  • polypeptide (a) is a polypeptide having an affinity for one or more components existing in the inner space of an envelope.
  • An envelope virus or inactivated envelope virus containing a protein comprising (a) a polypeptide having an affinity for one or more envelope viral components in combination with (b) a polypeptide capable of binding to one or more foreign substances to be introduced into a cell.
  • a fusion protein consisting of a polypeptide having an affinity for one or more envelope viral components, fused to an antibody binding polypeptide
  • said fusion protein is a polypeptide selected from the group consisting of:
  • a process for preparing an envelope virus or inactivated envelope virus containing a foreign substance within an envelope which comprises the steps consisting of
  • a process for preparing an envelope virus or inactivated envelope virus containing a foreign substance within an envelope which comprises the steps consisting of
  • a method for introducing one or more foreign substances into a cell with an envelope virus or inactivated envelope virus which comprises the steps consisting of
  • a method for introducing one or more foreign substances into a cell with an envelope virus or inactivated envelope virus which comprises the steps consisting of
  • a complex composed of (i) a protein comprising (a) a polypeptide having an affinity for one or more envelope viral components in combination with (b) a polypeptide capable of binding to one or more foreign substances to be introduced into a cell, and (ii) one or more foreign substances to be introduced into one or more cells.
  • proteins, antibodies and other foreign substances can be introduced into cells via simple operations without damaging their function, property and structure.
  • the foreign substance is an antibody
  • a variety of substances bound to the antibodies can be transferred into cells.
  • Antibodies against various intracellular materials can also be introduced and the function of such intracellular materials can be regulated.
  • antibodies against substances to be administered from the outside of cells as well as metabolites thereof and others can be introduced and therefore the intracellular physiological activity of said substance can also be regulated.
  • FIG. 1 shows SDS-PAGE patterns for ZZ-NP-containing inactivated HVJ envelope (lane 2), inactivated HVJ envelope alone (lane 1), ZZ-NP alone (lane 3), and molecular markers (lane 4).
  • FIG. 2 shows Western blotting patterns for mouse IgG-containing inactivated HVJ envelopes.
  • Lane 4 to 6 mouse IgG enclosed within ZZ-NP-containing inactivated HVJ envelopes.
  • Lane 1 to 3 mouse IgG Ab alone (1 ⁇ 3 ⁇ g, 1 ⁇ g, and 3 ⁇ g, respectively).
  • FIG. 3 shows photos for Anti-NPC (Nuclear Pore Complex) antibody, incorporated into HeLa S3 cell with inactivated HVJ envelope and, stained with Alexa 488-labeled goat Anti-Mouse IgG F(ab′) 2 fragment. Fluorescence image (1). Bright-field image (2).
  • FIG. 4 shows Western blotting patterns for mouse IgG antibody-containing inactivated HVJ envelopes.
  • Mouse IgG antibodies incorporated into inactivated HVJ envelopes (lane 1, 2).
  • Mouse IgG antibodies incorporated into inactivated HVJ envelopes containing the proteins of the present invention (ZZ-NP: lane 3, 4; ZZ-NPK1: lane 5, 6; ZZ-NPK2: lane 7, 8, ZZ-NPK3: lane 9, 10; ZZ-NPK4: lane 11, 12).
  • Lane 13 to 17 Mouse IgG Ab alone ( 1/16 ⁇ g, 1 ⁇ 8 ⁇ g, 1 ⁇ 4 ⁇ g, 1 ⁇ 2 ⁇ g, and 1 ⁇ g, respectively).
  • FIG. 5 shows photos for Anti-NPC (Nuclear Pore Complex) Ab, incorporated into HeLa S3 cell with inactivated HVJ envelope and, stained with Alexa 488-labeled goat Anti-Mouse IgG F(ab′) 2 fragment.
  • FIG. 6 shows SDS-PAGE patterns for inactivated HVJ envelopes within which G-NPK1 or GG-NPK1 made Rat IgG2b Ab be enclosed.
  • Rat IgG2b Ab incorporated into inactivated HVJ envelopes containing the proteins of the present invention (G-NPK1: 6, GG-NPK1: 9).
  • Molecular markers (lane 1).
  • Rat IgG2b Ab alone (lane 2).
  • Inactivated HVJ envelope alone (lane 3).
  • G-NPK1 alone (lane 4).
  • GG-NPK1 alone (lane 7).
  • Inactivated HVJ envelopes containing the proteins of the present invention (G-NPK1: lane 5, GG-NPK1: lane 8).
  • FIG. 7 shows SDS-PAGE patterns for inactivated HVJ envelopes within which L-NPK1 or LL-NPK1 made Rat IgG2b Ab be enclosed.
  • Rat IgG2b Ab incorporated into inactivated HVJ envelopes containing the proteins of the present invention (L-NPK1: 6, LL-NPK1: 9).
  • Molecular markers (lane 1).
  • Rat IgG2b Ab alone (lane 2).
  • Inactivated HVJ envelope alone (lane 3).
  • L-NPK1 alone (lane 4).
  • LL-NPK1 alone (lane 7).
  • Inactivated HVJ envelopes containing the proteins of the present invention L-NPK1: lane 5, LL-NPK1: lane 8).
  • FIG. 8 show photos for Rat IgG2b Ab, incorporated into HeLa S3 cells with inactivated HVJ envelope and, stained with Alexa 488-labeled chicken anti-Rat IgG Ab.
  • Cells receiving introduction of Rat IgG2b Ab with inactivated HVJ envelopes containing the proteins of the present invention [(G-NPK1: (1), GG-NPK1: (2), L-NPK1: (3), LL-NPK1: (4)].
  • FIG. 9 shows the detection, with Alexa 488-labeled goat anti-mouse IgG F(ab′) 2 fragments, of anti-NPC (Nuclear Pore Complex) antibodies introduced into HeLa S3 cells with inactivated HVJ envelopes.
  • Cells treated by G-NPK1- or GG-NPK1-containing inactivated HVJ envelope-mediated introduction of anti-NPC Ab [(1): G-NPK1, (2): GG-NPK1].
  • the envelope viruses that can be used herein include a variety of viruses as long as they have an envelope. It is known that the envelope virus delivers nucleic acid molecules, nucleocapsid proteins and others into host cells. Such viruses include those belonging to a viral family selected from the group consisting of Retroviridae, Togaviridae, Coronaviridae, Flaviviridae, Paramyxoviridae, Orthomyxoviridae, Bunyaviridae, Rhabdoviridae, Poxyiridae, Herpesviridae, Baculoviridae and Hepadnaviridae. Preferable viruses are those in the family Paramyxoviridae or Orthomyxoviridae, more preferably Paramyxoviridae.
  • viruses of the Paramyxovirinae include those of the genus Respirovirus , the genus Rubulavirus , and the genus Morbillivirus . Among them, viruses of the genus Respirovirus are preferable, more preferably Sendai virus.
  • Sendai virus and “HVJ” (Hemagglutinating virus of Japan) are interchangeably used herein.
  • the “inactivated” as used herein refers to inactivation of genomes when stated in connection with viruses.
  • the inactivated virus is replication-defective.
  • the genome inactivation can be achieved by, for example, UV or alkylating agent treatments.
  • Any of envelope viruses and inactivated envelope viruses can be used herein, and the inactivated envelope viruses are more desirable.
  • the phrases inactivated envelope viruses and inactivated viral envelopes are interchangeably used herein.
  • inventive proteins are those each comprising (a) a polypeptide having an affinity for (compatible to) one or more envelope viral components (or viral constituents) in combination with (b) a polypeptide capable of binding to one or more foreign substances (or exogenous molecules) to be introduced (or transferred) into one or more cells.
  • the polypeptide (a) is a polypeptide facilitating, through binding to the polypeptide (b), the state wherein said foreign substance is “enclosed within an envelope” or “incorporated into the inner space of an envelope”.
  • the polypeptide (a) is also a polypeptide elevating the level of said foreign substance enclosed within the envelope.
  • polypeptides (a) and (b) may be in the form of a complex wherein each polypeptide (a) is interacted with each polypeptide (b) to form a subunit, or in the form of a single fusion protein.
  • the fusion proteins are more desirable.
  • the “envelope” as used herein refers to a lipid bilayer-based membrane (or membranous structure) which surrounds the nucleocapsid existing in the envelope virus.
  • a specific substance (or specific molecule) is “enclosed within an (or the) envelope”, means that such a specific substance (or specific molecule) is present in an aqueous phase with which the inner space surrounded by the aforementioned lipid bilayer-based membranous structure is filled up, and/or, in the lipid of the membranous structure, being considered to be dependent on the surface properties (such as hydrophilicity or hydrophobicity) of proteins or foreign substances to be transferred into cells. It is desired that said substances are placed mainly in the aqueous layer inside the envelope of the inactivated virus. Further, the “incorporated in the envelope” as used herein means that a substance becomes “enclosed within the envelope” as aforementioned.
  • the above-described envelope viral components include proteins existing on or in the surface of the envelope, phosphorylated (P) proteins, matrix (M) proteins, nucleic acids, proteins associated with the nucleic acid, etc.
  • Embodiments of the above-described polypeptide (a) are proteins, or peptide fragments thereof, which have an affinity for (or are compatible to) one or more members selected from the group consisting of proteins existing on or in the surface of the envelope, phosphorylated proteins, matrix proteins, nucleic acids, and proteins associated with the nucleic acid.
  • the above-described polypeptide (a) is a protein, or a peptide fragment thereof, which is compatible to (or has an affinity for) one or more virus components existing in the inside of the envelope, more preferably a protein, or a peptide fragment thereof, which has an affinity for (or is compatible to) one or more members selected from the group consisting of the phosphorylated protein, the matrix protein, the nucleic acid, and the protein associated with the nucleic acid.
  • a more preferred embodiment of the polypeptide (a) is an envelope viral nucleocapsid protein, or a peptide fragment thereof.
  • the nucleocapsid protein (abbreviated to “NP”), derived from Sendai virus which is a preferred example of the envelope virus, has an affinity for the protein existing in the inside of the virus envelope and the virus nucleic acid (RNA).
  • the NP has also an affinity for interactions between the nucleocapsid proteins and allows the formation of complexes within the virus wherein the NP is closely associated with the nucleic acid.
  • the Sendai virus nucleocapsid protein includes preferably polypeptides of an amino acid sequence of SEQ ID NO: 2 or an amino acid sequence substantially identical thereto, or peptide fragments (partial peptides) thereof.
  • the peptide fragments are those each having an amino acid sequence selected from SEQ ID NOs: 27, 29, 31, and 33.
  • polypeptide capable of binding to one or more foreign substances may be any as long as it is a polypeptide comprising an amino acid sequence capable of binding to the foreign substance, and can be suitably selected, depending on the foreign substances to be treated.
  • the foreign substance to be transferred into one or more cells includes nucleic acid molecules, proteins, polysaccharides and lipids, preferably proteins, and more preferably antibodies.
  • the polypeptide (b) includes basic amino acid-rich polypeptides, polypeptides which can bind to the specific sequence of genes and others.
  • Embodiments of such polypeptides are DNA-binding sequences such as GAL4.
  • the polypeptide (b) includes avidins such as egg white-derived avidin, and streptavidin derived from bacteria.
  • the polypeptide (b) includes polypeptides capable of binding to an antibody (Ab) (antibody-binding polypeptides) or peptide fragments thereof.
  • Abs antibody-binding polypeptides
  • Desirable embodiments of the Ab-binding polypeptide are immunoglobulin-binding proteins and peptide fragments thereof. More preferable examples of the antibody-binding polypeptide are proteins capable of binding to an antibody Fc region or kappa ( ⁇ ) light chain and peptide fragments thereof.
  • ⁇ light chain-binding molecules will be selected. More specifically, they include Fc region-binding molecules, Protein A, Protein G, and Protein A/G; ⁇ light chain-binding molecules, Protein L; amino acid sequences substantially identical thereto or peptide fragments thereof.
  • immunoglobulin-binding proteins used herein may be whole protein units, or immunoglobulin-binding sequence fragments of such proteins. It is desirable that the aforementioned binding sequences are amino acid sequences which bind to an Fc region or ⁇ light chain.
  • the Fc region-binding sequence includes amino acid sequences for the A, B, C, D, and E regions of Protein A, said B region-derived Z region, and the C1, C2, and C3 regions of Protein G, and amino acid sequences substantially identical thereto. It is possible to use the member of such species alone or in combination with one or more sequences selected from the aforementioned sequences. More desirable examples thereof are the Z region having an amino acid sequence of SEQ ID NO: 4, the Protein G C1 region having an amino acid sequence of SEQ ID NO: 49, the Protein L B1 region having an amino acid sequence of SEQ ID NO: 51, and amino acid sequences substantially identical thereto.
  • the polypeptide (a) may be joined with the polypeptide (b) to form a fusion protein. Alternatively, they may be in the form of a complex wherein each polypeptide (a) is associated with each polypeptide (b) to form a subunit.
  • the fusion proteins are more preferable.
  • Such fusion proteins are molecules each comprising (a) the polypeptide having an affinity for one or more envelope viral components in combination with (b) the foreign substance-binding polypeptide, as aforementioned, wherein the polypeptide (b) may be positioned at either the N-terminal end (amino-terminal side) or the C-terminal end (carboxyl-terminal side) of the polypeptide (a).
  • More desirable embodiments are fusion proteins wherein the polypeptide (b) is positioned at the N terminal end of the polypeptide (a).
  • the fusion protein may contain a spacer region between the polypeptides (a) and (b) for avoiding interference due to steric hindrance.
  • the spacer region may be a glycine spacer, or composed of glycine and serine.
  • Preferable examples of the fusion protein molecule (4) are proteins each comprising an amino acid sequence of SEQ ID NO: 6, 13, 15, 17, 19, 53, 55, 57 or 59, or an amino acid sequence substantially identical thereto, more preferably proteins each comprising an amino acid sequence of SEQ ID NO: 6, 13, 55 or 59, or an amino acid sequence substantially identical thereto.
  • amino acid sequence substantially identical refers to amino acid sequences that have at least about 70%, preferably about 80%, more preferably about 90%, most preferably about 95% or higher amino acid residue identity (or homology).
  • identity means the percentage (%) of identical amino acid and similar amino acid residues as compared to all overlapped amino acid residues in optimal alignment of two amino acid sequences, when aligned using one of mathematical algorithms known in the art (preferably, said algorithm can take into account the presence of gaps, which need to be introduced into one or both sequences for the optimal alignment).
  • the “similar amino acid” refers to amino acids of like characteristics (e.g., physical and chemical properties), which include those classified into the same group, such as aromatic amino acids (Phe, Trp, Tyr), aliphatic amino acids (Ala, Leu, Ile, Val), polar amino acids (Gln, Asn), basic amino acids (Lys, Arg, His), acidic amino acids (Glu, Asp), hydroxyl group-containing amino acids (Ser, Thr), and small-sized side chain amino acids (Gly, Ala, Ser, Thr, Met). Replacements among such similar amino acids will be likely to be phenotypically silent in a protein (i.e., conservative amino acid substitutions). Embodiments of the conservative amino acid substitution are well known in the art and found in a variety of documents (see, for instance, Bowie et al., Science, 247: 1306-1310 (1990)).
  • Preferred examples of the “protein comprising an amino acid sequence substantially identical thereto” herein are proteins that are substantially identical in both amino acid sequence levels and qualitative activities, and the like.
  • the proteins according to the present invention encompass modified proteins (so-called “muteins”) comprising any of (1) deleted amino acid sequences which have one or more deletions of one or more amino acid residues (preferably about 1 to 30, still preferably about 1 to 10, and further preferably several (1 to 5) amino acid residues); (2) added amino acid sequences which have one or more additions of one or more amino acid residues (preferably about 1 to 30, still preferably about 1 to 10, and further preferably several (1 to 5) amino acid residues); (3) inserted amino acid sequences which have one or more insertions of one or more amino acid residues (preferably about 1 to 30, still preferably about 1 to 10, and further preferably several (1 to 5) amino acid residues); (4) substituted amino acid sequences which have one or more substitutions of one or more amino acid residues (preferably about 1 to 30, still preferably about 1 to 10, and further preferably several (1 to 5) amino acid residues) with one or more other amino acid residues; and (5) combinations wherein the aforementioned one or more amino acid deletions, additions, insertions
  • protein or peptide sequences are written in the N-terminal end to C-terminal end direction from left to right according to the conventional peptide notation.
  • the protein of the present invention is any of molecules having a carboxyl (—COOH), carboxylate (—COO ⁇ ), amide (—CONH 2 ) or ester (—COOR) group at the C-terminal end.
  • R is a C 1-6 alkyl group such as methyl, ethyl, n-propyl, isopropyl, and n-butyl; a C 3-8 cycloalkyl group such as cyclopentyl and cyclohexyl; a C 6-12 aryl group such as phenyl and ⁇ -naphtyl; a C 7-14 aralkyl group including a phenyl-C 1-2 alkyl group such as benzyl and phenethyl, and an ⁇ -naphtyl-C 1-2 alkyl group such as ⁇ -napthylmethyl; a pivaloyloxymethyl group, etc.
  • protein molecules wherein the carboxyl group is amidated or esterified are also included therein.
  • ester as used herein is selected from those listed in connection with the aforementioned C-terminal end ester.
  • proteins of the present invention also encompass protein molecules wherein the amino group of the N-terminal amino acid residue is protected with a protective group (for example, C 1-6 acyl including C 1-6 alkanoyl such as formyl and acetyl), those molecules wherein the N-terminal glutamine residue generated through in vivo cleavage is pyroglutamated, those molecules wherein a substituent on an intermolecular amino acid side chain (e.g., —OH, —SH, an amino group, an imidazole group, an indole group, a guanidino group and the like) is protected with a suitable protective group (e.g., C 1-6 acyl including C 1-6 alkanoyl such as formyl and acetyl), conjugated proteins such as so-called glycoproteins having one or more saccharide chains.
  • a protective group for example, C 1-6 acyl including C 1-6 alkanoyl such as formyl and acetyl
  • the subject protein-encoding DNA includes, for example, DNA molecules having a nucleotide sequence of SEQ ID NO: 5, 12, 14, 16, 18, 48, 50, 52, 54, 56 or 58, or DNA molecules that not only have a nucleotide sequence capable of hybridizing to said nucleotide sequence under high-stringent conditions but also encode a protein with the substantially same nature of activity, and others.
  • the DNA molecule, used herein, that is capable of hybridizing to said nucleotide sequence under high-stringent conditions includes DNA comprising a nucleotide sequence with at least about 50%, preferably at least about 6.0%, more preferably at least about 70%, particularly preferably at least about 80%, most preferably at least about 90% or higher homology to said nucleotide sequence.
  • hybridization can be performed according to methods known per se or equivalent techniques, for instance, the method disclosed in J. Sambrook et al., Molecular Cloning, Second Edition, Cold Spring Harbor Lab. Press, 1989, etc. When commercially available libraries are used, the hybridization can be carried out according to methods described in manuals attached thereto. Preferably, the hybridization can be achieved under high-stringent conditions.
  • High-stringent hybridization conditions referred to herein are conditions in about 19 to 40 mM, preferably about 19 to 20 mM, sodium salt at about 50 to 70° C., preferably about 60 to 65° C. Particularly, preferable high-stringent conditions are those in about 19 mM sodium salt at about 65° C.
  • DNA sequences can be converted with any of known kits including, for instance, MutanTM-Super Express Km (Takara Shuzo, Tokyo, Japan), MutanTM-K (Takara Shuzo, Tokyo, Japan) and other kits, based on methods known per se, such as ODA-LA PCR method, Gapped duplex method, and Kunkel method, or equivalent techniques.
  • Cloned DNA can be used without any modification, or after, as desired, digestion with restriction enzymes and/or addition of linkers, depending on targeted subjects.
  • the DNA may include the ATG translation initiation codon at the 5′-end and the translation stop codon, TAA, TGA or TAG, at the 3′-end. These translation initiation and stop codons can be added using suitable synthetic DNA adaptors.
  • Expression vectors that comprise DNA coding for a protein within the scope of the present invention can be constructed, for example, as follows: a DNA molecule that encodes the protein of the present invention is cut to give a target DNA fragment which is then inserted into a suitable expression vector downstream of a promoter in the vector.
  • the expression vector used herein includes E.
  • coli -derived plasmids e.g., pBR322, pBR325, pUC12, pUC13, and pET28a
  • Bacillus subtilis -derived plasmids e.g., pUB110, pTP5, and pC194
  • yeast-derived plasmids e.g., pSH19, and pSH15
  • insect cell expression plasmids e.g., pFast-Bac
  • animal cell expression plasmids e.g., pA1-11, pXT1, pRc/CMV, pRc/RSV, and pcDNAI/Neo
  • bacteriophages such as ⁇ phage
  • insect viral vectors such as baculoviral vectors (e.g., BmNPV, and AcNPV)
  • viral vectors derived from animal viruses such as retro-viruses, vaccinia viruses, and
  • any promoter may be employed as long as it serves as a suitable promoter for hosts used in gene expression.
  • examples of such promoters used herein are the SR ⁇ promoter, the SV40 or LTR promoter, the CMV (cytomegalovirus) and RSV (Rous sarcoma virus) promoters, MoMuLV (Moloney murine leukemia virus) LTR, the herpes simplex virus thymidine kinase promoter (HSV-TK), etc.
  • the CMV promoter, the SR ⁇ promoter and the like are preferable.
  • hosts are members of the genus Escherichia , trp promoter, lac, recA, ⁇ PL , lpp, T7 promoters and others are preferable.
  • hosts are members of the genus Bacillus , SPO1, SPO2, penP promoters and others are preferable.
  • hosts are yeast members, PHO5, PGK, GAP, ADH promoters and others are preferred.
  • the polyhedrin promoter, the P10 promoter and others are preferable.
  • the expression vector which can be used herein includes, in addition to the above, those comprising, if desired, an enhancer, a polyadenylation signal, a selectable marker, the SV40 origin of replication (hereinafter, often abbreviated to SV40 ori) and the like.
  • the selectable marker includes, for example, a dihydrofolate reductase gene (hereinafter, often abbreviated to dhfr, resistant to methotrexate (MTX)), ampicillin-resistant gene (hereinafter, often abbreviated to ampr), neomycin-resistant gene (hereinafter, often abbreviated to neor, resistant to G418) and others.
  • the target gene when dhfr gene-deficient Chinese hamster cells are used together with the dhfr gene as a selectable marker, the target gene may also be selected with a thymidine-free medium.
  • a nucleotide sequence coding for a signal sequence (signal codons) suitable for a host may be added to the 5′-terminal side of DNA encoding the protein or peptide fragment according to the present invention (or the native signal codons may be replaced with signal codons compatible with the host).
  • a PhoA signal sequence, an OmpA signal sequence, or the like can be used for Escherichia sp.
  • hosts an ⁇ -amylase signal sequence, a subtilisin signal sequence, or the like, for Bacillus sp. hosts; an MF ⁇ signal sequence, a SUC2 signal sequence, or the like, for yeast hosts; and an insulin signal sequence, an ⁇ -interferon signal sequence, an antibody molecule signal sequence, or the like, for animal cell hosts, respectively.
  • the Escherichia sp. used herein includes, for example, Escherichia coli K12•DH1 [Proc. Natl. Acad. Sci. USA, Vol. 60, 160 (1968)], Escherichia coli JM103 [Nucleic Acids Research, Vol. 9, 309 (1981)], Escherichia coli JA221 [Journal of Molecular Biology, Vol. 120, 517 (1978)], Escherichia coli HB101 [Journal of Molecular Biology, Vol. 41, 459 (1969)], Escherichia coli C600 [Genetics, Vol. 39, 440 (1954)], and Escherichia coli Rosetta (DE3) [Novagen].
  • Escherichia coli K12•DH1 Proc. Natl. Acad. Sci. USA, Vol. 60, 160 (1968)
  • Escherichia coli JM103 Nucleic Acids Research, Vol. 9, 309 (1981)
  • yeast used herein includes, for example, Bacillus subtilis MI114 [Gene, Vol. 24, 255 (1983)], Bacillus subtilis 207-21 [Journal of Biochemistry, Vol. 95, 87 (1984)].
  • the yeast used herein includes, for example, Saccharomyces cerevisiae AH22, AH22R ⁇ , NA87-11A, KDK-5D, 20B-12, Schizosaccharomyces pombe NCYC1913, NCYC2036, and Pichia pastoris KM71.
  • the insect cells used herein include, for example, when a virus is AcNPV, established insect cell lines, Spodoptera frugiperda cells (Sf cells), derived from a larva of cabbage army worm ( Spodoptera frugiperda ), MG1 cells, derived from the midgut of Trichoplusia ni , High FiveTM cells, derived from an egg of Trichoplusia ni , cells derived from Mamestra brassicae , cells derived from Estigmena acrea and others.
  • a virus is BmNPV
  • the insect cells used herein are established cell lines derived from silkworm ( Bombyx mori N cells; BmN cells) and the like.
  • Sf cells used herein are Sf9 cells (ATCC CRL1711), and Sf21 cells (both, Vaughn, J. L. et al., In Vivo, 13, 213-217 (1977)).
  • insects used herein are silkworm larva [Maeda et al., Nature, Vol. 315, 592 (1985)] and the like.
  • the animal cells used herein include, for example, monkey COS-7 cell lines, monkey Vero cells, Chinese hamster ovary (CHO) cells (hereinafter, abbreviated to CHO cells), dhfr gene-deficient Chinese hamster cell CHO (hereinafter, abbreviated to CHO(dhfr ⁇ ) cell), mouse L cells, mouse AtT-20 cells, mouse myeloma cells, rat GH3 cells, human FL cells and other cells.
  • CHO Chinese hamster ovary
  • Transformation can be performed according to known methods, depending on a variety of hosts.
  • Escherichia sp. bacteria can be transformed according to the methods disclosed in, for example, Proc. Natl. Acad. Sci. USA, Vol. 69, 2110 (1972) and Gene, Vol. 17, 107 (1982).
  • Bacillus sp. bacteria can be transformed according to the methods disclosed in, for example, Molecular & General Genetics, Vol. 168, 111 (1979).
  • Yeast cells can be transformed according to the methods disclosed in, for example, Methods in Enzymology, Vol. 194, 182-187 (1991); Proc. Natl. Acad. Sci. USA, Vol. 75, 1929 (1978) and others.
  • Insect cells and insects can be transformed according to the method disclosed in, for example, Bio/Technology, Vol. 6, 47-55 (1988).
  • Animal cells can be transformed according to the methods disclosed in, for example, Saibo Kougaku Bessatsu 8 Shin-Saibo Kougaku Jikken Purotokoru (Cell Technology Separate Volume 8 Novel Experimental Protocols for Cell Technology), 263-267 (1995, Shujunsha Co., Ltd., Tokyo, Japan), and Virology, Vol. 52, 456 (1973).
  • Cultivation of transformants or transfectants can be performed according to conventional techniques depending on a variety of hosts.
  • the transformant (transfectant) in which the host is Escherichia sp. or Bacillus sp. can be cultivated suitably in a liquid culture medium.
  • a preferred culture medium may contain carbon sources, nitrogen sources, minerals, and other elements, necessary for growing the transformant.
  • the carbon source may include, for example, glucose, dextrin, soluble starch, sucrose, etc.
  • the nitrogen source may include, for example, organic or inorganic substances such as ammonium salts, nitrates, corn steep liquor, peptone, casein, meat extracts, soybean-cakes, potato extracts.
  • Examples of the minerals are calcium chloride, sodium dihydrogen phosphate, magnesium chloride, etc. It may also be supplemented with yeast extracts, vitamins, growth promoting factors, etc.
  • the pH of the medium is desirably about 5 to 8.
  • a preferable example of the medium for culturing transformants wherein the host is Escherichia sp. is an M9 medium containing glucose and casamino acid (Miller, Journal of Experiments in Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972). If necessary, the medium may be supplemented with a drug such as 3 ⁇ -indolyl acrylic acid in order to make a promoter work efficiently.
  • the transformant is usually cultured at about 15 to 43° C. for about 3 to 24 hr. If necessary, aeration and stirring may be optionally applied.
  • the transformant When the host is Bacillus sp., the transformant is usually cultured at about 30 to 40° C. for about 6 to 24 hr. If necessary, aeration and stirring may be optionally applied.
  • examples of the medium for culturing the transformant are Burkholder minimum medium [Bostian, K. L. et al., Proc. Natl. Acad. Sci. USA, Vol. 77, 4505 (1980)], SD medium containing 0.5% casamino acid [Bitter, G. A. et al., Proc. Natl. Acad. Sci. USA, Vol. 81, 5330 (1984)], and others.
  • the pH of the medium is desirably about 5 to about 8.
  • Culturing is performed usually at about 20° C. to 35° C. for about 24 to 72 hr. If necessary, aeration and stirring may be optionally applied.
  • the medium used herein for culturing the transformant whose host is an insect cell or insect includes, for example, Grace's Insect Medium [Grace, T. C. C., Nature, Vol. 195, 788 (1962)] appropriately supplemented with an additive such as 10% inactivated bovine serum.
  • the pH of the medium is desirably about 6.2 to 6.4. Cultivation is usually carried out at about 27° C. for about 3 to 5 days. If necessary, aeration and stirring may be optionally applied.
  • the medium used herein for culturing the transformant whose host is an animal cell includes, for example, about 5 to 20% fetal bovine serum-containing minimum essential medium (MEM) [Science, Vol. 122, 501 (1952)], Dulbecco's modified Eagle's medium (DMEM) [Virology, Vol. 8, 396 (1959)], RPMI 1640 medium (The Journal of the American Medical Association, Vol. 199, 519 (1967)), 199 medium [Proceeding of the Society for the Biological Medicine, Vol. 73, 1 (1950)] and others.
  • MEM fetal bovine serum-containing minimum essential medium
  • DMEM Dulbecco's modified Eagle's medium
  • RPMI 1640 medium The Journal of the American Medical Association, Vol. 199, 519 (1967)
  • 199 medium Proceeding of the Society for the Biological Medicine, Vol. 73, 1 (1950)
  • the pH of the medium is desirably about 6 to 8.
  • Cultivation is usually carried out
  • the protein of the present invention can be extracted, isolated and/or purified from the resulting culture of said transformant according to methods known per se.
  • the proteins of the present invention are extracted from cultured bacteria or cell cytoplasms, the following techniques are appropriately used: the bacteria or cells are collected by a known method, next suspended in a suitable buffer, and then disrupted by sonication, lysozyme digestion and/or freeze-thawing, followed by centrifugation or filtration to afford a crude soluble protein extract.
  • the buffer may contain a protein-denaturing agent such as urea or guanidine hydrochloride, or a detergent such as Triton X-100.
  • the proteins of the present invention are extracted from the membrane fraction, the following processes are used: the bacteria or cells are disrupted in the same manner as aforementioned, and centrifuged at low speed to remove precipitated cell debris. The resultant supernatant is then centrifuged at high speed to precipitate a cell membrane-containing fraction (if necessary, the cell membrane fraction may be purified by density gradient centrifugation).
  • the proteins of the present invention are secreted outside of the microorganism (cell), culture supernatants are separated from the culture by centrifugation or filtration, and the resulting supernatants are collected.
  • the protein of the present invention contained in the resultant soluble fraction, membrane fraction or culture supernatant can be separated, isolated and/or purified according to methods known per se.
  • Such techniques used herein include methods of utilizing solubility such as salting-out and solvent precipitation; methods of utilizing mainly differences in molecular size such as dialysis, ultrafiltration, gel filtration and SDS-polyacrylamide gel electrophoresis (SDS-PAGE); methods of utilizing differences in electrical charge such as ion exchange chromatography; methods of utilizing specific affinity such affinity chromatography; methods of utilizing hydrophobic differences such as reversed-phase high-performance liquid chromatography (HPLC); methods of utilizing different isoelectric points such as isoelectric focusing; and other methods. These methods can be appropriately combined.
  • the protein or peptide When the protein or peptide is in a free form, it can be converted into its salt by methods known per se or modifications thereof. When the protein or peptide is obtained in the form of a salt, it can be converted into the free form or the form of another salt by methods known per se or modifications thereof.
  • the protein of the present invention produced by the transformant may be subjected to treatment with a suitable protein-modifying enzyme, before or after purification, to modify it appropriately or to partly remove a polypeptide fragment.
  • suitable protein-modifying enzyme examples include trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, and glycosidase.
  • the presence of the resultant protein of the present invention is determined by enzyme immunoassay or western blot with a specific antibody.
  • the protein of the present invention can be in vitro synthesized using, as a template, RNA corresponding to DNA coding for said protein in a cell-free protein translation system selected from the group consisting of rabbit reticulocyte lysate, wheat germ lysate, and others.
  • a cell-free protein translation system selected from the group consisting of rabbit reticulocyte lysate, wheat germ lysate, and others.
  • it can be synthesized using, as a template, DNA coding for the protein of the present invention in a cell-free transcription/translation system containing RNA polymerase.
  • the resultant proteins, peptide fragments thereof, antibodies and other molecules obtained herein can also be labeled, if necessary.
  • Labeling techniques are not limited to as long as they are conventionally known methods, but include preferably fluorescent labels, autoradiography, electron-dense materials, and insoluble pigment producing enzymes.
  • the labeling is a method of covalently binding with a fluorescent label.
  • Fluorescent substances to be used for the fluorescent label are not limited to, but include, for example, compounds having a fluorescent group such as pyrene, an anthraniloyl group, a dansyl group, fluorescein, rhodamine, and a nitrobenzoxadiazol group.
  • the compounds having the aforementioned fluorescent group are well known (see, for example, HIRATSUKA Toshiaki, “Tanpakushitsu Kakusan Kozo” (Protein Nucleic Acid and Enzyme), Vol. 42, No. 7 (1997), etc.), and can be incorporated into a protein molecule or peptide according to conventional methods.
  • the method of incorporating a foreign substance (non-self substance) into the envelope of an envelope virus or inactivated envelope virus with the protein of the present invention comprises the steps consisting of:
  • foreign substance-containing envelope viruses and inactivated envelope viruses can be obtained by the steps consisting of:
  • foreign substance-containing envelope viruses and inactivated envelope viruses can be obtained by the steps consisting of:
  • the treatment affecting the permeability of envelopes includes freeze-thawing, sonication, surface active agent addition and others.
  • the addition of surface active agents is more preferable.
  • the surface active agents used herein include octylglycoside, Triton-X 100, CHAPS, NP-40, etc. They can be used alone or in the form of combinations of two or more surface active agents.
  • the final concentration of surface active agents varies depending on kinds thereof but is 0.002 to 20%, desirably 0.02 to 2%.
  • the surface active agent in the application of the above method 1, it is desirable to remove the surface active agent prior to the step (3) of admixing with the foreign substance. In the application of the above method 2, it is desirable to remove the surface active agent after the step (3).
  • the removal of surface active agents includes methods of subjecting a mixture to centrifugation and discarding supernatants.
  • the surface active agent treatment is preferably conducted at low temperature, more preferably under ice-cooling.
  • envelope viruses and inactivated envelope viruses that contain foreign substances (non-self substances) within their envelopes allows the incorporation of said foreign substances into one or more cells.
  • Said foreign substances can be incorporated into one or more cells by contacting the cell or cells to be targeted for the incorporation with the envelope viruses and inactivated envelope viruses that contain the foreign substance (non-self substance) within their envelopes.
  • Cells to be targeted for the incorporation of foreign substances in the present invention are any of cultured cells and cells which compose tissues.
  • Preferred target cells are cultured cells.
  • Embodiments of the cells are any cell of mammals (e.g., human, bovine, simian, equine, swine, ovine, caprine, canine, feline, guinea pig, rat, mouse, rabbit, hamster, etc.).
  • Examples of such cells are hepatocytes, splenocytes, neurons, gliacytes, pancreatic ⁇ cells, myelocytes, mesangial cells, Langerhans' cells, epidermic cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, fibroblasts, fibrocytes, myocytes, fat cells (adipocytes), immune cells (e.g., macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), megakaryocytes, synovial cells, chondrocytes, bone cells (osteocytes), osteoblasts, osteoclasts, mammary gland cells and interstitial cells (Leydig's cells), precursor cells of these cells, stem cells and cancer cells and other cells.
  • immune cells e.g., macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eos
  • the targets for the incorporation of said foreign substances may be any tissue wherein one or more members of such cells exist, including, for example, brain, any part thereof (e.g., olfactory bulb, amygdaloid nucleus, cerebral basal ganglia, hippocampus, thalamus, hypothalamus, cerebral cortex, medulla oblongata, cerebellum), spinal cord, hypophysis, stomach, pancreas, kidney, liver, gonad, thyroid, gallbladder, bone marrow, adrenal gland, skin, lung, gastrointestinal tract (e.g., large intestine and small intestine), blood vessel, heart, thymus, spleen, submandibular gland, peripheral blood, prostate, testis, ovary, placenta, uterus, bone, joint, adipose tissue (e.g., brown adipose tissue, white adipose tissue), skeletal muscle and other tissues.
  • brain any part thereof (
  • a DNA fragment coding for the ZZ region which is the tandem repeat dimer of said region, was inserted into the site downstream of the T7 promoter of inducible expression vector pET28a (Novagen).
  • the above-described ZZ region DNA fragment was prepared by performing PCR using pEZZ18 (Amersham Biosciences) as a template.
  • Primers used in PCR were primer 1 (5′-GCAAATGCCATGGAACACGATGAAGCCGTAGACAA-3′; SEQ ID NO: 7) and primer 2 (5′-GGATCACCAAGCTTTTAGCTCGAATTCGCGTCTAC-3′; SEQ ID NO: 8), for generating NcoI and Hind III restriction sites at the upstream and downstream ends of the ZZ region in the resulting DNA fragment, respectively.
  • the PCR was done using Ex TaqTM (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to Ex TaqTM, 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of pEZZ18.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 94° C. for 30 sec, at 60° C. for 30 sec and at 72° C. for 30 sec, for a total of 30 cycles.
  • the resultant DNA fragment was digested with restriction enzymes, NcoI and Hind III, and then inserted into the site derived by cleavage with NcoI and Hind III from pET28a (Novagen) to create inducible expression vector pETZZ.
  • DNA coding for HVJ nucleocapsid protein was RT-PCR amplified from genome RNA of HVJ Z strain (furnished by courtesy of Professor Kaneda, Osaka University, Japan).
  • the aforementioned virus which can be used herein is HVJ virus in HVJ-inoculated chicken egg chorioallantoic fluid obtainable in the same manner as disclosed in Example 1 of US Patent Application Publication No. 2004/0253272 A1. Purification of total RNA was done with Viral RNA Mini (Viogene) from virus-infected chicken egg chorioallantoic fluid.
  • Reverse transcription was carried out with Ready-To-GoTM You-Prime First-Strand Beads (Amersham Biosciences) using purified total RNA in combination with virus genome RNA-specific primers, primer 3 (5′-ACCAAACAAGAGAAAAAACATGTAT-3′; SEQ ID NO: 9).
  • primer 3 5′-ACCAAACAAGAGAAAAAACATGTAT-3′; SEQ ID NO: 9
  • 20 ⁇ l of RNase free water was added to 10 ⁇ l of total RNA solution (16 ng/ ⁇ l) and the resultant mixture was allowed to stand at 65° C. for 10 min, and then put on ice for 2 min.
  • the resulting mix was added to First-Strand Reaction Mix Beads, admixed with 3 ⁇ l of primer 3 (10 ⁇ M), and then allowed to stand at 37° C. for 1 hr to give a virus genome cDNA solution.
  • PCR was performed using as a template the resulting virus genome cDNA from the above reaction in combination with primers, primer 4 (5′-TGCCAAAGGATCCGATGGCCGGGTTGTTGAGCACC-3′; SEQ ID NO: 10) and primer 5 (5′-GCCTCGTCTCGAGCTAGATTCCTCCTACCCCAGCT-3′; SEQ ID NO: 11), for generating BamH I and Xho I restriction sites at the upstream and downstream ends of the DNA region coding for the nucleocapsid protein in the resulting DNA fragment, respectively.
  • the PCR was done using Ex TaqTM (Takara) in 49 ⁇ l of reaction solution (a reaction buffer attached to Ex TaqTM, 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with 1 ⁇ l of the virus genome cDNA solution.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 94° C. for 30 sec, at 60° C. for 30 sec and at 72° C. for 2 min, for a total of 30 cycles. As a result, a DNA sample (1 ⁇ g) containing DNA coding for the nucleocapsid protein was obtained.
  • HVJ nucleocapsid protein-encoding DNA was inserted into the downstream site of the ZZ protein-encoding DNA sequence of pETZZ, obtained in the above Example 1, to create plasmid pETZZNP for expressing fusion proteins.
  • the DNA fragment, obtained in the above Example 2 was cleaved with BamH I and Xho I, and then inserted into the site, derived by cleavage with BamH I and Xho I from pETZZ, to create plasmid pETZZNP for expressing a fusion protein (ZZ-NP) with HVJ nucleocapsid protein on the C-terminal end of ZZ protein.
  • Fusion protein ZZ-NP was inducibly expressed in E. coli using T7 expression system.
  • E. coli Rosetta (DE3) (Novagen) was transformed with pETZZNP, obtained in the above Example 3, and the resulting transformants (transformed cells) were incubated in LB culture medium (30 ml) containing kanamycin (final concentration: 20 ⁇ g/ml) and chloramphenicol (final concentration: 34 ⁇ g/ml) at 37° C. overnight.
  • the resultant culture was used as a preculture solution.
  • This preculture solution was transferred in 1 L of LB culture medium containing kanamycin (final concentration: 20 ⁇ g/ml) and chloramphenicol (final concentration: 34 ⁇ g/ml), and incubated.
  • the crude extract was centrifuged (at 4° C., 20,000 g for 30 min), and the resulting supernatant was subjected to affinity purification using IgG SepharoseTM 6 Fast Flow (Amersham Biosciences) according to attached manuals.
  • the column was equilibrated with 0.5M acetic acid (pH3.4), TST (50 mM Tris buffer, pH7.6, 150 mM NaCl, 0.05% Tween 20) and loaded with the above supernatant.
  • the column was washed with TST, 5 mM NH 4 Ac (pH5.0), and then eluted with 0.5M acetic acid (pH3.4).
  • the fusion protein containing eluate (2 ml) was dialyzed against 300 mL of PBS for 2 hr, and further against 300 ml of PBS overnight, with dialysis membrane, Spectra/Por Membrane MWCO: 1,000 (Spectrum). After the dialysis, the resulting fusion protein solution (2 ml) was sterilized by filtration through a 0.22 ⁇ m filter. A fusion protein ZZ-NP solution (2 ml) with the fusion protein ZZ-NP concentration of 2 mg/mL was obtained.
  • the suspension was centrifuged (at 4° C., 10,000 g, for 5 min) and supernatants were removed.
  • To the resultant pellets was gently added 20 ⁇ l of PBS, the mixture was centrifuged (4° C., 10,000 g, 5 min), and supernatants were removed.
  • To the resultant pellets was added 10 ⁇ l of PBS to form a suspension.
  • To the above suspension was also added 10 ⁇ l of 2 ⁇ sample buffer and the mixture was boiled for 5 min. The resulting mixture was applied to SDS-PAGE (10% acrylamide gel). After the electrophoresis, samples were electroblotted (60 min, 2 mA/cm 2 ) onto a PVDF membrane (ATTO).
  • mouse IgG is present in inactivated HVJ envelope pellets receiving the above treatment.
  • ZZ-NP used increased the amounts of incorporated antibodies in a dose-dependent manner (lane 4: 0.8 ⁇ g, lane 5: 4 ⁇ g, lane 6: 20 ⁇ g).
  • HeLa S3 cells (furnished by courtesy of Professor Kaneda, Osaka University, Japan) were seeded at 1 ⁇ 10 cells/0.7 cm 2 , and incubated in a carbon dioxide gas incubator overnight.
  • To the resulting cultured cells was added 5 ⁇ l of Anti-NPC antibody-containing inactivated HVJ envelope suspension, obtained in the above Example 7, and the cells were placed in a carbon dioxide gas incubator.
  • the cells were washed twice with PBS, and fixed by treatment with a PBS solution containing 4% paraformaldehyde for 15 min.
  • the cells were washed twice with PBS, and then perfused in a PBS solution containing 0.2% Triton X-100 for 5 min.
  • the cells were washed twice with PBS for 5 min, and then treated with a PBS solution containing 1% BSA for 10 min. Next, the cells were allowed to stand in a PBS solution containing 1% BSA and 4 ⁇ g/ml Alexa Fluor 488-labeled Goat Anti-Mouse IgG, F(ab′) 2 fragment (Invitrogen) for 1 hr. After washing 3 times with PBS for 5 min at the final step, the cells were observed using confocal laser microscope FV300 (OLYMPUS, Japan) equipped with an Argon laser ( FIG. 3 ).
  • Nucleocapsid-encoding DNA of pETZZNP obtained in the above Example 3, was partly deleted to construct pET-ZZK1 to 4.
  • primer 6 (5′-TAGCTCGAGCACCACCAC-3′; SEQ ID NO: 20) in combination with primer 7 (5′-GTTTGCCAGATGATGTCT-3′; SEQ ID NO: 21); primer 6 (5′-TAGCTCGAGCACCACCAC-3′; SEQ ID NO: 20) in combination with primer 8 (5′-AAGGTATGTCCTCCCTGT-3′; SEQ ID NO: 22); and primer 6 (5′-TAGCTCGAGCACCACCAC-3′; SEQ ID NO: 20) in combination with primer 9 (5′-AACTATCTGGATGTTCTT-3′; SEQ ID NO: 23), respectively.
  • the PCR was done using Pyrobest DNA Polymerase (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to PyrobestTM DNA Polymerase, 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of pETZZNP.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 98° C. for 10 sec, at 60° C. for 30 sec and at 72° C. for 7 min, for a total of 30 cycles.
  • the resultant DNA fragments were subjected to self-ligation to form pET-ZZK1, pET-ZZK2 and pET-ZZK3.
  • PCR was performed using as a template pET-ZZK1 in combination with primers, primer 10 (5′-CGGATCCCCGGGTACCGAGCTCGAATT-3′; SEQ ID NO: 24) and primer 11 (5′-GGGAACTACATCCGAGATGCAG-3′; SEQ ID NO: 25) in the same fashion as in the above (1) to create pET-ZZK4.
  • Fusion proteins ZZ-NPK1, ZZ-NPK2, ZZ-NPK3, and ZZ-NPK4 were inducibly expressed in E. coli using T7 expression system. Specifically, E. coli Rosetta (DE3) (Novagen) was transformed with pET-ZZK1, pET-ZZK2, pET-ZZK3, and pET-ZZK4, respectively, obtained in the above Example 9, and the resulting transformants (transformed cells) were incubated in LB culture medium (30 ml) containing kanamycin (final concentration: 20 ⁇ g/ml) and chloramphenicol (final concentration: 34 ⁇ g/ml) at 37° C. overnight. Each of the resultant cultures was used as a preculture solution.
  • E. coli Rosetta (DE3) (Novagen) was transformed with pET-ZZK1, pET-ZZK2, pET-ZZK3, and pET-ZZK4, respectively, obtained in the above Example 9, and the resulting transformants (
  • This preculture solution was transferred in 1 L of LB culture medium containing kanamycin (final concentration: 20 ⁇ g/ml) and chloramphenicol (final concentration: 34 ⁇ g/ml), and incubated.
  • kanamycin final concentration: 20 ⁇ g/ml
  • chloramphenicol final concentration: 34 ⁇ g/ml
  • expression was induced by adding 1 ml of 1M IPTG solution.
  • the culture was centrifuged (at 4° C., 6,000 rpm for 15 min), and cells were collected and stored overnight at ⁇ 20° C.
  • the resultant cells were suspended in 20 ml of TST solution (50 mM Tris, pH7.6, 150 mM NaCl, 0.05% Tween 20) and then disrupted by sonication.
  • the crude extract was centrifuged (at 4° C., 20,000 g for 30 min), and the resulting supernatant was subjected to affinity purification using IgG Sepharose 6 Fast Flow (Amersham Biosciences) according to attached manuals. Briefly, the column was equilibrated with 0.5M acetic acid (pH3.4), TST (50 mM Tris buffer, pH7.6, 150 mM NaCl, 0.05% Tween 20) and loaded with the above supernatant. The column was washed with TST, 5 mM NH 4 Ac (pH5.0), and then eluted with 0.5M acetic acid (pH3.4) to obtain target products.
  • TST 50 mM Tris buffer, pH7.6, 150 mM NaCl, 0.05% Tween 20
  • the fusion protein containing eluate (2 ml) was dialyzed against 300 mL of PBS for 2 hr, and further against 300 ml of PBS overnight, with dialysis membrane, Spectra/Por Membrane MWCO: 1,000 (Spectrum). After the dialysis, the resulting fusion protein solution (2 ml) was sterilized by filtration through a 0.22 ⁇ m filter to give target fusion protein, ZZ-NPK1, ZZ-NPK2, ZZ-NPK3, or ZZ-NPK4.
  • NPK1 is shown in SEQ ID NO: 26 and 27, NPK2 in SEQ ID NO: 28 and 29, NPK3 in SEQ ID NO: 30 and 31, NPK4 in SEQ ID NO: 32 and 33, respectively.
  • the resulting respective ZZ fusion protein-containing inactivated HVJ envelope was suspended in 10 ⁇ l of antibody solution (1 mg/ml Mouse IgG (SIGMA), PBS) and then allowed to stand on ice for 5 min. The suspension was centrifuged (at 4° C., 10,000 g, for 5 min) and supernatants were removed. To the resultant pellets was gently added 10 ⁇ l of PBS, the mixture was recentrifuged (4° C., 10,000 g, 5 min), and supernatants were removed. To the resultant pellets was added 10 ⁇ l of PBS to form a suspension. To the above suspension was also added 10 ⁇ l of 2 ⁇ sample buffer and the mixture was boiled for 5 min.
  • antibody solution (1 mg/ml Mouse IgG (SIGMA), PBS
  • the resulting mixture was applied to SDS-PAGE (10% acrylamide gel). After the electrophoresis, samples were electroblotted (60 min, 2 mA/cm 2 ) onto a PVDF membrane (ATTO). After the blotting, blocking was done by dipping this PVDF membrane into TBS-T solution (0.1% Tween 20-containing TBS) containing 5% skim milk for 1 hr.
  • this blocked PVDF membrane was washed with TBS-T for 5 min, and then dipped into a TBS-T solution containing Peroxidase-conjugated AffiniPure F(ab′) 2 Fragment Goat Anti-Mouse IgG (H+L) (Jackson Immuno Research Laboratories) and 0.1% BSA for 1 hr. Thereafter, 5 min washing with a TBS-T solution was repeated 3 times and mouse IgG was then detected with ECL PLUS western blotting detection reagents kit (Amersham Biosciences) ( FIG. 4 ). It has been disclosed that mouse IgG is present in inactivated HVJ envelope pellets receiving the above treatments with fusion proteins. Among fusion proteins used, the highest incorporation level of mouse IgG into the inactivated HVJ envelope was observed when ZZ-NPK1 was used.
  • HeLa S3 cells (furnished by courtesy of Professor Kaneda, Osaka University, Japan) were seeded at 1 ⁇ 10 4 cells/0.7 cm 2 , and incubated in a carbon dioxide gas incubator overnight.
  • To the resulting cultured cells was added 10 ⁇ l of Anti-NPC Ab-containing inactivated HVJ envelope suspension, obtained in the above Example 12, and the mixture was placed in a carbon dioxide gas incubator.
  • the cells were washed twice with PBS, and fixed by treatment with a PBS solution containing 4% paraformaldehyde for 15 min.
  • the cells were washed twice with PBS, and then perfused in a PBS solution containing 0.2% Triton X-100 for 5 min.
  • the cells were washed twice with PBS for 5 min, and then treated with a PBS solution containing 1% BSA for 10 min. Next, the cells were allowed to stand in a PBS solution containing 1% BSA and 4 ⁇ g/ml Alexa Fluor 488-labeled Goat Anti-Mouse IgG, F(ab′) 2 fragment (Invitrogen) for 1 hr. After washing 3 times with PBS for 5 min at the final step, the cells were analyzed using confocal laser microscope FV300 (OLYMPUS, Japan) equipped with an Argon laser. For controls, the same treatments were repeated using a solution containing Anti-NPC Ab alone in place of the above-described Anti-NPC Ab-containing inactivated HVJ envelope suspension.
  • PCR was performed using as a template pET28a (Novagen) in combination with primers, primer 12 (5′-GAAGCGGTTGATGCTGCTACCGCAGAAAAAGTTTTCAAACAGTA CGCTAACAGCATGACTGGTGGACAG-3′: SEQ ID NO: 34) and primer 13 (5′-AGTAGTGGTTTCGCCTTTCAAGGTTTTACCATTCAGGATTAATT TGTAGGTCATGGTATATCTCCTTCT-3′: SEQ ID NO: 35).
  • the PCR was done using Pyrobest DNA Polymerase (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to Pyrobest DNA Polymerase, 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of template plasmid.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 98° C. for 10 sec, at 58° C. for 30 sec and at 72° C. for 6 min, for a total of 30 cycles.
  • the resultant DNA fragment was subjected to self-ligation to form pETG1 coding for an half (N-terminal side) of the Protein G C1 region.
  • PCR was performed using as a template pETG1, in combination with primers, primer 14 (5′-GTTACTGAAAAACCGGAAGTGATCGATGCGTCTGAATTAACCCC GGCGGTGACCCTGAGATCCGGCTGCTAA-3′: SEQ ID NO: 36) and primer 15 (5′-GGTAAAGGTTTTAGTCGCATCGTCGTAGGTCCATTCACCGTCAA CACCGTTGTCGTTAGCGTACTGTTTGAA-3′; SEQ ID NO: 37).
  • primer 14 5′-GTTACTGAAAAACCGGAAGTGATCGATGCGTCTGAATTAACCCC GGCGGTGACCCTGAGATCCGGCTGCTAA-3′: SEQ ID NO: 36
  • primer 15 5′-GGTAAAGGTTTTAGTCGCATCGTCGTAGGTCCATTCACCGTCAA CACCGTTGTCGTTAGCGTACTGTTTGAA-3′; SEQ ID NO: 37.
  • the PCR was done in the same fashion as above, followed by self-ligation, to obtain pET
  • PCR was performed using as a template pETG2, in combination with primers, primer 16 (5′-ATGCGTCCGGCGTAGA-3′; SEQ ID NO: 38) and primer 17 (5′-TAGCAGTTGGAATTCGCGGTCACCGCCGGGGTTAA-3′; SEQ ID NO: 39).
  • the PCR was done using Ex Taq (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to Ex Taq (Takara), 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of template plasmid.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 94° C. for 30 sec, at 58° C. for 30 sec and at 72° C. for 1 min, for a total of 30 cycles.
  • the resultant DNA fragment was digested with restriction enzymes, Xba I and EcoR I, and then inserted into the site derived by cleavage with Xba I and EcoR I from pET-ZZNPK1 to create plasmid pET-GNPK1 for expressing a fusion protein composed of Protein G C1 region and NPK1.
  • PCR was performed using as a template pETG2 in combination with primers, primer 18 (5′-AGGAGATAGAATTCTACCTACAAATTAATCCTGAA-3′; SEQ ID NO: 40) and primer 17 (5′-AAGGTATGTCCTCCCTGT-3′; SEQ ID NO: 39).
  • the PCR was done in the same fashion as above.
  • the resultant DNA fragment was digested with restriction enzyme, EcoR I, and then inserted into the site derived by cleavage with EcoR I from pET-GNPK1 to create plasmid pET-GGNPK1 for expressing a fusion protein composed of the tandem repeat dimer of Protein G C1 region and NPK1.
  • PCR was performed using as a template pET28a (Novagen) in combination with primers, primer 19 (5′-AACCTGATCTTTGCGAACGGCAGCACCCAGACCGCGGAATTTAAA GGCACCTTTGAAAAAAGCATGACTGGTGGACAG-3′; SEQ ID NO: 41) and primer 20 (5′-CGCTTTGATGGTCACTTCTTCTTCGCTATCGGTTTCCGGGGTTTC CGGGGTTTCTTCTTTCATGGTATATCTCCTTCT-3′; SEQ ID NO: 42).
  • the PCR was done using Pyrobest DNA Polymerase (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to Pyrobest DNA Polymerase, 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of template plasmid.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 98° C. for 10 sec, at 58° C. for 30 sec and at 72° C. for 6 min, for a total of 30 cycles.
  • the resultant DNA fragment was subjected to self-ligation to form pETL1 coding for an half (N-terminal side) of the Protein L B1 region.
  • PCR was performed using as a template pETL1 in combination with primers, primer 21 (5′-TGAATATACCGTGGATGTGGCGGATAAAGGTTATACCCTGAA CATTAAATTTGCGGGTTTTTTGCTGAAAGGA-3′; SEQ ID NO: 43) and primer 22 (5′-CCGTTATCTTTTTTCAGGGTATCCGCATACGCATACGCTTCGC TGGTCGCTTTTTCAAAGGTACCTTTAA-3′; SEQ ID NO: 44).
  • primer 21 5′-TGAATATACCGTGGATGTGGCGGATAAAGGTTATACCCTGAA CATTAAATTTGCGGGTTTTTTGCTGAAAGGA-3′; SEQ ID NO: 43
  • primer 22 5′-CCGTTATCTTTTTTCAGGGTATCCGCATACGCATACGCTTCGC TGGTCGCTTTTTCAAAGGTACCTTTAA-3′; SEQ ID NO: 44.
  • the PCR was done in the same fashion as above, followed by self-ligation, to obtain pETL2
  • PCR was performed using as a template pETL2, in combination with primers, primer 23 (5′-ATGCGTCCGGCGTAGA-3′; SEQ ID NO: 45) and primer 24 (5′-TCCCCTGTCGAATTCGCACCCGCAAATTTAATGT-3′; SEQ ID NO: 46).
  • the PCR was done using Ex Taq (Takara), in 50 ⁇ l of reaction solution (a reaction buffer attached to Ex Taq (Takara), 200 ⁇ M each of dATP, dTTP, dCTP and dGTP, and 400 nM each of said primers) admixed with about 1 ng of template plasmid.
  • the amplification was performed using GeneAmp PCR system 9700 (PE Applied Biosystems) set to repeat steps at 94° C. for 30 sec, at 58° C. for 30 sec and at 72° C. for 1 min, for a total of 30 cycles.
  • the resultant DNA fragment was digested with restriction enzymes, Xba I and EcoR I, and then inserted into the site derived by cleavage with Xba I and EcoR I from pET-ZZNPK1 to create plasmid pET-LNPK1 for expressing a fusion protein composed of Protein L B1 region and NPK1.
  • PCR was also performed using as a template pETL2 in combination with primers, primer 25 (5′-AGGAGATAGAATTCTAAAGAAGAAACCCCGGAAAC-3′; SEQ ID NO: 47) and primer 24 (5′-AAGGTATGTCCTCCCTGT-3′; SEQ ID NO: 46).
  • the PCR was done in the same fashion as above.
  • the resultant DNA fragment was digested with restriction enzyme, EcoR I, and then inserted into the site derived by cleavage with EcoR I from pET-LNPK1 to create plasmid pET-LLNPK1 for expressing a fusion protein composed of the tandem repeat dimer of Protein L B1 region and NPK1.
  • Fusion proteins G-NPK1, GG-NPK1, L-NPK1 and LL-NPK1 were inducibly expressed in E. coli expression systems utilizing T7 RNA polymerase.
  • E. coli Rosetta (DE3) (Novagen) was transformed with pET-GNPK1 and pET-GGNPK1, obtained in the above Example 14, and pET-LNPK1 and pET-LLNPK1, obtained in the above Example 15, and treated in the same fashion as in Example 10 to give target fusion proteins G-NPK1, GG-NPK1, L-NPK1 and LL-NPK1, respectively, provided that, for eluents upon the affinity purification of G-NPK1 and GG-NPK1, 0.1M glycine-HCl (pH2.7) was used in place of 0.5M acetic acid (pH3.4).
  • the resulting G-NPK1, GG-NPK1, L-NPK1 or LL-NPK1-containing inactivated HVJ envelopes were suspended in 5 ⁇ l of antibody solution (1 mg/ml Rat IgG2b (R&D systems)) and then allowed to stand on ice for 5 min. The suspension was centrifuged (at 4° C., 10,000 g, for 5 min) and supernatants were removed. To the resultant pellets was gently added 20 ⁇ l of PBS, the mixture was recentrifuged (4° C., 10,000 g, 5 min), and supernatants were removed. To the resultant pellets was added 10 ⁇ l of PBS to form a suspension.
  • antibody solution 1 mg/ml Rat IgG2b (R&D systems)
  • the resultant G-NPK1, GG-NPK1, L-NPK1 or LL-NPK1-containing inactivated HVJ envelopes were suspended in 5 ⁇ l of antibody solution (rat IgG2b (R&D systems), 1 mg/ml) After standing on ice for 5 min, the mixture was centrifuged (4° C., 10,000 g, 5 min) and supernatants were removed. To the resulting rat IgG2b-containing inactivated HVJ envelopes was added 12.5 ⁇ l of PBS, and further 12.5 ⁇ l of 1 mg/ml protamine sulfate solution to form a suspension.
  • HeLa S3 cells (furnished by courtesy of Professor Kaneda, Osaka University, Japan) were seeded at 1 ⁇ 10 4 cells/0.7 cm 2 , and incubated in a carbon dioxide gas incubator (5% CO 2 , 37° C.) overnight.
  • a carbon dioxide gas incubator 5% CO 2 , 37° C.
  • To the resulting cultured cells was added 10 ⁇ l of rat IgG2b-containing inactivated HVJ envelope suspension, obtained in the above Example 18, and the mixture was placed in a carbon dioxide gas incubator. Four hours later, the cells were washed twice with PBS, and fixed by treatment with a PBS solution containing 4% paraformaldehyde for 15 min.
  • the cells were washed twice with PBS, and then perfused in a PBS solution containing 0.2% Triton X-100 for 5 min. After the treatment, the cells were washed twice with PBS for 5 min, and then treated with a PBS solution containing 1% BSA for 10 min. Next, the cells were allowed to stand in a PBS solution containing 1% BSA and 4 ⁇ g/ml Alexa Fluor 488-labeled chicken Anti-rat IgG (Invitrogen) for 1 hr at room temperature. After washing 3 times with PBS for 5 min at the final step, the cells were analyzed using confocal laser microscope FV300 (OLYMPUS, Japan) equipped with an Argon laser ( FIG. 8 ). It has been verified that rat IgG2b antibodies are present homogeneously within cytoplasms transfected with rat IgG2b Ab-containing inactivated HVJ envelopes.
  • the resulting G-NPK1, GG-NPK1, L-NPK1 or LL-NPK1-containing inactivated HVJ envelopes were suspended in 5 ⁇ l of antibody solution (Anti-NPC Ab, 1 mg/ml). After standing on ice for 5 min, the suspension was centrifuged (4° C., 10,000 g, 5 min) and supernatants were removed. To the resulting Anti-NPC Ab-containing inactivated HVJ envelopes was added 12.5 ⁇ l of PBS, and further 12.5 ⁇ l of 1 mg/ml protamine sulfate solution to form a suspension.
  • proteins containing a envelope viral component-compatible polypeptide, and envelope viruses or inactivated envelope viruses in combination with said proteins enables proteins, antibodies, and other foreign substances (non-self substances) to be incorporated into one or more cells without damaging their properties, functions and structures, and such intracellular delivery techniques are time and quantity controllable and facilitate efficient introduction of foreign substances into cells.
  • the present invention provides targeting techniques for a wide variety of cells including various adhesive and suspension cells and primarily cultured cells and applicable to animals (in vivo), as well as techniques which can adapt, combinations of molecules to be intracellularly introduced with targets thereof, toward diverse applications.
  • applications of the present invention facilitate in vivo and in vitro function analysis in unknown genes, in vivo and in vitro function analysis in targeted genes, all aspects of gene therapy including clinical studies and clinical trials, studies to evaluate the onset mechanisms of diseases and to prevent the disease onset, DDS (Drug Delivery System) studies and uses, function examinations of novel useful materials and new drug candidates and application studies thereof, and applications to Drug Discovery.
  • the present invention allows the development and research of antineoplastic actions, cytotoxic actions and control actions such as gene expression.
US12/223,464 2006-01-31 2007-01-31 Polypeptide Having Affinity for Envelope Virus Constituent and Use Thereof in Transferring Substance Into Cell Abandoned US20090053790A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2006-021882 2006-01-31
JP2006021882 2006-01-31
JP2006-230513 2006-08-28
JP2006230513 2006-08-28
PCT/JP2007/051587 WO2007088882A1 (ja) 2006-01-31 2007-01-31 エンベロープウイルスの構成成分に親和性を有するポリペプチドと細胞内物質導入への使用

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/051587 A-371-Of-International WO2007088882A1 (ja) 2006-01-31 2007-01-31 エンベロープウイルスの構成成分に親和性を有するポリペプチドと細胞内物質導入への使用

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/546,359 Division US8388979B2 (en) 2006-01-31 2012-07-11 Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell

Publications (1)

Publication Number Publication Date
US20090053790A1 true US20090053790A1 (en) 2009-02-26

Family

ID=38327453

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/223,464 Abandoned US20090053790A1 (en) 2006-01-31 2007-01-31 Polypeptide Having Affinity for Envelope Virus Constituent and Use Thereof in Transferring Substance Into Cell
US13/546,359 Active US8388979B2 (en) 2006-01-31 2012-07-11 Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/546,359 Active US8388979B2 (en) 2006-01-31 2012-07-11 Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell

Country Status (4)

Country Link
US (2) US20090053790A1 (ja)
EP (1) EP1980615A4 (ja)
JP (1) JP5102635B2 (ja)
WO (1) WO2007088882A1 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9493543B2 (en) 2010-02-16 2016-11-15 Novo Nordisk A/S Factor VIII fusion protein

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5645155B2 (ja) * 2009-08-31 2014-12-24 国立大学法人 琉球大学 分子量マーカー及び分子量マーカーの作製方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6913923B2 (en) * 2000-02-02 2005-07-05 Anges Mg, Inc. Virus envelope vector for gene transfer
WO2006001120A1 (ja) * 2004-06-24 2006-01-05 Dnavec Research Inc. マイナス鎖rnaウイルスを含む抗癌剤

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9400088D0 (sv) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US6432699B1 (en) * 1997-03-28 2002-08-13 New York University Viral vectors having chimeric envelope proteins containing the IgG-binding domain of protein A
JP3942362B2 (ja) 2000-02-02 2007-07-11 アンジェスMg株式会社 遺伝子導入のためのウイルスエンベロープベクター
WO2003014338A1 (fr) 2001-08-02 2003-02-20 Anges Mg, Inc. Procede de production d'enveloppes virales inactivees
US20040028687A1 (en) * 2002-01-15 2004-02-12 Waelti Ernst Rudolf Methods and compositions for the targeted delivery of therapeutic substances to specific cells and tissues
CA2494485A1 (en) 2002-07-25 2004-02-05 Medimmune, Inc. Methods of treating and preventing rsv, hmpv, and piv using anti-rsv, anti-hmpv, and anti-piv antibodies
JP3901653B2 (ja) * 2003-03-20 2007-04-04 東邦瓦斯株式会社 熱風発生バーナ
JP2005052083A (ja) 2003-08-05 2005-03-03 Rikogaku Shinkokai タンパク質の細胞内導入方法
JP2005343874A (ja) * 2004-06-07 2005-12-15 Ishihara Sangyo Kaisha Ltd 不活性化ウイルスエンベロープを用いる細胞などへの外来物質の導入方法

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6913923B2 (en) * 2000-02-02 2005-07-05 Anges Mg, Inc. Virus envelope vector for gene transfer
WO2006001120A1 (ja) * 2004-06-24 2006-01-05 Dnavec Research Inc. マイナス鎖rnaウイルスを含む抗癌剤
US20080031855A1 (en) * 2004-06-24 2008-02-07 Dnavec Research Inc. Anticancer Agent Containing Minus-Strand Rna Virus

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Fujii et al., "Conserved and non-conserved regions in the Sendai virus genome: evolution of a gene possessing overlapping reading frames," 22(1): 47-52 (2001). *
Fujii et al., "Identification of mutations associated with attenuation of virulence of a field Sendai virus isolate by egg passage." Virus Genes 25:189-193(2002) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9493543B2 (en) 2010-02-16 2016-11-15 Novo Nordisk A/S Factor VIII fusion protein

Also Published As

Publication number Publication date
EP1980615A4 (en) 2010-04-21
EP1980615A1 (en) 2008-10-15
JPWO2007088882A1 (ja) 2009-06-25
US8388979B2 (en) 2013-03-05
WO2007088882A1 (ja) 2007-08-09
US20120301961A1 (en) 2012-11-29
JP5102635B2 (ja) 2012-12-19

Similar Documents

Publication Publication Date Title
EP1991560B1 (en) Peptide having cell membrane penetrating activity
JP4188909B2 (ja) 細胞質残留性細胞膜透過ペプチド及びこれの用途{CytoplasmicTransductionPeptidesandUsesthereof}
US20040265300A1 (en) Chimeric molecules containing a module able to target specific cells and a module regulating the apoptogenic function of the permeability transition pore complex (PTPC)
CN101437833A (zh) 作为心脏功能调节剂的磷酸酶抑制剂蛋白-1
KR102060411B1 (ko) 세포 침투성 펩타이드, 이를 포함하는 융합 화합물 및 이 융합 화합물을 포함하는 약학 조성물
WO2021087282A1 (en) Recombinant cdkl5 proteins, gene therapy and production methods
AU2003244303A1 (en) Methods for transducing fusion molecules
US8388979B2 (en) Polypeptide having affinity for envelope virus constituent and use thereof in transferring substance into cell
CA3103684A1 (en) Solubilized apyrases, methods and use
JP2022549057A (ja) 分子の細胞内送達のための複合体
US20020142355A1 (en) Methods for the in vivo biotin labeling of polypeptides
US6395875B1 (en) Recombinant soluble adenovirus receptor
KR101263424B1 (ko) 세포투과성 엔도스타틴 재조합 단백질, 이를 코딩하는 폴리뉴클레오티드 및 이를 유효성분으로 함유하는 항암 조성물
KR101636538B1 (ko) 인간 NLBP 유래의 NP2 폴리펩티드 또는 dNP2 폴리펩티드를 포함하는 세포 투과 펩티드 및 이를 이용한 카고 전달 시스템
EP4086275A1 (en) Multimerization delivery system for intracellular delivery of molecule
US20140303093A1 (en) Micro-utrophin polypeptides and methods
KR20090122769A (ko) 세포내로 단백질을 전달하는 방법 및 이를 위한 펩타이드
EP1949907B1 (en) Novel application of heparin-binding epidermal growth factor-like growth factor for medical purposes
KR101636542B1 (ko) 인간 nlbp 유래의 np12 폴리펩티드 또는 np21 폴리펩티드를 포함하는 세포 투과 펩티드 및 이를 이용한 카고 전달 시스템
WO2024034200A1 (ja) 膜貫通タンパク質を標的オルガネラに局在化させるためのペプチドリンカー及び局在化方法並びに局在化する融合タンパク質
KR102556731B1 (ko) 단백질 수송 도메인, 이를 포함하는 융합 화합물 및 이 융합 화합물을 포함하는 약학 조성물
KR102267495B1 (ko) 미토콘드리아 타겟팅용 폴리펩타이드 및 그 용도
KR101705603B1 (ko) 엔테로박테리아세아 유래 세포 투과 단백질 및 이의 용도
JP2003169673A (ja) タンパク質分解排除酵素とその用途
KR20140046995A (ko) 인간 nlbp 단백질 유래의 np1 폴리펩티드를 포함하는 세포 투과 펩티드 및 이를 이용한 카고 전달 시스템

Legal Events

Date Code Title Description
AS Assignment

Owner name: ISHIHARA SANGYO KAISHA, LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FUSHIKIDA, KUNI;KONDO, YOSHITAKA;REEL/FRAME:021359/0034

Effective date: 20080716

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION