US20040254347A1 - Method of cross-linking peptides - Google Patents

Method of cross-linking peptides Download PDF

Info

Publication number
US20040254347A1
US20040254347A1 US10/307,562 US30756202A US2004254347A1 US 20040254347 A1 US20040254347 A1 US 20040254347A1 US 30756202 A US30756202 A US 30756202A US 2004254347 A1 US2004254347 A1 US 2004254347A1
Authority
US
United States
Prior art keywords
cross
linking
sequence
peptide
transglutaminase
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
US10/307,562
Inventor
Han-Jia Lin
Yee-Hsiung Chen
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.)
Academia Sinica
Original Assignee
Academia Sinica
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 Academia Sinica filed Critical Academia Sinica
Priority to US10/307,562 priority Critical patent/US20040254347A1/en
Assigned to ACADEMIA SINICA reassignment ACADEMIA SINICA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, YEE-HSIUNG, LIN, HAN-JIA
Publication of US20040254347A1 publication Critical patent/US20040254347A1/en
Priority to US11/649,176 priority patent/US7396656B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/104Aminoacyltransferases (2.3.2)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1075General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of amino acids or peptide residues
    • 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

Definitions

  • the present invention relates to a method of cross-linking peptides through a novel amino acid sequence Q-X-K-(S/T), which was initially identified from mouse seminal vesicle secretion (SVS) III protein.
  • the peptides containing Q-X-K-(S/T) sequence may be cross-linked by any transgluatminase (TGase).
  • Transgluatminase is a kind of enzyme with the ability to catalyze protein-protein cross-linking reaction.
  • Transglutaminase was first reported by Heinrich Waelsch in 1959. He isolated the enzyme from the liver of guinea pig. In the presence of calcium ion, this enzyme shows trans-amidation activity which binding glutamine in proteins to primary amines covalently. Therefore this enzyme was designed as transglutaminase (EC.2.3.2.13). In recent years, more transglutaminases have been purified from different spices and different tissues.
  • TGases can be divided into five types: (1) tissue type TGase (TG2), which is commonly expressed in all kinds of tissues, might involve cell-programmed death; (2) epidermal TGase (TG1), found in the wounded epidermal tissues, enable epidermal proteins to cross-link into keratin; (3) hair-follicle TGase, found in hair-follicle cell, can cross-link hair protein; (4) plasma factor XIII, when catalyzed by thrombin, it can cross-link fibrin to stabilize the structure of thrombus; (5) prostate TGase (TG4), found in the coagulating gland of rodent, can catalyze seminal vesicle secreted proteins to copulatory plug.
  • tissue type TGase TG2
  • epidermal TGase TG1
  • hair-follicle TGase found in hair-follicle cell
  • plasma factor XIII when catalyzed by thrombin,
  • TGases Although these different kinds of TGases have great difference in size and sequences, their catalytic mechanisms are similar. Each of them has a cystine residue in their active site and the enzyme activity is calcium dependent. TGase has great usages in industry. For examples: adding TGase to meat will increase its tenacity and savor in food processing; Enzymes can be fixed by cross-linking of TGase in enzyme engineering; TGase has been used to construct tissues' frame in tissue engineering et cetera.
  • TGase has two substrates: one is usually defined as glutamine in a peptides chain and serves as an acyl donor; the other should be a primary amine, which serves as an acyl acceptor.
  • TGases demand more specificity of acyl donor than of acyl acceptor in usual. Therefore, lots of polyamines (spermine and histamine for example) also can be acyl acceptors and covalently bind with proteins as a kind of post-translational modification.
  • this invention didn't define characteristics and effects of the amino acids in Y position. Moreover, the peptide fragments in this invention were only effective under the action of human plasma factor XIII that limits the usage of other sources of TGases and also restricts the utilities of these peptide fragments. Besides, the length of the defmed fragment was too long and the reaction efficiency was low.
  • the purpose of this invention is to take the reactive site in SVS III protein as the substrate of transglutaminase. It provides a novel way to cross-link, fix or polymerize proteins by TGase.
  • the present invention defines the reactive site of TGase from mouse SVS III gene sequence (SEQ ID NO: 1).
  • the minimum effective unit of reaction site includes four amino acids (SEQ ID NO: 2), which are defined as Q-X-K-(S/T).
  • Q represents glutamine
  • X represents aliphatic side chain of amino acids (Leu, Val, Ile, Ala etc., for example);
  • K is lysine;
  • (S/T) can be serine or threonine.
  • the examples of this invention claim that the minimum effective unit can be the substrate of TGase.
  • peptides When peptides have repetitive minimum effective units (SEQ ID NO: 3 including five minimum effective units, for example), they are better substrates of TGase.
  • the rearranged sequence of minimum effective unit also can be the substrate of TGase.
  • “Cross-linking fragment” is defined as peptides with one or more minimum effective unit or with deformed sequence. Accordingly, the present invention encompasses a peptide for cross-linking, comprising a -QX- sequence at the N-terminal and a -XK(S/T) sequence at the C-terminal, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine.
  • cross-linking fragment is a good substrate for TGase from different sources including plasma factor XIII, TGase of guinea pig and TGase in mouse coagulate land secretion.
  • This peptide fragment can be synthesized directly (Merrifiedld, R. B. 1963. J. Amer. Chem. Soc. 85, 2149-2154), or produced by constructing the cDNA sequence in recombinant plasmid to produce fusion protein.
  • the fusion protein containing the cross-linking fragment can be cross-linked to the plastic surface with primary amine by TGase, which retains its enzyme activity.
  • FIG. 1 depicts construction of the recombinant plasmid, where (A) is a map of the recombinant expression vector, pGEX-4T and the sites of restriction enzymes recognized, and (B) shows sequences of each pair of synthesized nucleotides in each clone were aligned in the annealed form;
  • FIG. 2 shows cross-linking of GST fusion proteins by guinea pig liver transglutaminase, where GST protein (lanes 1 and 2), FP#1 (lanes 3 and 4), FP#2 (lanes 5 and 6) and FP#3 (lanes 7 and 8) were cross-linked by guinea pig liver transglutaminase in a reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2) with (lanes 1, 3, 5, and 7) or without (lanes 2, 4, 6 and 8) 50 mM EDTA;
  • a reaction buffer 50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2
  • FIG. 3 shows that a fusion protein containing QXKS/T is a good substrate for different sources of transglutaminase, where FP#1 was cross-linked by different sources of transglutaminases, including mouse coagulating gland fluid (C), human blood factor XIII (F), and guinea pig transglutaminase (T), in a reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2) with (lanes 1, 3, and 5) or without (lanes 2, 4, and 6) 50 mM EDTA; and
  • C mouse coagulating gland fluid
  • F human blood factor XIII
  • T guinea pig transglutaminase
  • FIG. 4 shows that GST fused with QXKS/T tandem repeats segment can be fixed on the surface of primary amine containing microplate, where (A) shows GST activity in the reaction mixture after the cross-linking reaction, and (B) shows GST activity in the well of microplate after the cross-linking reaction (see “example 2” for detail experiment condition).
  • This Data represent the means of three experiments, and error bars represent S.D.
  • cross-linking fragment means the peptide having a Q-X-K-(S/T) sequence, wherein Q represents glutamine, X represents amino acids having an aliphatic side chain such as Leu, Val, Ile, Ala etc.; K is lysine; (S/T) can be either serine or threonine; the term “fusion protein”, “fusion peptide” or “fusion polypeptide” means a peptide containing fragments from different origins.
  • TGase represents transglutaminase
  • GST represents glutathione S-transferase
  • All fusion proteins identified in the present invention were prepared by inserting a cross-linking fragment into GST sequence by recombinant DNA technology.
  • E. Coli expression vector, pGEX4-T (Amersham-Pharmacia, Freiburg, Germany), was used to produce fusion proteins.
  • Restriction enzymes, Bam HI and EcoRI, were used to cut the pGEX4-T vector.
  • the vector was then purified by 1% agarose gel electrophoresis and recovered from the gel by QiagelElution kit (Qiagen, Hilden, Germany).
  • a sense and an anti-sense DNA fragment encoded each protein fragment were synthesized based on mouse SVS III cDNA (SEQ ID NO: 1).
  • a BamHI site and an EcoRI site were added during nucleotide synthesizing (cf. FIG. 1).
  • 20 ⁇ l of both strands of the synthesized oligonucleotides, in the concentration of 100 ⁇ M, were mixed together and heated to 95° C. for 10 min, then annealed at 55° C. for 10 min and room temperature for another 10 min.
  • the annealed insert DNA fragment was mix with the BamHI/EcoRI-treated pGEX4-T vector and reacted by T4 DNA ligase at 4° C. overnight.
  • the reaction mixture was use to transform host cells, E. Coli strain JM109, by conventional transformation technology. Positive clones, which were able to produce a fusion polypeptide were screened and identified by DNA sequencing.
  • each bacteria clone was transferred into 100 ml of LB broth containing 100, ⁇ g/ml ampicillin and cultured at 37° C. with 200 rpm shaking overnight.
  • the bacteria broth was subcultured in 900 ml LB medium containing 100 ⁇ g/ml ampicillin in the next morning.
  • OD 600 of the bacteria broth reached 0.6
  • IPTG 0.5 mM of final conc.
  • the pellet was resuspensed by 10 ml of phosphate buffer saline and mixed with complete protease inhibitor cocktail (Roch, Germany).
  • the bacteria suspension was sonicated for 5 min and the cell lysate was centrifuged at 15,000 rpm for 20 min. The supernatant, so-called the crude extract, was collected for further purification.
  • the fusion proteins were purified from the crude extract by affinity chromatography on a column of glutathione agarose bead (Amersham-Pharmacia, Freiburg, Germany). Glutathione agarose was packaged into a column with 20 mm inner diagram. The height of the gel was 40 mm. 50 ml of phosphate buffer saline was flowed through the column for equivalence. The crude extract was loaded into the column and then another 50 ml of PBS was flowed through to wash out unbound proteins. Finally, 20 ml of elution buffer (10 mM reduction form glutathione in PBS) was applied into the column and the purified protein was collected. After dialysis and measuring protein concentration, the recombinant proteins were adjusted to 1 ⁇ g/ml and stored in ⁇ 20° C.
  • each fusion protein was mixed with tissue transglutaminase from guinea pig's liver in 40 ⁇ l reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl 2 pH7.5) and the reaction mixture was incubated at 37° C. for 1 hour. Since the action of transglutaminase is calcium dependent, every experiment had a control, which substitute 50 mM EDTA for 7.5 mM CaCl 2 . After 1 hour of incubation, 40 ⁇ l of 2 ⁇ Laemmli sample buffer was added to stop the reaction. Each reaction result was resolved by SDS-PAGE.
  • GST itself is not a substrate of TGase (lane 1) and was not cross-linked by the enzyme, while GST fused with insert SEQ ID NO:2 (Fusion Protein No:1, FP#1, lane 3), SEQ ID NO:3 (FP#2, lane 5) and SEQ ID NO:4 (FP#3, lane 7) showed the ability to be cross-linked by transglutaminase.
  • SEQ ID NO:2 Fusion Protein No:1, FP#1, lane 3
  • SEQ ID NO:3 FP#2, lane 5
  • SEQ ID NO:4 FP#3, lane 7
  • the presence of EDTA in the reaction mixture prevents the fusion proteins to form polymers, because of the lack of calcium ion.
  • the enzyme-catalyzed cross-links of FP#2 were very striking (cf. lane 5 of FIG. 2). There were almost no FP#2 monomer left after the enzyme reaction.
  • FP#2 was intermolecularly cross-linked by the enzyme reaction.
  • FP#1 was cross-linked to a dimer by the enzyme reaction (cf. lane 3 of FIG. 2), manifesting the transglutaminase substrate activity of the short peptide QIKS.
  • the enzyme was able to cross-link FP#3, which was a mutant of FP#2 with a QI at the N-terminal and a KS at the C-terminal of the cross-linking segment but the inner glutamine (Q) and lysine (K) residues were replaced by glycine residues.
  • the four-peptide segment of QXK(S/T) is the essential sequence for cross-linking by transglutaminase. While one segment of QXK(S/T) is sufficient for the transglutaminase-catalyzed protein cross-linking, the more QXK(S/T) repeats the fusion protein contains, the stronger is the cross-linking ability.
  • the sequence of QXK(S/T) can also be rearranged and still maintain its cross-linking ability by transglutaminase.
  • Fusion proteins containing QXK(S/T) sequences are not only cross-linked by guinea pig liver transglutaminase but also good substrates to other types of transglutaminases.
  • Thrombin-activated factor XIII (F), guinea pig liver transglutaminase (T), or mouse coagulating gland fluid (M) was incubated with FP#1 (15 ⁇ g) in 40 ⁇ l reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl 2 pH7.5) and incubated at 37° C. for 1 hour.
  • transglutaminase Since the action of transglutaminase is calcium dependent, every experiment had a control, which 7.5 mM CaCl 2 was substituted by 50 mM EDTA. After 1 hour of incubation, 40 ⁇ l of 2 ⁇ Laemmli sample buffer was added to stop the reaction and the reaction mixture was resolved by SDS/PAGE on a 14% gel slab.
  • FP#1 with the essential sequence of QXKS/T, is a good substrate of transglutaminases from different sources, including mouse coagulating gland transglutaminase (TG4), human blood factor XIII and guinea pig liver transglutaminase (TG2).
  • TG4 mouse coagulating gland transglutaminase
  • TG2 human blood factor XIII
  • TG2 guinea pig liver transglutaminase
  • transglutaminase Since transglutaminase has the ability to transfer an acyl group from a molecule to a primary amine so as to form a covalent bond, it is possible to fix a fusion protein which containing QXK(S/T) sequence to a solid phase having primary amine on its surface by the action of transglutaminase.
  • the reaction mixture contained 1 ⁇ g of FP#2, 0.1 ⁇ g of guinea pig liver transglutaminase, 50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl 2 in pH 7.5.
  • reaction mixture was loaded into wells of a microplate which contains primary amine on the surface (COSTAR amine surface stripwell, Corning, USA.) and incubated at 37° C. for 2 hrs.
  • CaCl 2 in the reaction was replaced by 50 mM EDTA.
  • the supernatant in each well was collected to a microcentrifuge tube and the wells were washed twice by PBS.
  • the supernatant was then mixed with 1 ml of assay reagent (100 mM potassium phosphate buffer pH6.5, 1 mM glutathione, 1 mM 1-Chloro-2,4-dinitrobezene) to test the enzyme activity of GST.
  • the reaction was carried out at 37° C.
  • transglutaminase can be used to fix a fusion protein having QXK(S/T) sequence to a solid phase.
  • the reaction mixture contained 50 ⁇ g of FP#2 as an antigen, 1 ⁇ g of guinea pig liver transglutaminase, 50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl 2 in pH 7.5.
  • the reaction was carried on at 37° C. for 1 hrs.
  • the CaCl 2 in the reaction was replaced by 50 mM EDTA in the control.
  • an equal volume of Freund's incomplete adjuvant was added into each reaction mixture and mixed well with the reaction mixture to form an antigen injection mixture.
  • mice were challenged with the same antigen and received the second challenge after another 3 weeks.
  • the antiserum was collected two weeks after the final challenge.
  • the FP#2 protein was resolved by SDS-PAGE and transferred to a nitrocellulose membrane. After transfer, the protein blots were immunodetected by the Western blot procedure, using the antiserum as the primary antibody diluted to 1:10000 in a blocking solution (5% nonfat skimmed milk in PBS), and a goat anti-mouse IgG was conjugated with horseradish peroxidase as the secondary antibody diluted to 1:10000 in the blocking solution.
  • the enzyme-staining bands were enhanced by chemiluminescence detection using an ECL kit (Amersham-Pharmacia, Freiburg, Germany) according to the manufacturer's instruction.
  • the result showed a strong immunoreaction to the antigen (FP#2), while the signal in the control serum was weak.
  • FP#2 the antigen
  • the antigenicity of the antigen was improved by the polymerization of the FP#2 through the action of transglutaminase.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A method of cross-linking a peptide to form a homopolymer of the peptide, to immobilize the peptide on a solid phase and to enhance antigenicity of the peptide is disclosed. The method comprises the steps of preparing a fusion peptide by incorporating a cross-linking segment including a tetrapeptide sequence QXK(S/T) into the peptide and cross-linking the peptide by a glutaminase.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0001]
  • The present invention relates to a method of cross-linking peptides through a novel amino acid sequence Q-X-K-(S/T), which was initially identified from mouse seminal vesicle secretion (SVS) III protein. The peptides containing Q-X-K-(S/T) sequence may be cross-linked by any transgluatminase (TGase). [0002]
  • 2. Description of the Related Art [0003]
  • Cross-linking of biomolecules has been noted for decades throughout the world of biotechnology. Cross-linking of proteins with different functions can produce a new molecule with multi functions. Enzymes cross-link to a solid phase makes its activity retaining that can reduce manufactory costs. Biomolecules can be cross-linked by chemical reactions while these reactions are not very specific and probably reduced the activity of enzymes. An alternative way to cross-link biomolecules is enzyme-catalyzed reaction. [0004]
  • Transgluatminase (TGase) is a kind of enzyme with the ability to catalyze protein-protein cross-linking reaction. Transglutaminase (TGase) was first reported by Heinrich Waelsch in 1959. He isolated the enzyme from the liver of guinea pig. In the presence of calcium ion, this enzyme shows trans-amidation activity which binding glutamine in proteins to primary amines covalently. Therefore this enzyme was designed as transglutaminase (EC.2.3.2.13). In recent years, more transglutaminases have been purified from different spices and different tissues. Until now, TGases can be divided into five types: (1) tissue type TGase (TG2), which is commonly expressed in all kinds of tissues, might involve cell-programmed death; (2) epidermal TGase (TG1), found in the wounded epidermal tissues, enable epidermal proteins to cross-link into keratin; (3) hair-follicle TGase, found in hair-follicle cell, can cross-link hair protein; (4) plasma factor XIII, when catalyzed by thrombin, it can cross-link fibrin to stabilize the structure of thrombus; (5) prostate TGase (TG4), found in the coagulating gland of rodent, can catalyze seminal vesicle secreted proteins to copulatory plug. Although these different kinds of TGases have great difference in size and sequences, their catalytic mechanisms are similar. Each of them has a cystine residue in their active site and the enzyme activity is calcium dependent. TGase has great usages in industry. For examples: adding TGase to meat will increase its tenacity and savor in food processing; Enzymes can be fixed by cross-linking of TGase in enzyme engineering; TGase has been used to construct tissues' frame in tissue engineering et cetera. [0005]
  • However, there are limits in industrial applications for TGase. First, most TGase was isolated from animal source and they are also difficult to prepare by recombinant techniques. Second, most TGases have specificities to their substrates that limits the application of TGase. J. E. Folk and his group made a series of studies on tissue type TGase of guinea pig's liver and human's plasma factor XIII (Gorman, J. J. and Folk, J. E. 1980. J. Biol. Chem. 2255,4419-427; Schhrode, J. and Folk, J. E. 1979. J. Biol. Chem. 254,653-661). Their works may help to understand the substrate specificity of TGase. TGase has two substrates: one is usually defined as glutamine in a peptides chain and serves as an acyl donor; the other should be a primary amine, which serves as an acyl acceptor. TGases demand more specificity of acyl donor than of acyl acceptor in usual. Therefore, lots of polyamines (spermine and histamine for example) also can be acyl acceptors and covalently bind with proteins as a kind of post-translational modification. [0006]
  • Some works have been done to define the effective sites from substrates of TGases to produce cross-linking peptides fragment. For example, in U.S. Pat. Nos. 5,428,014 and 5,939,385, peptide sequences from human plasma fibrinogen have been studied. These peptide fragments are proved to be cross-linkable by human plasma factor XIII and this characteristic has been applied in tissue engineering. This invention generalized an S1-Y-S2 formula from plasma fibrinogen. In this case, SI represents T-I-G-E-G-Q, Y is 0˜7 interval amino acids, and S2 is X-K-X-A-G-D-V (U.S. Pat. No. 5,428,014, claim [0007] 1). Yet, this invention didn't define characteristics and effects of the amino acids in Y position. Moreover, the peptide fragments in this invention were only effective under the action of human plasma factor XIII that limits the usage of other sources of TGases and also restricts the utilities of these peptide fragments. Besides, the length of the defmed fragment was too long and the reaction efficiency was low.
  • In the present invention, we have found a better substrate of TGase from other sources. Seminal vesicles secretions of rodent have been reported as good substrate of TGase (Notides, A. C. and Williams-Ashman, H. G. 1967. Proc. Natl. Acad. Sci. U.S.A. 58, 1991-1995). Notides and Williams-Ashman found a protein (18 kDa) secreted from guinea pig's seminal vesicle. This protein can readily be polymerized by a TGase secreted from coagulating gland. Following study also proved that SVS II protein from mouse and rat seminal vesicle secretions are substrates of TGase (Harris, S. E. et. al. 1990. J. Biol. Chem. 265, 9896-9903; Lundwall, A. et al. 1997. Eur. J. Biochem. 249,39-44). Though human seminal secretions will not solidify to become copulatory plugs, it has been proved that SgI and SgII proteins from human seminal vesicle are also substrates of TGase (Peter, A. et. A1. 1998. Eur. J. Biochem. 252, 216-221). However, the molecular mechanism of these proteins have never been studied. In this invention, we isolated a new protein, SVS III, from mouse seminal vesicle and proved it a good substrate of transglutaminase. The present invention also provides an effective sequence from SVS III and related applications. [0008]
  • SUMMARY OF THE INVENTION
  • The purpose of this invention is to take the reactive site in SVS III protein as the substrate of transglutaminase. It provides a novel way to cross-link, fix or polymerize proteins by TGase. The present invention defines the reactive site of TGase from mouse SVS III gene sequence (SEQ ID NO: 1). The minimum effective unit of reaction site includes four amino acids (SEQ ID NO: 2), which are defined as Q-X-K-(S/T). Q represents glutamine, X represents aliphatic side chain of amino acids (Leu, Val, Ile, Ala etc., for example); K is lysine; (S/T) can be serine or threonine. The examples of this invention claim that the minimum effective unit can be the substrate of TGase. [0009]
  • When peptides have repetitive minimum effective units (SEQ ID NO: 3 including five minimum effective units, for example), they are better substrates of TGase. [0010]
  • Examples in this invention claim that the rearranged sequence of minimum effective unit (SEQ ID NO: 4) also can be the substrate of TGase. “Cross-linking fragment” is defined as peptides with one or more minimum effective unit or with deformed sequence. Accordingly, the present invention encompasses a peptide for cross-linking, comprising a -QX- sequence at the N-terminal and a -XK(S/T) sequence at the C-terminal, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine. [0011]
  • Examples in this invention also prove that the cross-linking fragment is a good substrate for TGase from different sources including plasma factor XIII, TGase of guinea pig and TGase in mouse coagulate land secretion. [0012]
  • This peptide fragment can be synthesized directly (Merrifiedld, R. B. 1963. J. Amer. Chem. Soc. 85, 2149-2154), or produced by constructing the cDNA sequence in recombinant plasmid to produce fusion protein. In examples of this invention, we construct the cDNA sequence in recombinant plasmid to produce fusion protein with this cross-linking fragment. Therefore, the fusion protein has the ability to cross-link by TGase. In examples of this invention, we explain how to produce a fusion protein with cross-linking fragment. [0013]
  • The fusion protein containing the cross-linking fragment can be cross-linked to the plastic surface with primary amine by TGase, which retains its enzyme activity. [0014]
  • In examples of this invention, we also proved that fusion protein with the cross-linking fragment could become polymer, which can induce stronger immune reaction when it is injected into animals as an antigen. This method makes an improvement on the production of vaccines. [0015]
  • Other objects and features of the present invention will become apparent from the following detailed description considered in conjunction with the accompanying drawings. It is to be understood, however, that the drawings are designed solely for purposes of illustration and not as a definition of the limits of the invention, for which reference should be made to the appended claims. It should be further understood that the drawings are not necessarily drawn to scale and that, unless otherwise indicated, they are merely intended to conceptually illustrate the structures and procedures described herein. [0016]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • In the drawings: [0017]
  • FIG. 1 depicts construction of the recombinant plasmid, where (A) is a map of the recombinant expression vector, pGEX-4T and the sites of restriction enzymes recognized, and (B) shows sequences of each pair of synthesized nucleotides in each clone were aligned in the annealed form; [0018]
  • FIG. 2 shows cross-linking of GST fusion proteins by guinea pig liver transglutaminase, where GST protein ([0019] lanes 1 and 2), FP#1 (lanes 3 and 4), FP#2 (lanes 5 and 6) and FP#3 (lanes 7 and 8) were cross-linked by guinea pig liver transglutaminase in a reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2) with ( lanes 1, 3, 5, and 7) or without ( lanes 2, 4, 6 and 8) 50 mM EDTA;
  • FIG. 3 shows that a fusion protein containing QXKS/T is a good substrate for different sources of transglutaminase, where [0020] FP#1 was cross-linked by different sources of transglutaminases, including mouse coagulating gland fluid (C), human blood factor XIII (F), and guinea pig transglutaminase (T), in a reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2) with ( lanes 1, 3, and 5) or without ( lanes 2, 4, and 6) 50 mM EDTA; and
  • FIG. 4 shows that GST fused with QXKS/T tandem repeats segment can be fixed on the surface of primary amine containing microplate, where (A) shows GST activity in the reaction mixture after the cross-linking reaction, and (B) shows GST activity in the well of microplate after the cross-linking reaction (see “example 2” for detail experiment condition). This Data represent the means of three experiments, and error bars represent S.D. [0021]
  • DETAILED DESCRIPTION OF THE PRESENTLY PREFERRED EMBODIMENTS
  • As used herein, the terms “cross-linking fragment”, “cross-linking sequence”, “cross-linking segment”, or “minimum effective unit” means the peptide having a Q-X-K-(S/T) sequence, wherein Q represents glutamine, X represents amino acids having an aliphatic side chain such as Leu, Val, Ile, Ala etc.; K is lysine; (S/T) can be either serine or threonine; the term “fusion protein”, “fusion peptide” or “fusion polypeptide” means a peptide containing fragments from different origins. [0022]
  • “TGase” represents transglutaminase; [0023]
  • “GST” represents glutathione S-transferase. [0024]
  • The present invention is further explained and illustrated in the following examples, which represent particular embodiments of, but not limitations to, the present invention. [0025]
  • EXAMPLE 1 Preparation of Fusion Proteins Containing Cross-linking Effective Sequence
  • All fusion proteins identified in the present invention were prepared by inserting a cross-linking fragment into GST sequence by recombinant DNA technology. [0026] E. Coli expression vector, pGEX4-T (Amersham-Pharmacia, Freiburg, Germany), was used to produce fusion proteins. Restriction enzymes, Bam HI and EcoRI, were used to cut the pGEX4-T vector. To remove the restriction enzymes and small nucleotides, the vector was then purified by 1% agarose gel electrophoresis and recovered from the gel by QiagelElution kit (Qiagen, Hilden, Germany). A sense and an anti-sense DNA fragment encoded each protein fragment were synthesized based on mouse SVS III cDNA (SEQ ID NO: 1). At the beginning and the end of each DNA fragment, a BamHI site and an EcoRI site were added during nucleotide synthesizing (cf. FIG. 1). 20 μl of both strands of the synthesized oligonucleotides, in the concentration of 100 μM, were mixed together and heated to 95° C. for 10 min, then annealed at 55° C. for 10 min and room temperature for another 10 min. The annealed insert DNA fragment was mix with the BamHI/EcoRI-treated pGEX4-T vector and reacted by T4 DNA ligase at 4° C. overnight. The reaction mixture was use to transform host cells, E. Coli strain JM109, by conventional transformation technology. Positive clones, which were able to produce a fusion polypeptide were screened and identified by DNA sequencing.
  • To produce the recombinant proteins, each bacteria clone was transferred into 100 ml of LB broth containing 100, μg/ml ampicillin and cultured at 37° C. with 200 rpm shaking overnight. The bacteria broth was subcultured in 900 ml LB medium containing 100 μg/ml ampicillin in the next morning. When OD[0027] 600 of the bacteria broth reached 0.6, IPTG (0.5 mM of final conc.) were added into the broth and continuously cultured at 37° C. with 200 rpm shaking for 5 hours. Then the broth was centrifuged at 5,000 rpm for 10 min and discarded the supernatant. The pellet was resuspensed by 10 ml of phosphate buffer saline and mixed with complete protease inhibitor cocktail (Roch, Germany). The bacteria suspension was sonicated for 5 min and the cell lysate was centrifuged at 15,000 rpm for 20 min. The supernatant, so-called the crude extract, was collected for further purification.
  • The fusion proteins were purified from the crude extract by affinity chromatography on a column of glutathione agarose bead (Amersham-Pharmacia, Freiburg, Germany). Glutathione agarose was packaged into a column with 20 mm inner diagram. The height of the gel was 40 mm. 50 ml of phosphate buffer saline was flowed through the column for equivalence. The crude extract was loaded into the column and then another 50 ml of PBS was flowed through to wash out unbound proteins. Finally, 20 ml of elution buffer (10 mM reduction form glutathione in PBS) was applied into the column and the purified protein was collected. After dialysis and measuring protein concentration, the recombinant proteins were adjusted to 1 μg/ml and stored in −20° C. [0028]
  • EXAMPLE 2 Cross-Linking Ability of The Fusion Proteins
  • To test the cross-linking abilities of different fusion proteins produced in Example 1, each fusion protein was mixed with tissue transglutaminase from guinea pig's liver in 40 μl reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl[0029] 2 pH7.5) and the reaction mixture was incubated at 37° C. for 1 hour. Since the action of transglutaminase is calcium dependent, every experiment had a control, which substitute 50 mM EDTA for 7.5 mM CaCl2. After 1 hour of incubation, 40 μl of 2× Laemmli sample buffer was added to stop the reaction. Each reaction result was resolved by SDS-PAGE.
  • As shown in FIG. 2, GST itself is not a substrate of TGase (lane 1) and was not cross-linked by the enzyme, while GST fused with insert SEQ ID NO:2 (Fusion Protein No:1, [0030] FP#1, lane 3), SEQ ID NO:3 (FP#2, lane 5) and SEQ ID NO:4 (FP#3, lane 7) showed the ability to be cross-linked by transglutaminase. The presence of EDTA in the reaction mixture prevents the fusion proteins to form polymers, because of the lack of calcium ion. Note worthily, the enzyme-catalyzed cross-links of FP#2 were very striking (cf. lane 5 of FIG. 2). There were almost no FP#2 monomer left after the enzyme reaction. Based on the relation between molecular size and protein mobility on SDS/PAGE, dimers, trimers, tetramers, pentamers and hexamers of FP#2 were clearly identified. Homopolymers larger than hexamers were also detected. Apparently, FP#2 was intermolecularly cross-linked by the enzyme reaction. FP#1 was cross-linked to a dimer by the enzyme reaction (cf. lane 3 of FIG. 2), manifesting the transglutaminase substrate activity of the short peptide QIKS. Likewise, the enzyme was able to cross-link FP#3, which was a mutant of FP#2 with a QI at the N-terminal and a KS at the C-terminal of the cross-linking segment but the inner glutamine (Q) and lysine (K) residues were replaced by glycine residues. Apparently, the four-peptide segment of QXK(S/T) is the essential sequence for cross-linking by transglutaminase. While one segment of QXK(S/T) is sufficient for the transglutaminase-catalyzed protein cross-linking, the more QXK(S/T) repeats the fusion protein contains, the stronger is the cross-linking ability. The sequence of QXK(S/T) can also be rearranged and still maintain its cross-linking ability by transglutaminase.
  • EXAMPLE 3 Cross-linking Abilities of the Fusion Proteins by Transglutaminases from Different Sources
  • Fusion proteins containing QXK(S/T) sequences are not only cross-linked by guinea pig liver transglutaminase but also good substrates to other types of transglutaminases. Thrombin-activated factor XIII (F), guinea pig liver transglutaminase (T), or mouse coagulating gland fluid (M) was incubated with FP#1 (15 μg) in 40 μl reaction buffer (50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl[0031] 2 pH7.5) and incubated at 37° C. for 1 hour. Since the action of transglutaminase is calcium dependent, every experiment had a control, which 7.5 mM CaCl2 was substituted by 50 mM EDTA. After 1 hour of incubation, 40 μl of 2× Laemmli sample buffer was added to stop the reaction and the reaction mixture was resolved by SDS/PAGE on a 14% gel slab.
  • As shown in FIG. 3, [0032] FP#1, with the essential sequence of QXKS/T, is a good substrate of transglutaminases from different sources, including mouse coagulating gland transglutaminase (TG4), human blood factor XIII and guinea pig liver transglutaminase (TG2).
  • EXAMPLE 4 Fixation of Fusion Protein Containing QXK(S/T) Sequence to a Solid Phase
  • Since transglutaminase has the ability to transfer an acyl group from a molecule to a primary amine so as to form a covalent bond, it is possible to fix a fusion protein which containing QXK(S/T) sequence to a solid phase having primary amine on its surface by the action of transglutaminase. In a volume of 50 μl, the reaction mixture contained 1 μg of [0033] FP#2, 0.1 μg of guinea pig liver transglutaminase, 50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2 in pH 7.5. The reaction mixture was loaded into wells of a microplate which contains primary amine on the surface (COSTAR amine surface stripwell, Corning, USA.) and incubated at 37° C. for 2 hrs. As a control, CaCl2 in the reaction was replaced by 50 mM EDTA. After reaction was complete, the supernatant in each well was collected to a microcentrifuge tube and the wells were washed twice by PBS. The supernatant was then mixed with 1 ml of assay reagent (100 mM potassium phosphate buffer pH6.5, 1 mM glutathione, 1 mM 1-Chloro-2,4-dinitrobezene) to test the enzyme activity of GST. The reaction was carried out at 37° C. for 5 min and the absorption at 340 nm was observed. The enzyme activity in the wells were also tested by pouring 100 μl of assay reagent (100 mM potassium phosphate buffer pH6.5, 1 mM glutathione, 1 mM 1-Chloro-2,4-dinitrobezene) into each well and incubated the plate at 37° C. for 5 min. The solution of each well was collected and its absorption at 340 nm was measured. A stronger absorption represents a higher enzyme activity. The highest enzyme activity was found in the control supernatant (FIG. 4A). However, under the action of transglutaminase, the GST activity of FP#2 was remained in the well of microplate (FIG. 4B). Thus, it demonstrates that transglutaminase can be used to fix a fusion protein having QXK(S/T) sequence to a solid phase.
  • EXAMPLE 5 Polymerization of the Fusion Proteins to Improve Antigenicity
  • Polymerization of an antigen containing QXK(S/T) sequence by transglutaminase can improve antigenicity. In a volume of 50 μl, the reaction mixture contained 50 μg of [0034] FP#2 as an antigen, 1 μg of guinea pig liver transglutaminase, 50 mM Tris-HCl, 150 mM NaCl and 7.5 mM CaCl2 in pH 7.5. The reaction was carried on at 37° C. for 1 hrs. The CaCl2 in the reaction was replaced by 50 mM EDTA in the control. After the reaction was completed, an equal volume of Freund's incomplete adjuvant was added into each reaction mixture and mixed well with the reaction mixture to form an antigen injection mixture. These antigen injection mixtures were used to inject 12-week-old female mice subcutaneously. Three weeks after the first boost, mice were challenged with the same antigen and received the second challenge after another 3 weeks. The antiserum was collected two weeks after the final challenge. The FP#2 protein was resolved by SDS-PAGE and transferred to a nitrocellulose membrane. After transfer, the protein blots were immunodetected by the Western blot procedure, using the antiserum as the primary antibody diluted to 1:10000 in a blocking solution (5% nonfat skimmed milk in PBS), and a goat anti-mouse IgG was conjugated with horseradish peroxidase as the secondary antibody diluted to 1:10000 in the blocking solution. The enzyme-staining bands were enhanced by chemiluminescence detection using an ECL kit (Amersham-Pharmacia, Freiburg, Germany) according to the manufacturer's instruction. The result showed a strong immunoreaction to the antigen (FP#2), while the signal in the control serum was weak. Apparently, the antigenicity of the antigen was improved by the polymerization of the FP#2 through the action of transglutaminase.
  • Thus, while there have shown and described and pointed out fundamental novel features of the invention as applied to a preferred embodiment thereof, it will be understood that various omissions and substitutions and changes in the form and details of the devices illustrated, and in their operation, may be made by those skilled in the art without departing from the spirit of the invention. For example, it is expressly intended that all combinations of those elements and/or method steps which perform substantially the same function in substantially the same way to achieve the same results are within the scope of the invention. Moreover, it should be recognized that structures and/or elements and/or method steps shown and/or described in connection with any disclosed form or embodiment of the invention may be incorporated in any other disclosed or described or suggested form or embodiment as a general matter of design choice. It is the intention, therefore, to be limited only as indicated by the scope of the claims appended hereto. [0035]
  • 1 15 1 265 PRT Murine sp. 1 Met Lys Ser Ile Phe Phe Ser Leu Ser Leu Leu Leu Leu Leu Glu Lys 1 5 10 15 Lys Ala Ala Gly Ile Glu Leu Tyr Ala Gly Gly Thr Lys Gly His Phe 20 25 30 Leu Val Lys Thr Ser Pro Leu Met Phe Ile Gly Lys Asn Gln Phe Leu 35 40 45 Tyr Gly His Lys Glu Glu Gln Glu Glu Ala Pro Glu Glu Ser Ile Phe 50 55 60 Val Gln Thr Lys His His Ala Tyr Gly Gln Asp Ala Asp Ala Asp Met 65 70 75 80 Gly Gly Ala Leu Ser Ser Gln Glu Leu Thr Ser Leu Lys Glu Asp Ile 85 90 95 Val Cys Glu Glu Glu Asp Glu Leu Ala Gln Gln Lys Ser Gln Leu Pro 100 105 110 Ser Gln Ser Gln Ile Lys Ser Gln Thr Gln Val Lys Ser Tyr Ala Ala 115 120 125 Gln Leu Lys Ser Gln Pro Gly Gln Leu Lys Thr Ile Gly Gln Val Lys 130 135 140 Ser Gln Thr Met Leu Lys Ser His Gly Ala Pro Leu Lys Ser Phe Lys 145 150 155 160 Ala Arg Leu Asn Leu Arg Glu Asp Ile Pro Gln Gln Val Lys Gly Arg 165 170 175 Gly Tyr Gly Leu Ala Glu Asp Leu Ala Gln Val Arg Gln Gln Pro Ala 180 185 190 Lys Val His Arg Leu Lys Gly Lys His Arg Gln Ser Arg Lys Thr Ala 195 200 205 Ala Phe Tyr Pro Gln Phe Arg Arg His Ser Arg Pro Tyr Pro Arg Tyr 210 215 220 Phe Val Gln Phe Gln Glu Gln Leu Gln Gly Ser Val His His Thr Lys 225 230 235 240 Ser Phe Tyr Pro Gly Pro Gly Met Cys Tyr Cys Pro Arg Gly Gly Val 245 250 255 Ile Leu Tyr Gln Asp Ala Phe Thr Asp 260 265 2 4 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 2 Gln Ile Lys Ser 1 3 30 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 3 Gln Ile Lys Ser Gln Thr Gln Val Lys Ser Tyr Ala Ala Gln Leu Lys 1 5 10 15 Ser Gln Pro Gly Gln Leu Lys Thr Ile Gly Gln Val Lys Ser 20 25 30 4 30 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 4 Gln Ile Gly Ser Gly Thr Gly Val Gly Ser Tyr Ala Ala Gly Leu Gly 1 5 10 15 Ser Gly Pro Gly Gly Leu Gly Thr Ile Gly Gly Val Lys Ser 20 25 30 5 18 DNA Artificial Sequence Description of Artificial Sequence Primer 5 gatcccaaat aaaatccg 18 6 96 DNA Artificial Sequence Description of Artificial Sequence Synthetic nucleotide sequence 6 gatcccaaat aaaatcccaa actcaagtaa aatcctacgc agcccaactg aagtcccaac 60 caggccagct aaaaaccata gggcaggtga agtcag 96 7 96 DNA Artificial Sequence Description of Artificial Sequence Synthetic nucleotide sequence 7 gatcccaaat aggttccggc actggggtag gttcctacgc agccggcctg ggttccgggc 60 caggcggtct aggtaccata gggggcgtga agtcag 96 8 57 DNA Artificial Sequence CDS (1)..(54) Description of Artificial Sequence Primer 8 ctg gtt ccg cgt gga tcc cca gga att ccc ggg tcg act cga gcg gcc 48 Leu Val Pro Arg Gly Ser Pro Gly Ile Pro Gly Ser Thr Arg Ala Ala 1 5 10 15 gca tcg tga 57 Ala Ser 9 18 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 9 Leu Val Pro Arg Gly Ser Pro Gly Ile Pro Gly Ser Thr Arg Ala Ala 1 5 10 15 Ala Ser 10 6 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 10 Thr Ile Gly Glu Gly Gln 1 5 11 7 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 11 Xaa Lys Xaa Ala Gly Asp Val 1 5 12 18 DNA Artificial Sequence Description of Artificial Sequence Primer 12 aattcggatt ttatttgg 18 13 96 DNA Artificial Sequence Description of Artificial Sequence Synthetic nucleotide sequence 13 aattctgact tcacctgccc tatggttttt agctggcctg gttgggactt cagttgggct 60 gcgtaggatt ttacttgagt ttgggatttt atttgg 96 14 95 DNA Artificial Sequence Description of Artificial Sequence Synthetic nucleotide sequence 14 aattctgact tcacgccccc tatggtacct agacgcctgg cccggaaccc aggccggctg 60 cgtaggaacc taccccagtg ccggaaccta tttgg 95 15 4 PRT Artificial Sequence Description of Artificial Sequence Synthetic peptide 15 Gln Xaa Lys Xaa 1

Claims (30)

We claim:
1. A method of cross-linking a peptide to form polymers of said peptide, comprising the steps of
(a) incorporating a cross-linking segment containing a Q-X-K-(S/T) sequence into said peptide to form a fusion polypeptide, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine; and
(b) cross-linking said fusion polypeptide by a transglutaminase in a cross-linking reaction buffer to form said polymers.
2. The method of claim 1, wherein said cross-linking segment comprises at least two tandem repeats of said Q-X-K-(S/T) sequence.
3. The method of claim 2, wherein said cross-linking segment comprises SEQ ID No: 3.
4. The method of claim 1, wherein said Q-X-K-(S/T) sequence is SEQ ID No: 2.
5. The method of claim 1, wherein said transglutaminase is from mouse coagulating gland fluid.
6. The method of claim 1, wherein said transglutaminase is from human blood factor XIII.
7. The method of claim 1, wherein said transglutaminase is guinea pig liver transglutaminase.
8. A method of cross-linking a peptide to form polymers of said peptide, comprising the steps of incorporating a cross-linking segment containing SEQ ID No: 4 into said peptide to form a fusion polypeptide and cross-linking said fusion polypeptide by a transglutaminase in a cross-linking reaction buffer to form said polymers.
9. A method of immobilize a peptide on a solid phase having a surface containing a primary amine moiety, comprising the steps of
(a) preparing a fusion polypeptide by incorporating a cross-linking segment containing a Q-X-K-(S/T) sequence into said peptide, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine; and
(b) cross-linking said fusion polypeptide with the surface of said solid phase in the presence of a cross-linking buffer by a transglutaminase to immobilize said fusion polypeptide.
10. The method of claim 9, wherein said cross-linking segment comprises at least two tandem repeats of said Q-X-K-(S/T) sequence.
11. The method of claim 10, wherein said cross-linking segment comprises SEQ ID No: 3.
12. The method of claim 9, wherein said Q-X-K-(S/T) sequence is SEQ ID No: 2.
13. The method of claim 9, wherein said transglutaminase is from mouse coagulating gland fluid.
14. The method of claim 9, wherein said transglutaminase is from human blood factor XIII.
15. The method of claim 9, wherein said transglutaminase is guinea pig liver transglutaminase.
16. A method of enhancing antigenicity of a peptide, comprising the steps of
(a) preparing a fusion polypeptide by incorporating a cross-linking segment containing a Q-X-K-(S/T) sequence into said peptide, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine;
(b) cross-linking said fusion polypeptide by a transglutaminase in a cross-linking buffer to form polymers of said peptide; and
(c) immunizing an animal by injecting said animal with said polymers of said peptide to produce an antibody against said peptide.
17. The method of claim 16, wherein said cross-linking segment comprises at least two tandem repeats of said Q-X-K-(S/T) sequence.
18. The method of claim 17, wherein said cross-linking segment comprises SEQ ID No: 3.
19. The method of claim 16, wherein said Q-X-K-(S/T) sequence is SEQ ID No: 2.
20. The method of claim 16, wherein said transglutaminase is from mouse coagulating gland fluid.
21. The method of claim 16, wherein said transglutaminase is from human blood factor XIII.
22. The method of claim 16, wherein said transglutaminase is guinea pig liver transglutaminase.
23. A peptide for cross-linking, comprising a -QX- sequence and a -XK(S/T) sequence, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine, said -QK- sequence and said -XK(S/T) sequence being spaced apart by at most 25 amino acid residues.
24. The peptide of claim 23 having a sequence identical to SEQ ID No: 4.
25. A fusion polypeptide, comprising a cross-linking segment containing a Q-X-K-(S/T) sequence, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine.
26. The fusion polypeptide of claim 25, wherein said cross-linking segment comprises at least two tandem repeats of said Q-X-K-(S/T) sequence.
27. The fusion polypeptide of claim 26, wherein said cross-linking segment comprises SEQ ID No: 3.
28. The fusion polypeptide of claim 25, wherein said Q-X-K-(S/T) sequence is SEQ ID No: 2.
29. A fusion polypeptide, comprising a cross-linking segment containing a -QX- sequence and a -XK(S/T)- sequence, wherein Q is glutamine, X is an amino acid having an aliphatic side chain, K is lysine and (S/T) is either serine or threonine, said -QK- sequence and said -XK(S/T) sequence being spaced apart by at most 25 amino acid residues
30. The fusion peptide of claim 23, wherein said cross-linking segment has a sequence identical to SEQ ID No: 4.
US10/307,562 2002-11-25 2002-11-25 Method of cross-linking peptides Abandoned US20040254347A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/307,562 US20040254347A1 (en) 2002-11-25 2002-11-25 Method of cross-linking peptides
US11/649,176 US7396656B2 (en) 2002-11-25 2007-01-03 Method of cross-linking peptides

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/307,562 US20040254347A1 (en) 2002-11-25 2002-11-25 Method of cross-linking peptides

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/649,176 Division US7396656B2 (en) 2002-11-25 2007-01-03 Method of cross-linking peptides

Publications (1)

Publication Number Publication Date
US20040254347A1 true US20040254347A1 (en) 2004-12-16

Family

ID=33510227

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/307,562 Abandoned US20040254347A1 (en) 2002-11-25 2002-11-25 Method of cross-linking peptides
US11/649,176 Expired - Fee Related US7396656B2 (en) 2002-11-25 2007-01-03 Method of cross-linking peptides

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/649,176 Expired - Fee Related US7396656B2 (en) 2002-11-25 2007-01-03 Method of cross-linking peptides

Country Status (1)

Country Link
US (2) US20040254347A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040001848A1 (en) * 2002-03-01 2004-01-01 Szu-Yi Chou Method of producing disease-specific antigens
US20040005654A1 (en) * 2002-03-01 2004-01-08 Szu-Yi Chou Method of producing polyvalent antigens

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030219853A1 (en) * 2002-03-01 2003-11-27 Szu-Yi Chou Method of cross-linking a compound
US7807624B2 (en) * 2006-01-11 2010-10-05 Affinergy, Inc. Methods and compositions for promoting attachment of cells of endothelial cell lineage to medical devices
WO2017147542A2 (en) 2016-02-26 2017-08-31 Regeneron Pharmaceuticals, Inc. Optimized transglutaminase site-specific antibody conjugation

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428014A (en) * 1993-08-13 1995-06-27 Zymogenetics, Inc. Transglutaminase cross-linkable polypeptides and methods relating thereto
US5939385A (en) * 1993-08-13 1999-08-17 Zymogenetics, Inc. Transglutaminase cross-linkable polypeptides and methods relating thereto
US20020107215A1 (en) * 2000-08-01 2002-08-08 Lifespan Biosciences, Inc. Tissue-associated proteins and their uses
US20030064074A1 (en) * 1999-11-12 2003-04-03 Chang Robert C. Recombinant gelatins in vaccines
US7060467B2 (en) * 2000-03-13 2006-06-13 Monsanto Technology Llc Recombinant proteins containing repeating units

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428014A (en) * 1993-08-13 1995-06-27 Zymogenetics, Inc. Transglutaminase cross-linkable polypeptides and methods relating thereto
US5939385A (en) * 1993-08-13 1999-08-17 Zymogenetics, Inc. Transglutaminase cross-linkable polypeptides and methods relating thereto
US20030064074A1 (en) * 1999-11-12 2003-04-03 Chang Robert C. Recombinant gelatins in vaccines
US7060467B2 (en) * 2000-03-13 2006-06-13 Monsanto Technology Llc Recombinant proteins containing repeating units
US20020107215A1 (en) * 2000-08-01 2002-08-08 Lifespan Biosciences, Inc. Tissue-associated proteins and their uses

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040001848A1 (en) * 2002-03-01 2004-01-01 Szu-Yi Chou Method of producing disease-specific antigens
US20040005654A1 (en) * 2002-03-01 2004-01-08 Szu-Yi Chou Method of producing polyvalent antigens
US7485438B2 (en) * 2002-03-01 2009-02-03 Szu-Yi Chou Method of producing polyvalent antigens

Also Published As

Publication number Publication date
US7396656B2 (en) 2008-07-08
US20070190616A1 (en) 2007-08-16

Similar Documents

Publication Publication Date Title
CA2064689C (en) Stabilized protein or peptide conjugates
ES2880336T3 (en) Methods and products for the synthesis of fusion proteins
EP1260582B1 (en) Conjugates of factor IX and a biocompatible polymer
KR102264478B1 (en) Recombinant Clostridium botulinum neurotoxins
CN102089328B (en) Streptokinase mutant and covalent modification thereof
JP2019506163A (en) Split intein with exceptional splicing activity
CN110582566A (en) Peptide ligase and uses thereof
CA2638862A1 (en) Pegylated mutated clostridium botulinum toxin
IL83878A (en) Soluble protein corresponding to tnf inhibitory protein its preparation and pharmaceutical compositions containing it
US7396656B2 (en) Method of cross-linking peptides
Nakajima et al. Chemical modification of earthworm fibrinolytic enzyme with human serum albumin fragment and characterization of the protease as a therapeutic enzyme
US20220403012A1 (en) Peptide-hinge-free flexible antibody-like molecule
US20090169553A1 (en) Novel Protein Fusion/Tag Technology
Liu et al. Preparation, characterization and in vitro bioactivity of N-terminally PEGylated staphylokinase dimers
JP6014194B2 (en) IGF-I poly (ethylene glycol) conjugate
CA2229540A1 (en) Epitope tagging system
US20080139400A1 (en) Molecular display on multimeric protein scaffolds
EP0753304A1 (en) Thrombolytic enzyme and method of obtaining same
Wang et al. A PEGylation technology of L-asparaginase with monomethoxy polyethylene glycol-propionaldehyde
JP2015534814A (en) Recombinant Clostridium botulinum neurotoxin
CN116261568A (en) Binding proteins for Complement Factor H (CFH)
Tomasselli et al. Recombinant Human Cytomegalovirus Protease with a C-Terminal (His) 6Extension: Purification, Autocatalytic Release of the Mature Enzyme, and Biochemical Characterization
Yokoigawa et al. Purification of high molecular weight urokinase by reverse-immunoadsorption
McCaman et al. Affinity binding of a vampire bat plasminogen activator to SEC resins
JPH0335782A (en) Soluble t11 protein which inhibits activation of t11-mediated t cell

Legal Events

Date Code Title Description
AS Assignment

Owner name: ACADEMIA SINICA, TAIWAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIN, HAN-JIA;CHEN, YEE-HSIUNG;REEL/FRAME:013936/0001;SIGNING DATES FROM 20030326 TO 20030327

STCB Information on status: application discontinuation

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