CN111269322B - Fusion protein of streptavidin and Gaussian luciferase and application thereof - Google Patents

Fusion protein of streptavidin and Gaussian luciferase and application thereof Download PDF

Info

Publication number
CN111269322B
CN111269322B CN201811473694.5A CN201811473694A CN111269322B CN 111269322 B CN111269322 B CN 111269322B CN 201811473694 A CN201811473694 A CN 201811473694A CN 111269322 B CN111269322 B CN 111269322B
Authority
CN
China
Prior art keywords
fusion protein
sequence
gly
seq
protein
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.)
Active
Application number
CN201811473694.5A
Other languages
Chinese (zh)
Other versions
CN111269322A (en
Inventor
李静
王佑富
郑越
董宇亮
章文蔚
徐崇钧
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.)
Qingdao Huada Zhizao Technology Co ltd
Original Assignee
Shenzhen Huada Zhizaojichuang Technology Co 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 Shenzhen Huada Zhizaojichuang Technology Co ltd filed Critical Shenzhen Huada Zhizaojichuang Technology Co ltd
Priority to CN201811473694.5A priority Critical patent/CN111269322B/en
Publication of CN111269322A publication Critical patent/CN111269322A/en
Application granted granted Critical
Publication of CN111269322B publication Critical patent/CN111269322B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/36Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Actinomyces; from Streptomyces (G)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/82Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving vitamins or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/61Fusion polypeptide containing an enzyme fusion for detection (lacZ, luciferase)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

The fusion protein comprises a streptavidin full-length protein and a gauss luciferase full-length protein or a continuous luminescence mutant, wherein the streptavidin full-length protein is connected with the gauss luciferase full-length protein or the continuous luminescence mutant through a flexible peptide. The fusion protein can conveniently and specifically identify biotin molecules, has the function of catalyzing substrate oxidation luminescence, and is convenient to detect. Compared with the coupling protein of the Gaussian luciferase and the streptavidin in the prior art, the fusion protein has the advantages of simple process and convenient production.

Description

Fusion protein of streptavidin and Gaussian luciferase and application thereof
Technical Field
The invention relates to the field of genetic engineering, in particular to a fusion protein of streptavidin and Gaussian luciferase and application thereof.
Background
Streptavidin (SA) was found by Chaiet in 1963 as a product secreted by Streptomyces avidinii during culture. One SA peptide chain has 159 amino acid residues and molecular weight of 16.5 kDa. The complete SA protein consists of 4 SA polypeptide chains, forming a tetrameric structure. Each polypeptide chain can specifically bind to one Biotin (Biotin) molecule, and thus 1 molecule of SA can bind to 4 molecules of Biotin. The interaction between the two exists in a non-covalent binding form, and the affinity reaches 10-15the/M, several orders of magnitude higher than the binding constant of antigen-antibody, is the most stable protein-small molecule interaction occurring in nature. The strong binding between the two results in biotin and SA being frequently used to develop labeling and detection tools. When the kit is used, biotin is usually labeled on a substrate to be detected, the biotin labeled on the substrate is combined by SA, then an antibody of the SA is combined with the SA, meanwhile, a luminescent group is connected to the antibody in advance, and the antibody can be excited to emit light, and finally, the substrate is detected by detecting the light emission.
Luciferase is a generic term for a class of enzymes that catalyze the oxidative luminescence of luciferin or fatty aldehydes in vivo. Is usually found in lower animals. The luciferase commonly used at present comprises firefly luciferase, renilla luciferase and gauss luciferase. Firefly luciferase requires ATP and Mg for luminescence2+Assisted by Renilla luciferase, although ATP and Mg-independent2+And the like, but the luminous intensity is weaker, and more sensitive detection is needed in application.
Gaussia luciferase (Gluc) well makes up the defects of firefly luciferase and renilla luciferase, and ATP and Mg are not required during luminescence2+The cofactors also have the luminous efficiency 100 times higher than that of renilla luciferase. The gauss luciferase is the luciferase secreted by a marine flexo-phalangeal animal, is the luciferase with the minimum molecular weight discovered at present, has a signal peptide, can be secreted to the outside of cells, and is convenient for activity detection. In thatUnder the condition of oxygen, the Gauss luciferase can spontaneously catalyze the oxidation luminescence of a substrate coelenterazine. The luminous intensity is the highest among the luciferases discovered at present, and the detection is easy. Due to the advantages, the Gauss luciferase is widely applied to the fields of living cell detection, protein-protein interaction, protein positioning, small interference RNA silencing technology, high-throughput drug screening and the like.
At present, two methods are mainly used for identifying and detecting biotin: one is the combination of SA and anti-SA antibodies, and the other is the combination of SA and Gauss luciferase. For the combination of SA and anti-SA antibodies, mainly SA is used for specifically recognizing biotin, then anti-SA antibody with a luminescent group is used for recognizing SA, and finally the biotin is detected by detecting the luminescence of the luminescent group on the antibody. The method is complicated and takes a long time. The coupling protein of SA and Gauss luciferase is prepared through chemical coupling modification of one biotin on Gauss luciferase molecule and further reaction of 1: 1, SA is added, and the SA is combined with biotin on Gaussian luciferase to form a coupled protein, and finally the combined ratio is 1: 1. The protein can be specifically combined with SA, and has the self-luminous ability, thereby being convenient for detection. However, the preparation process of the coupling protein is complicated, the process is complex, the production efficiency is low, the amplification of the yield is not facilitated, and only 1 molecule of luminescent protein exists on 1 molecule of SA, and the signal amplification function is not realized.
Disclosure of Invention
The invention provides a streptavidin and gauss luciferase fusion protein and application thereof, wherein the fusion protein can be obtained only by once expression and purification, the production process is simple, and the method is beneficial to large-scale production.
According to a first aspect, in one embodiment there is provided a streptavidin-gauss luciferase fusion protein comprising a full-length streptavidin protein and a full-length gauss luciferase protein or a persistent luminescent mutant, said full-length streptavidin protein being linked to said full-length gauss luciferase protein or persistent luminescent mutant by a flexible peptide.
In a preferred embodiment, the streptavidin full-length protein is as set forth in SEQ ID NO: 1 is shown.
In a preferred embodiment, the sequence of the full-length protein of the gauss luciferase is shown as SEQ ID NO: 2, respectively.
In a preferred embodiment, the above-mentioned continuous light-emitting mutant has a sequence shown in SEQ ID NO: 3, respectively.
In a preferred embodiment, the flexible peptide sequence is as set forth in SEQ ID NO: 4, respectively.
In a preferred embodiment, the above fusion protein further comprises: a signal peptide for directing secretion of the fusion protein from the inside to the outside of the expression cell.
In a preferred embodiment, the signal peptide is located at the N-terminus of the fusion protein.
In a preferred embodiment, the signal peptide sequence is as set forth in SEQ ID NO: 5, respectively.
In a preferred embodiment, the above fusion protein further comprises: a fusion tag for affinity purification of the above fusion protein.
In a preferred embodiment, the fusion tag is a 6 × histidine tag.
In a preferred embodiment, the sequence of the fusion protein is shown in SEQ ID NO: 6 or SEQ ID NO: shown at 7.
According to a second aspect, in one embodiment there is provided an isolated nucleic acid encoding the fusion protein of the first aspect.
In a preferred embodiment, the isolated nucleic acid comprises a coding sequence for a full-length streptavidin protein and a coding sequence for a full-length gauss luciferase protein or a coding sequence for a persistent luminescent mutant, wherein the coding sequence for the full-length streptavidin protein is linked to the coding sequence for the full-length gauss luciferase protein or the coding sequence for the persistent luminescent mutant by a linker sequence.
In a preferred embodiment, the coding sequence of the streptavidin full-length protein is shown in SEQ ID NO: shown in fig. 8.
In a preferred embodiment, the coding sequence of the full-length protein of the gauss luciferase is shown as SEQ ID NO: shown at 9.
In a preferred embodiment, the coding sequence of the above-mentioned continuous light-emitting mutant is shown in SEQ ID NO: shown at 10.
In a preferred embodiment, the linker sequence is as set forth in SEQ ID NO: shown at 11.
In a preferred embodiment, the above isolated nucleic acid further comprises: a signal peptide coding sequence which codes for a signal peptide for directing secretion of the above fusion protein from the inside to the outside of the expression cell.
In a preferred embodiment, the signal peptide coding sequence is located 5' to the isolated nucleic acid.
In a preferred embodiment, the signal peptide coding sequence is as set forth in SEQ ID NO: shown at 12.
In a preferred embodiment, the above isolated nucleic acid further comprises: a fusion tag coding sequence which codes for a fusion tag for affinity purification of the above fusion protein.
In a preferred embodiment, the fusion tag coding sequence is a sequence encoding a 6 × histidine tag.
In a preferred embodiment, the isolated nucleic acid sequence is as set forth in SEQ ID NO: 13 or SEQ ID NO: as shown at 14.
According to a third aspect, there is provided in one embodiment an expression vector comprising a nucleic acid sequence encoding the fusion protein of the first aspect, and a vector backbone sequence.
According to a fourth aspect, there is provided in one embodiment a recombinant host cell comprising within it an expression vector of the third aspect.
According to a fifth aspect, there is provided in one embodiment the use of the fusion protein of the first aspect, the isolated nucleic acid of the second aspect, the expression vector of the third aspect or the recombinant host cell of the fourth aspect in the detection of a biotin molecule.
According to the invention, SA and Gaussian luciferase genes are subjected to fusion expression by a genetic engineering technology to form a fusion protein, and the fusion protein can conveniently and specifically identify biotin molecules and has the function of catalyzing substrate oxidation luminescence. The fusion protein can be obtained only by once expression and purification, the production process is simple, the scale production is facilitated, and compared with the coupling protein of the Gaussian luciferase and the streptavidin in the prior art, the fusion protein has the advantages of simple process and convenience in production. The fusion protein has wide application prospect in the fields of DNA hybridization, immunodetection, biochemical diagnosis and the like.
Drawings
FIG. 1 is a schematic diagram showing the construction of a fusion vector according to an embodiment of the present invention, wherein SA represents a streptavidin full-length protein coding sequence, Gluc represents a Gaussian luciferase full-length protein coding sequence or a continuous light-emitting mutant coding sequence, Linker represents a Linker sequence, Signal peptide represents a Signal peptide coding sequence, and Overlap PCR represents bridge PCR.
FIG. 2 is a SDS-PAGE detection result of the fusion protein in the example of the present invention, wherein lane 1: SA-Gluc fusion protein, lane 2: marker.
FIG. 3 is a graph showing the results of bioluminescence assay of a wild-type SA-Gluc fusion protein and a mutant SA-G2L fusion protein in examples of the present invention, wherein SA-Gluc represents the wild-type fusion protein, SA-G2L represents the mutant fusion protein, Gluc represents Gauss luciferase, abscissa represents Time (Time), and ordinate represents luminescence intensity (CL values).
FIG. 4 is a graph showing the results of measuring the binding force of the wild-type SA-Gluc fusion protein and the mutant SA-G2L fusion protein to biotin by the Elisa method in examples of the present invention, in which SA-Gluc represents the wild-type fusion protein, SA-G2L represents the mutant fusion protein, the abscissa represents the Concentration (Concentration) value, and the ordinate represents the OD450nm absorbance value.
Detailed Description
The present invention will be described in further detail with reference to the following detailed description and accompanying drawings. In the following description, numerous specific details are set forth in order to provide a thorough understanding of the present invention. However, those skilled in the art will readily recognize that some of the features may be omitted in different instances or may be replaced by other materials, methods.
Furthermore, the features, operations, or characteristics described in the specification may be combined in any suitable manner to form various embodiments. Also, the various steps or actions in the method descriptions may be transposed or transposed in order, as will be apparent to one of ordinary skill in the art. Thus, the various sequences in the specification and drawings are for the purpose of describing certain embodiments only and are not intended to imply a required sequence unless otherwise indicated where such sequence must be followed.
The invention aims to provide a fusion protein which can specifically bind Biotin and catalyze substrate coelenterazine to emit light, so as to achieve the purpose of conveniently and rapidly detecting any molecule with a Biotin (Biotin) label.
The invention belongs to the field of genetic engineering, and particularly relates to preparation and application of a fusion protein (SA-Gluc) of streptavidin and Gaussian luciferase. The invention uses gene engineering technology to connect the Gauss luciferase full-length protein (or continuous luminous mutant G2L) and the streptavidin full-length protein through flexible peptide (linker) to obtain the fusion protein. The C end of the fusion protein is connected with a polyhistidine tag, expression and nickel column purification are carried out through CHO cells, and the obtained fusion protein can specifically bind to biotin on one hand and oxidize a substrate coelenterazine to emit light on the other hand. The self-luminescence can be detected by instruments such as an enzyme-labeling instrument and the like, can be observed by naked eyes under certain conditions, and can conveniently detect any molecule with a biotin label without the participation of any antibody and other luminescent molecules.
A coding Gaussian luciferase full-length gene (or a continuous luminescence mutant G2L gene) and a streptavidin full-length gene are constructed into a eukaryotic recombinant expression vector by utilizing a PCR and double enzyme digestion technology, a signal peptide is added at the N end of a target gene, a 6 × histidine (6 × His) tag is added at the C end, and a coding gene of a flexible peptide is added between the Gaussian luciferase gene and the streptavidin gene. The constructed expression vector plasmid was transformed into CHO cells by the liposome method for transient expression. After the protein is expressed in the cell, the protein is secreted into the cell culture fluid under the action of the signal peptide, and the signal peptide is cut off in the secretion process. After the fermentation was completed, the cell culture fluid was collected by centrifugation. And purifying the collected culture solution by a Ni column to obtain the fusion protein. The fusion protein has the specific binding capacity with biotin and the capacity of catalyzing the oxidization and luminescence of the coelenterazine as a substrate.
The invention provides a fusion protein of streptavidin and gauss luciferase, which comprises a streptavidin full-length protein and gauss luciferase full-length protein or a continuous luminescence mutant, wherein the streptavidin full-length protein is connected with the gauss luciferase full-length protein or the continuous luminescence mutant through a flexible peptide.
In the fusion protein of the present invention, the connection mode (i.e., the sequence of the streptavidin full-length protein and the gauss luciferase full-length protein or the continuous luminescent mutant) of the streptavidin full-length protein and the gauss luciferase full-length protein or the continuous luminescent mutant can be adjusted, for example, the streptavidin full-length protein is at the N-terminal of the fusion protein, and the gauss luciferase full-length protein or the continuous luminescent mutant is at the C-terminal of the fusion protein; or the Gaussian luciferase full-length protein or the continuous luminescence mutant is arranged at the N end of the fusion protein, and the streptavidin full-length protein is arranged at the C end of the fusion protein. In a preferred embodiment, the streptavidin full-length protein is at the N-terminus of the fusion protein and the gauss luciferase full-length protein or the persistent light mutant is at the C-terminus of the fusion protein.
In the fusion protein of the present invention, the persistent luminescent mutant refers to a persistent luminescent mutant of the full-length protein of the gauss luciferase, and can refer to any mutant which has one or more amino acid mutations relative to the full-length protein of the wild-type gauss luciferase and still maintains the luminescent properties of the full-length protein of the gauss luciferase. In a preferred embodiment, the persistent luminescent mutant is persistent luminescent mutant G2L, described in more detail below.
In a preferred embodiment, the full length streptavidin protein is as follows SEQ ID NO: 1, and the following components:
MAEAGITGTWYNQLGSTFIVTAGADGALTGTYESAVGNAESRYVLTGRYDSAPATDGSGTALGWTVAWKNNYRNAHSATTWSGQYVGGAEARINTQWLLTSGTTEANAWKSTLVGHDTFTKVKPSAAS(SEQ ID NO:1)。
in a preferred embodiment, the full-length protein sequence of the gauss luciferase is as follows SEQ ID NO: 2, as shown in the figure:
KPTENNEDFNIVAVASNFATTDLDADRGKLPGKKLPLEVLKEMEANARKAGCTRGCLICLSHIKCTPKMKKFIPGRCHTYEGDKESAQGGIGEAIVDIPEIPGFKDLEPMEQFIAQVDLCVDCTTGCLKGLANVQCSDLLKKWLPQRCATFASKIQGQVDKIKGAGGD(SEQ ID NO:2)。
in a preferred embodiment, the persistent luminescent mutant is a G2L mutant having the sequence set forth in SEQ ID NO: 3, showing:
KPTENNEDFNIVAVASNFATTDLDADRGKLPGKKLPLEVLKELEANARKAGCTRGCLICLSHIKCTPKMKKFIPGRCHTYEGDKESAQGGIGEAIVDIPEIPGFKDLEPLEQFIAQVDLCVDCTTGCLKGLANVQCSDLLKKWLPQRCATFASKIQGQVDKIKGAGGD (SEQ ID NO: 3). Among them, one with underlining lines "L"refers to the mutated amino acid positions relative to the full-length protein sequence of the wild-type gauss luciferase, namely, the mutations M43L and M110L.
It is noted that the fusion protein of the present invention includes, in addition to the fusion protein using SEQ ID NO: 1 and SEQ ID NO: 2, or SEQ ID NO: 1 and SEQ ID NO: 3, and also includes a sequence form having the same function although the sequence has a certain difference. For example, for the full length streptavidin protein, the sequence can be the same as that of SEQ ID NO: 1, but also has the corresponding streptavidin full-length protein affinity activity, as shown in SEQ ID NO: 1 sequence with several amino acids added or reduced at both ends but with the same affinity activity; for the full-length protein sequence of the gauss luciferase, the sequence can be the same as that of SEQ ID NO: 2, but also has the activity of the full-length protein sequence of the gauss luciferase, as shown in SEQ ID NO: 2, the sequence with several amino acids increased or decreased at both ends of the sequence but the activity of the full-length protein sequence of the same Gauss luciferase is maintained; for the persistent light mutant sequence, it can be a nucleotide sequence similar to that of SEQ ID NO: 3, but also has the sequence activity of a persistent light-emitting mutant, as shown in SEQ ID NO: 3 sequence with several amino acids added or reduced at both ends but maintaining the activity of the same continuous light-emitting mutant sequence.
In the fusion protein of the present invention, the flexible peptide having a linking function may be selected from the following sequences:
GGGGSGGGGSGGGGS(SEQ ID NO:4);
GGGGGS(SEQ ID NO:15);
GQGQGQGQGQG(SEQ ID NO:16);
GSTSGSGKSSEKGKG(SEQ ID NO:17);
VPGVGVPGVG(SEQ ID NO:18);
SAPGTPSR(SEQ ID NO:19);
EGKSSGSGSESKEF(SEQ ID NO:20);
GSGGSG(SEQ ID NO:21);
GSGGSGGG(SEQ ID NO:22)。
in a preferred embodiment, the flexible peptide sequence is as set forth in SEQ ID NO: 4, and (2) is as follows:
GGGGSGGGGSGGGGS(SEQ ID NO:4)。
in a preferred embodiment, the fusion protein further comprises: a signal peptide for directing secretion of the fusion protein from the expressing cell to the outside.
The signal peptide may be located at the N-terminus or C-terminus of the fusion protein of the present invention, and in a preferred embodiment, the signal peptide is located at the N-terminus of the above fusion protein without affecting the affinity activity and the luminescence activity in the fusion protein of the present invention.
In a preferred embodiment, the signal peptide sequence is as set forth in SEQ ID NO: and 5, as follows:
MGVKVLFALICIAVAEA(SEQ ID NO:5)。
in the present invention, methods useful for purifying fusion proteins include, but are not limited to: nickel column purification, ion exchange chromatography (Q column and P column), hydrophobic chromatography, heparin column chromatography, ammonium sulfate precipitation, etc.
In a preferred embodiment, the fusion protein of the invention further comprises: a fusion tag for affinity purification of the fusion protein. Specifically, the fusion tag is affinity-bound to a purification apparatus (e.g., a purification column) used in various purification methods, and then the purified fusion protein is obtained by an appropriate method. The fusion tag may be located at the N-terminus or C-terminus, preferably at the C-terminus, of the fusion protein of the invention without affecting the affinity activity and the luminescence activity in the fusion protein of the invention.
The fusion tag can be a 6 × histidine tag (6 × His tag) or other fusion tags such as GST, MBP, SUMO and the like, and an affinity purification method corresponding to the fusion tag is selected according to the difference of the fusion tag. In a preferred embodiment, the fusion tag is a 6 × histidine tag.
In a most preferred embodiment, the fusion protein of the invention has the following sequence SEQ ID NO: 6 or SEQ ID NO: 7, and:
MGVKVLFALICIAVAEAMAEAGITGTWYNQLGSTFIVTAGADGALTGTYESAVGNAESRYVLTGRYDSAPATDGSGTALGWTVAWKNNYRNAHSATTWSGQYVGGAEARINTQWLLTSGTTEANAWKSTLVGHDTFTKVKPSAASGGGGSGGGGSGGGGSKPTENNEDFNIVAVASNFATTDLDADRGKLPGKKLPLEVLKEMEANARKAGCTRGCLICLSHIKCTPKMKKFIPGRCHTYEGDKESAQGGIGEAIVDIPEIPGFKDLEPMEQFIAQVDLCVDCTTGCLKGLANVQCSDLLKKWLPQRCATFASKIQGQVDKIKGAGGDHHHHHH(SEQ ID NO:6);
MGVKVLFALICIAVAEAMAEAGITGTWYNQLGSTFIVTAGADGALTGTYESAVGNAESRYVLTGRYDSAPATDGSGTALGWTVAWKNNYRNAHSATTWSGQYVGGAEARINTQWLLTSGTTEANAWKSTLVGHDTFTKVKPSAASGGGGSGGGGSGGGGSKPTENNEDFNIVAVASNFATTDLDADRGKLPGKKLPLEVLKELEANARKAGCTRGCLICLSHIKCTPKMKKFIPGRCHTYEGDKESAQGGIGEAIVDIPEIPGFKDLEPLEQFIAQVDLCVDCTTGCLKGLANVQCSDLLKKWLPQRCATFASKIQGQVDKIKGAGGDHHHHHH(SEQ ID NO:7)。
wherein the underlined sequence (MGVKVLFALICIAVAEA) Represents a signal peptide; underlined sequence (GGGGSGGGGSGGGGS) Indicates a flexible peptide; underlined sequence (HHHHHH) Represents the 6 × histidine tag sequence.
One embodiment of the invention provides an isolated nucleic acid encoding the fusion protein of the first aspect.
In a preferred embodiment, the isolated nucleic acid comprises a coding sequence for a full-length streptavidin protein and a coding sequence for a full-length gauss luciferase protein or a coding sequence for a persistent luminescent mutant, the coding sequence for the full-length streptavidin protein being linked to the coding sequence for the full-length gauss luciferase protein or the coding sequence for the persistent luminescent mutant by a linker sequence.
It is to be noted that, due to the degeneracy of codons, nucleotide sequences encoding the same polypeptide or protein may differ, and therefore, an isolated nucleic acid of the present invention may be any nucleic acid capable of encoding a fusion protein of the present invention. The sequence is not particularly limited.
In a preferred embodiment, the full-length streptavidin protein coding sequence is as follows SEQ ID NO: 8, showing:
ATGGCTGAGGCTGGAATCACCGGAACATGGTACAACCAGCTGGGCAGCACCTTTATCGTGACAGCTGGAGCTGATGGCGCCCTGACCGGCACATATGAGTCCGCTGTGGGCAATGCTGAGAGCAGATACGTGCTGACCGGCCGGTATGATTCTGCTCCAGCTACCGACGGATCCGGAACAGCTCTGGGATGGACCGTGGCCTGGAAGAACAATTACAGGAACGCTCACTCTGCCACCACATGGTCCGGACAGTATGTGGGAGGAGCTGAGGCTCGGATCAACACACAGTGGCTGCTGACCAGCGGCACCACAGAGGCTAATGCCTGGAAGTCTACCCTGGTGGGCCATGATACCTTCACAAAGGTGAAGCCCTCCGCCGCTAGC(SEQ ID NO:8)。
in a preferred embodiment, the full-length protein coding sequence for gauss luciferase is as follows SEQ ID NO: 9 is as follows:
AAGCCAACAGAGAACAATGAGGATTTCAACATCGTGGCTGTGGCCAGCAATTTTGCTACCACAGACCTGGATGCCGACAGAGGCAAGCTGCCAGGCAAGAAGCTGCCCCTGGAGGTGCTGAAGGAGATGGAGGCTAACGCTAGGAAGGCTGGATGTACCAGGGGATGCCTGATCTGTCTGTCTCACATCAAGTGCACACCTAAGATGAAGAAGTTCATCCCAGGCCGCTGTCATACCTACGAGGGCGATAAGGAGTCCGCTCAGGGAGGAATCGGAGAGGCCATCGTGGATATCCCCGAGATCCCTGGCTTCAAGGACCTGGAGCCCATGGAGCAGTTTATCGCTCAGGTGGATCTGTGCGTGGACTGTACCACAGGCTGCCTGAAGGGCCTGGCCAATGTGCAGTGTTCCGACCTGCTGAAGAAGTGGCTGCCTCAGAGGTGCGCTACCTTTGCCAGCAAGATCCAGGGCCAGGTGGATAAGATCAAGGGAGCTGGAGGCGAC(SEQ ID NO:9)。
in a preferred embodiment, the persistent light mutant coding sequence is as set forth in SEQ ID NO: 10, and:
AAGCCAACAGAGAACAATGAGGATTTCAACATCGTGGCTGTGGCCAGCAATTTTGCTACCACAGACCTGGATGCCGACAGAGGCAAGCTGCCAGGCAAGAAGCTGCCCCTGGAGGTGCTGAAGGAGCTCGAGGCTAACGCTAGGAAGGCTGGATGTACCAGGGGATGCCTGATCTGTCTGTCTCACATCAAGTGCACACCTAAGATGAAGAAGTTCATCCCAGGCCGCTGTCATACCTACGAGGGCGATAAGGAGTCCGCTCAGGGAGGAATCGGAGAGGCCATCGTGGATATCCCCGAGATCCCTGGCTTCAAGGACCTGGAGCCCCTCGAGCAGTTTATCGCTCAGGTGGATCTGTGCGTGGACTGTACCACAGGCTGCCTGAAGGGCCTGGCCAATGTGCAGTGTTCCGACCTGCTGAAGAAGTGGCTGCCTCAGAGGTGCGCTACCTTTGCCAGCAAGATCCAGGGCCAGGTGGATAAGATCAAGGGAGCTGGAGGCGAC (SEQ ID NO: 10). Wherein the underlined codons denote relative to Gaussian fluorescenceMutant bases of the coding sequence of the full-length protein of the luciferase.
In a preferred embodiment, the linker sequence is as set forth in SEQ ID NO: 11, and:
GGAGGAGGAGGATCTGGAGGAGGAGGATCCGGAGGAGGAGGATCT(SEQ ID NO:11)。
in preferred embodiments, the isolated nucleic acid further comprises: a signal peptide coding sequence which encodes a signal peptide for directing secretion of the fusion protein from the expressing cell to the outside.
Since the signal peptide in the fusion protein of the present invention may be located at the N-terminus or C-terminus of the fusion protein, the signal peptide coding sequence may be located at the 5 'end or 3' end of the isolated nucleic acid. The signal peptide is located at the N-terminus of the above fusion protein without affecting the affinity and luminescence activity in the fusion protein of the present invention, and thus in a preferred embodiment, the signal peptide coding sequence is located at the 5' end of the isolated nucleic acid.
In a preferred embodiment, the signal peptide coding sequence is as set forth in SEQ ID NO: 12, and:
ATGGGCGTGAAGGTGCTGTTCGCCCTGATCTGCATCGCCGTGGCTGAGGCC(SEQ ID NO:12)。
in a preferred embodiment, the isolated nucleic acid further comprises a fusion tag coding sequence encoding a fusion tag for affinity purification of the above-described fusion protein.
In a preferred embodiment, the fusion tag coding sequence is a sequence encoding a 6 × histidine tag.
The fusion tag coding sequence can be located at the 3 ' end or 5 ' end of the isolated nucleic acid, and in preferred embodiments, the fusion tag coding sequence is located at the 3 ' end of the isolated nucleic acid and does not affect the expression of the affinity and light-emitting active portions of the fusion protein.
In a most preferred embodiment, the isolated nucleic acid sequence of the invention is as set forth in SEQ ID NO: 13 or SEQ ID NO: 14, in the following:
ATGGGCGTGAAGGTGCTGTTCGCCCTGATCTGCATCGCCGTGGCTGAGGCCATGGCTGAGGCTGGAATCACCGGAACATGGTACAACCAGCTGGGCAGCACCTTTATCGTGACAGCTGGAGCTGATGGCGCCCTGACCGGCACATATGAGTCCGCTGTGGGCAATGCTGAGAGCAGATACGTGCTGACCGGCCGGTATGATTCTGCTCCAGCTACCGACGGATCCGGAACAGCTCTGGGATGGACCGTGGCCTGGAAGAACAATTACAGGAACGCTCACTCTGCCACCACATGGTCCGGACAGTATGTGGGAGGAGCTGAGGCTCGGATCAACACACAGTGGCTGCTGACCAGCGGCACCACAGAGGCTAATGCCTGGAAGTCTACCCTGGTGGGCCATGATACCTTCACAAAGGTGAAGCCCTCCGCCGCTAGCGGAGGAGGAGGAT CTGGAGGAGGAGGATCCGGAGGAGGAGGATCTAAGCCAACAGAGAACAATGAGGATTTCAACATCGTGGCTGTGGCCAGCAATTTTGCTACCACAGACCTGGATGCCGACAGAGGCAAGCTGCCAGGCAAGAAGCTGCCCCTGGAGGTGCTGAAGGAGATGGAGGCTAACGCTAGGAAGGCTGGATGTACCAGGGGATGCCTGATCTGTCTGTCTCACATCAAGTGCACACCTAAGATGAAGAAGTTCATCCCAGGCCGCTGTCATACCTACGAGGGCGATAAGGAGTCCGCTCAGGGAGGAATCGGAGAGGCCATCGTGGATATCCCCGAGATCCCTGGCTTCAAGGACCTGGAGCCCATGGAGCAGTTTATCGCTCAGGTGGATCTGTGCGTGGACTGTACCACAGGCTGCCTGAAGGGCCTGGCCAATGTGCAGTGTTCCGACCTGCTGAAGAAGTGGCTGCCTCAGAGGTGCGCTACCTTTGCCAGCAAGATCCAGGGCCAGGTGGATAAGATCAAGGGAGCTGGAGGCGACCACCATCACCATCACCAT(SEQ ID NO:13);
ATGGGCGTGAAGGTGCTGTTCGCCCTGATCTGCATCGCCGTGGCTGAGGCCATGGCTGAGGCTGGAATCACCGGAACATGGTACAACCAGCTGGGCAGCACCTTTATCGTGACAGCTGGAGCTGATGGCGCCCTGACCGGCACATATGAGTCCGCTGTGGGCAATGCTGAGAGCAGATACGTGCTGACCGGCCGGTATGATTCTGCTCCAGCTACCGACGGATCCGGAACAGCTCTGGGATGGACCGTGGCCTGGAAGAACAATTACAGGAACGCTCACTCTGCCACCACATGGTCCGGACAGTATGTGGGAGGAGCTGAGGCTCGGATCAACACACAGTGGCTGCTGACCAGCGGCACCACAGAGGCTAATGCCTGGAAGTCTACCCTGGTGGGCCATGATACCTTCACAAAGGTGAAGCCCTCCGCCGCTAGCGGAGGAGGAGGAT CTGGAGGAGGAGGATCCGGAGGAGGAGGATCTAAGCCAACAGAGAACAATGAGGATTTCAACATCGTGGCTGTGGCCAGCAATTTTGCTACCACAGACCTGGATGCCGACAGAGGCAAGCTGCCAGGCAAGAAGCTGCCCCTGGAGGTGCTGAAGGAGCTCGAGGCTAACGCTAGGAAGGCTGGATGTACCAGGGGATGCCTGATCTGTCTGTCTCACATCAAGTGCACACCTAAGATGAAGAAGTTCATCCCAGGCCGCTGTCATACCTACGAGGGCGATAAGGAGTCCGCTCAGGGAGGAATCGGAGAGGCCATCGTGGATATCCCCGAGATCCCTGGCTTCAAGGACCTGGAGCCCCTCGAGCAGTTTATCGCTCAGGTGGATCTGTGCGTGGACTGTACCACAGGCTGCCTGAAGGGCCTGGCCAATGTGCAGTGTTCCGACCTGCTGAAGAAGTGGCTGCCTCAGAGGTGCGCTACCTTTGCCAGCAAGATCCAGGGCCAGGTGGATAAGATCAAGGGAGCTGGAGGCGACCACCATCACCATCACCAT(SEQ ID NO:14)。
wherein, SEQ ID NO: 13 or SEQ ID NO: among the 14 sequences, underlined sequences from the 5 'end to the 3' end are a signal peptide coding sequence, a flexible peptide coding sequence (linker sequence), and a sequence for coding a 6 × histidine tag, respectively.
One embodiment of the invention provides an expression vector comprising a nucleic acid sequence encoding a fusion protein of the invention, and a vector backbone sequence. The vector backbone sequence may be any suitable expression vector sequence, for example, in one embodiment, the vector backbone sequence is pcDNA3.1 (+).
One embodiment of the present invention provides a recombinant host cell comprising an expression vector of the present invention. The host cell may be any host cell capable of expressing the fusion protein of the invention, e.g., in one embodiment, a CHO cell.
The invention also provides the use of the fusion protein, the isolated nucleic acid, the expression vector or the recombinant host cell in the detection of a biotin molecule.
The invention is useful for detecting biotin molecules, which can be in free form, independently, or any molecule bearing a biotin label, such as a biotin-labeled protein. In some embodiments, the biotin is labeled on a substrate to be detected, and the substrate can be detected by detecting a biotin molecule on the substrate. The invention has wide application in the fields of DNA hybridization, immunodetection, biochemical diagnosis and the like.
The fusion protein can conveniently and specifically identify biotin molecules, has the function of catalyzing substrate oxidation luminescence, and is convenient to detect. Compared with the coupling protein of the Gaussian luciferase and the streptavidin in the prior art, the fusion protein has the advantages of simple process and convenient production.
The technical solutions of the present invention are described in detail below by way of examples, and it should be understood that the examples are only illustrative and should not be construed as limiting the scope of the present invention.
Example 1: construction of fusion expression vectors
As shown in FIG. 1, synthetic primers were designed using SA gene and Gauss luciferase gene as templates, and PCR amplification was performed to obtain individual gene amplification products of SA and Gauss luciferase, respectively. Based on the PCR product, a second round of bypass PCR is carried out to obtain the SA and Gaussian luciferase fusion gene amplification product (the sequence is shown as SEQ ID NO: 13). And (3) carrying out electrophoresis detection on the PCR amplification product by using 1% agarose gel, and recovering the product by using a gel recovery kit. The recovered product and the vector plasmid pcDNA3.1(+) are respectively subjected to double enzyme digestion reaction by EcoR I and Hind III, electrophoresis detection is carried out by 1% agarose gel after the reaction is finished, and the product is recovered by a gel recovery kit. The PCR-digested product and the plasmid-digested product were mixed at a molar ratio of 3:1 and ligated with T4 DNA ligase. The ligation products were transformed into TOP10 competent, plated on LB plates containing 100 ng/. mu.L ampicillin. The next day, single clones were picked from the plates, plasmids were extracted after extensive culture, and sequencing was performed to ensure correct sequence after PCR was identified as positive.
Mutant G2L fusion expression vector on the basis of the successfully prepared wild-type fusion protein expression vector, plasmid PCR (wherein, SA and continuous luminescent mutant fusion gene sequences are shown in SEQ ID NO: 14) is carried out by designing a primer containing a mutant gene, and then the PCR product is treated by Dpn I enzyme and reacted for 2h at 37 ℃. The product was transformed into TOP10 competent cells and plated on LB plates containing 100 ng/. mu.L ampicillin. The next day, single clones were picked from the plates, plasmids were extracted after extensive culture, and sequencing ensured that the mutation sites were correct.
Example 2: transfection, expression and purification of fusion expression vectors
25 mu g of the constructed pcDNA3.1-SA-Gluc fusion expression plasmid was mixed with 1ml of cell culture medium. Another 920. mu.L of cell culture medium was mixed with 80. mu.L of lipo2000 transfection reagent (invitrogen). The diluted expression vector and the diluted transfection reagent were mixed well and left at room temperature for 20 minutes. 25ml of 2X 10 solution was prepared6For the mixed cells, the mixed pcDNA3.1-SA-Gluc fusion expression plasmid and transfection reagent mixture was added to CHO cells and shaken gently. 37 ℃ and 8% CO2Cultured under the conditions for 3 days.
The cell culture fluid was collected, centrifuged at 6000rpm for 10min, and the supernatant was collected. The supernatant was filtered and transferred to a nickel column and 10ml of PBS was equilibrated. Then 10ml was washed with a buffer (20mM Tris, pH 8.0, 150mM NaCl, 20mM imidazole) to remove the hetero-protein, and finally eluted with a buffer (20mM Tris, pH 8.0, 150mM NaCl, 500mM imidazole). The eluted proteins were detected by 12% SDS-PAGE, and the results of the SDS-PAGE detection of the SA-Gluc fusion protein are shown in FIG. 2. Showing that the SA-Gluc fusion protein is successfully expressed.
The pcDNA3.1-SA-G2L fusion expression plasmid was constructed as described above.
Example 3: fusion protein luciferase bioluminescent assay
The SA-Gluc fusion protein and the SA-G2L fusion protein were each diluted to 3nM with a substrate diluent (50mM Tris, pH 8.0, 100mM NaCl) and 10. mu.L was added to a 96-well plate. Then, 90. mu.L of coelenterazine diluted to 10. mu.M with the same solution was added, and the luminescence intensity was read by a luminescence module using a microplate reader, and the result is shown in FIG. 3, in which the luminescence intensity of the wild-type SA-Gluc fusion protein was equivalent to that of Gluc protein of the same concentration and was slightly weaker. The mutant fusion protein SA-G2L has the luminous intensity equivalent to that of Gluc, but obviously shows the continuous luminous characteristic, and the luminescence is not obviously reduced after 30 s.
Example 4: elisa test experiment for binding capacity of fusion protein and biotin
Preparing a solution:
PBS: 138mM NaCl, 2.6mM KCl (in 10mM potassium phosphate solution, pH 7.4);
PBST: PBS plus 0.05% Tween 20;
sealing liquid: PBS added with 40 mug/mL BSA;
diluting the solution: PBS was added with 0.15. mu.g/mL BSA.
(1) A96-well plate was used, 100. mu.L of biotin-labeled BSA (10 ng/. mu.g) was added to each well, and the plate was coated at 37 ℃ for 1 hour.
(2) The coating solution was decanted off and washed 3 times with 100. mu.L PBST for 5 minutes each.
(3) Add 100. mu.L of blocking solution to each well and block for 1 hour at 37 ℃.
(4) The blocking solution was decanted off and washed 3 times with 100. mu.L of PBST for 5 minutes each.
(5) The SA-Gluc fusion protein and the SA-G2L fusion protein were diluted to 500nM with the dilutions, and then diluted to 100nM,20nM,4nM,0.8nM,0.16nM and 0.032nM with 5-fold gradients, and the dilutions of each gradient were added to a 96-well plate in sequence, with 3 replicate wells per gradient. The diluent without the fusion protein was used as a negative control. Binding was carried out at 37 ℃ for 1 hour.
(6) The plate was decanted and washed 3 times with 100. mu.L of PBST for 5 minutes each.
(7) The horseradish peroxidase-labeled SA antibody was diluted with a diluent and added to a 96-well plate at 100. mu.L per well, and bound at 37 ℃ for 1 hour.
(8) The plate was decanted and washed 3 times with 100. mu.L of PBST for 5 minutes each. The cells were washed 3 times with 100. mu.L PBS by gentle shaking for 5 minutes each.
(9) The luminescence was developed using a DMB luminescence kit (Biotech). The kit is operated according to the kit instructions, and after color development, the absorbance value of 450nM is read by a microplate reader.
(10) And (4) counting the light absorption values of all concentrations, taking the average of 3 repeated holes, making a corresponding curve of the concentrations and the light absorption values, and calculating the binding force constant.
The results are shown in FIG. 4, which shows that the binding force of the wild-type SA-Gluc fusion protein and the fusion protein of the persistent luminescent mutant SA-G2L to biotin is 10-10An order of magnitude.
The present invention has been described in terms of specific examples, which are provided to aid understanding of the invention and are not intended to be limiting. For a person skilled in the art to which the invention pertains, several simple deductions, modifications or substitutions may be made according to the idea of the invention.
SEQUENCE LISTING
<110> Shenzhen Huashengshengsciences institute
Fusion protein of <120> streptavidin and Gaussian luciferase and application thereof
<130> 18I27210
<160> 22
<170> PatentIn version 3.3
<210> 1
<211> 128
<212> PRT
<213> Artificial sequence
<400> 1
Met Ala Glu Ala Gly Ile Thr Gly Thr Trp Tyr Asn Gln Leu Gly Ser
1 5 10 15
Thr Phe Ile Val Thr Ala Gly Ala Asp Gly Ala Leu Thr Gly Thr Tyr
20 25 30
Glu Ser Ala Val Gly Asn Ala Glu Ser Arg Tyr Val Leu Thr Gly Arg
35 40 45
Tyr Asp Ser Ala Pro Ala Thr Asp Gly Ser Gly Thr Ala Leu Gly Trp
50 55 60
Thr Val Ala Trp Lys Asn Asn Tyr Arg Asn Ala His Ser Ala Thr Thr
65 70 75 80
Trp Ser Gly Gln Tyr Val Gly Gly Ala Glu Ala Arg Ile Asn Thr Gln
85 90 95
Trp Leu Leu Thr Ser Gly Thr Thr Glu Ala Asn Ala Trp Lys Ser Thr
100 105 110
Leu Val Gly His Asp Thr Phe Thr Lys Val Lys Pro Ser Ala Ala Ser
115 120 125
<210> 2
<211> 168
<212> PRT
<213> Artificial sequence
<400> 2
Lys Pro Thr Glu Asn Asn Glu Asp Phe Asn Ile Val Ala Val Ala Ser
1 5 10 15
Asn Phe Ala Thr Thr Asp Leu Asp Ala Asp Arg Gly Lys Leu Pro Gly
20 25 30
Lys Lys Leu Pro Leu Glu Val Leu Lys Glu Met Glu Ala Asn Ala Arg
35 40 45
Lys Ala Gly Cys Thr Arg Gly Cys Leu Ile Cys Leu Ser His Ile Lys
50 55 60
Cys Thr Pro Lys Met Lys Lys Phe Ile Pro Gly Arg Cys His Thr Tyr
65 70 75 80
Glu Gly Asp Lys Glu Ser Ala Gln Gly Gly Ile Gly Glu Ala Ile Val
85 90 95
Asp Ile Pro Glu Ile Pro Gly Phe Lys Asp Leu Glu Pro Met Glu Gln
100 105 110
Phe Ile Ala Gln Val Asp Leu Cys Val Asp Cys Thr Thr Gly Cys Leu
115 120 125
Lys Gly Leu Ala Asn Val Gln Cys Ser Asp Leu Leu Lys Lys Trp Leu
130 135 140
Pro Gln Arg Cys Ala Thr Phe Ala Ser Lys Ile Gln Gly Gln Val Asp
145 150 155 160
Lys Ile Lys Gly Ala Gly Gly Asp
165
<210> 3
<211> 168
<212> PRT
<213> Artificial sequence
<400> 3
Lys Pro Thr Glu Asn Asn Glu Asp Phe Asn Ile Val Ala Val Ala Ser
1 5 10 15
Asn Phe Ala Thr Thr Asp Leu Asp Ala Asp Arg Gly Lys Leu Pro Gly
20 25 30
Lys Lys Leu Pro Leu Glu Val Leu Lys Glu Leu Glu Ala Asn Ala Arg
35 40 45
Lys Ala Gly Cys Thr Arg Gly Cys Leu Ile Cys Leu Ser His Ile Lys
50 55 60
Cys Thr Pro Lys Met Lys Lys Phe Ile Pro Gly Arg Cys His Thr Tyr
65 70 75 80
Glu Gly Asp Lys Glu Ser Ala Gln Gly Gly Ile Gly Glu Ala Ile Val
85 90 95
Asp Ile Pro Glu Ile Pro Gly Phe Lys Asp Leu Glu Pro Leu Glu Gln
100 105 110
Phe Ile Ala Gln Val Asp Leu Cys Val Asp Cys Thr Thr Gly Cys Leu
115 120 125
Lys Gly Leu Ala Asn Val Gln Cys Ser Asp Leu Leu Lys Lys Trp Leu
130 135 140
Pro Gln Arg Cys Ala Thr Phe Ala Ser Lys Ile Gln Gly Gln Val Asp
145 150 155 160
Lys Ile Lys Gly Ala Gly Gly Asp
165
<210> 4
<211> 15
<212> PRT
<213> Artificial sequence
<400> 4
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15
<210> 5
<211> 17
<212> PRT
<213> Artificial sequence
<400> 5
Met Gly Val Lys Val Leu Phe Ala Leu Ile Cys Ile Ala Val Ala Glu
1 5 10 15
Ala
<210> 6
<211> 334
<212> PRT
<213> Artificial sequence
<400> 6
Met Gly Val Lys Val Leu Phe Ala Leu Ile Cys Ile Ala Val Ala Glu
1 5 10 15
Ala Met Ala Glu Ala Gly Ile Thr Gly Thr Trp Tyr Asn Gln Leu Gly
20 25 30
Ser Thr Phe Ile Val Thr Ala Gly Ala Asp Gly Ala Leu Thr Gly Thr
35 40 45
Tyr Glu Ser Ala Val Gly Asn Ala Glu Ser Arg Tyr Val Leu Thr Gly
50 55 60
Arg Tyr Asp Ser Ala Pro Ala Thr Asp Gly Ser Gly Thr Ala Leu Gly
65 70 75 80
Trp Thr Val Ala Trp Lys Asn Asn Tyr Arg Asn Ala His Ser Ala Thr
85 90 95
Thr Trp Ser Gly Gln Tyr Val Gly Gly Ala Glu Ala Arg Ile Asn Thr
100 105 110
Gln Trp Leu Leu Thr Ser Gly Thr Thr Glu Ala Asn Ala Trp Lys Ser
115 120 125
Thr Leu Val Gly His Asp Thr Phe Thr Lys Val Lys Pro Ser Ala Ala
130 135 140
Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
145 150 155 160
Lys Pro Thr Glu Asn Asn Glu Asp Phe Asn Ile Val Ala Val Ala Ser
165 170 175
Asn Phe Ala Thr Thr Asp Leu Asp Ala Asp Arg Gly Lys Leu Pro Gly
180 185 190
Lys Lys Leu Pro Leu Glu Val Leu Lys Glu Met Glu Ala Asn Ala Arg
195 200 205
Lys Ala Gly Cys Thr Arg Gly Cys Leu Ile Cys Leu Ser His Ile Lys
210 215 220
Cys Thr Pro Lys Met Lys Lys Phe Ile Pro Gly Arg Cys His Thr Tyr
225 230 235 240
Glu Gly Asp Lys Glu Ser Ala Gln Gly Gly Ile Gly Glu Ala Ile Val
245 250 255
Asp Ile Pro Glu Ile Pro Gly Phe Lys Asp Leu Glu Pro Met Glu Gln
260 265 270
Phe Ile Ala Gln Val Asp Leu Cys Val Asp Cys Thr Thr Gly Cys Leu
275 280 285
Lys Gly Leu Ala Asn Val Gln Cys Ser Asp Leu Leu Lys Lys Trp Leu
290 295 300
Pro Gln Arg Cys Ala Thr Phe Ala Ser Lys Ile Gln Gly Gln Val Asp
305 310 315 320
Lys Ile Lys Gly Ala Gly Gly Asp His His His His His His
325 330
<210> 7
<211> 334
<212> PRT
<213> Artificial sequence
<400> 7
Met Gly Val Lys Val Leu Phe Ala Leu Ile Cys Ile Ala Val Ala Glu
1 5 10 15
Ala Met Ala Glu Ala Gly Ile Thr Gly Thr Trp Tyr Asn Gln Leu Gly
20 25 30
Ser Thr Phe Ile Val Thr Ala Gly Ala Asp Gly Ala Leu Thr Gly Thr
35 40 45
Tyr Glu Ser Ala Val Gly Asn Ala Glu Ser Arg Tyr Val Leu Thr Gly
50 55 60
Arg Tyr Asp Ser Ala Pro Ala Thr Asp Gly Ser Gly Thr Ala Leu Gly
65 70 75 80
Trp Thr Val Ala Trp Lys Asn Asn Tyr Arg Asn Ala His Ser Ala Thr
85 90 95
Thr Trp Ser Gly Gln Tyr Val Gly Gly Ala Glu Ala Arg Ile Asn Thr
100 105 110
Gln Trp Leu Leu Thr Ser Gly Thr Thr Glu Ala Asn Ala Trp Lys Ser
115 120 125
Thr Leu Val Gly His Asp Thr Phe Thr Lys Val Lys Pro Ser Ala Ala
130 135 140
Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
145 150 155 160
Lys Pro Thr Glu Asn Asn Glu Asp Phe Asn Ile Val Ala Val Ala Ser
165 170 175
Asn Phe Ala Thr Thr Asp Leu Asp Ala Asp Arg Gly Lys Leu Pro Gly
180 185 190
Lys Lys Leu Pro Leu Glu Val Leu Lys Glu Leu Glu Ala Asn Ala Arg
195 200 205
Lys Ala Gly Cys Thr Arg Gly Cys Leu Ile Cys Leu Ser His Ile Lys
210 215 220
Cys Thr Pro Lys Met Lys Lys Phe Ile Pro Gly Arg Cys His Thr Tyr
225 230 235 240
Glu Gly Asp Lys Glu Ser Ala Gln Gly Gly Ile Gly Glu Ala Ile Val
245 250 255
Asp Ile Pro Glu Ile Pro Gly Phe Lys Asp Leu Glu Pro Leu Glu Gln
260 265 270
Phe Ile Ala Gln Val Asp Leu Cys Val Asp Cys Thr Thr Gly Cys Leu
275 280 285
Lys Gly Leu Ala Asn Val Gln Cys Ser Asp Leu Leu Lys Lys Trp Leu
290 295 300
Pro Gln Arg Cys Ala Thr Phe Ala Ser Lys Ile Gln Gly Gln Val Asp
305 310 315 320
Lys Ile Lys Gly Ala Gly Gly Asp His His His His His His
325 330
<210> 8
<211> 384
<212> DNA
<213> Artificial sequence
<400> 8
atggctgagg ctggaatcac cggaacatgg tacaaccagc tgggcagcac ctttatcgtg 60
acagctggag ctgatggcgc cctgaccggc acatatgagt ccgctgtggg caatgctgag 120
agcagatacg tgctgaccgg ccggtatgat tctgctccag ctaccgacgg atccggaaca 180
gctctgggat ggaccgtggc ctggaagaac aattacagga acgctcactc tgccaccaca 240
tggtccggac agtatgtggg aggagctgag gctcggatca acacacagtg gctgctgacc 300
agcggcacca cagaggctaa tgcctggaag tctaccctgg tgggccatga taccttcaca 360
aaggtgaagc cctccgccgc tagc 384
<210> 9
<211> 504
<212> DNA
<213> Artificial sequence
<400> 9
aagccaacag agaacaatga ggatttcaac atcgtggctg tggccagcaa ttttgctacc 60
acagacctgg atgccgacag aggcaagctg ccaggcaaga agctgcccct ggaggtgctg 120
aaggagatgg aggctaacgc taggaaggct ggatgtacca ggggatgcct gatctgtctg 180
tctcacatca agtgcacacc taagatgaag aagttcatcc caggccgctg tcatacctac 240
gagggcgata aggagtccgc tcagggagga atcggagagg ccatcgtgga tatccccgag 300
atccctggct tcaaggacct ggagcccatg gagcagttta tcgctcaggt ggatctgtgc 360
gtggactgta ccacaggctg cctgaagggc ctggccaatg tgcagtgttc cgacctgctg 420
aagaagtggc tgcctcagag gtgcgctacc tttgccagca agatccaggg ccaggtggat 480
aagatcaagg gagctggagg cgac 504
<210> 10
<211> 504
<212> DNA
<213> Artificial sequence
<400> 10
aagccaacag agaacaatga ggatttcaac atcgtggctg tggccagcaa ttttgctacc 60
acagacctgg atgccgacag aggcaagctg ccaggcaaga agctgcccct ggaggtgctg 120
aaggagctcg aggctaacgc taggaaggct ggatgtacca ggggatgcct gatctgtctg 180
tctcacatca agtgcacacc taagatgaag aagttcatcc caggccgctg tcatacctac 240
gagggcgata aggagtccgc tcagggagga atcggagagg ccatcgtgga tatccccgag 300
atccctggct tcaaggacct ggagcccctc gagcagttta tcgctcaggt ggatctgtgc 360
gtggactgta ccacaggctg cctgaagggc ctggccaatg tgcagtgttc cgacctgctg 420
aagaagtggc tgcctcagag gtgcgctacc tttgccagca agatccaggg ccaggtggat 480
aagatcaagg gagctggagg cgac 504
<210> 11
<211> 45
<212> DNA
<213> Artificial sequence
<400> 11
ggaggaggag gatctggagg aggaggatcc ggaggaggag gatct 45
<210> 12
<211> 51
<212> DNA
<213> Artificial sequence
<400> 12
atgggcgtga aggtgctgtt cgccctgatc tgcatcgccg tggctgaggc c 51
<210> 13
<211> 1002
<212> DNA
<213> Artificial sequence
<400> 13
atgggcgtga aggtgctgtt cgccctgatc tgcatcgccg tggctgaggc catggctgag 60
gctggaatca ccggaacatg gtacaaccag ctgggcagca cctttatcgt gacagctgga 120
gctgatggcg ccctgaccgg cacatatgag tccgctgtgg gcaatgctga gagcagatac 180
gtgctgaccg gccggtatga ttctgctcca gctaccgacg gatccggaac agctctggga 240
tggaccgtgg cctggaagaa caattacagg aacgctcact ctgccaccac atggtccgga 300
cagtatgtgg gaggagctga ggctcggatc aacacacagt ggctgctgac cagcggcacc 360
acagaggcta atgcctggaa gtctaccctg gtgggccatg ataccttcac aaaggtgaag 420
ccctccgccg ctagcggagg aggaggatct ggaggaggag gatccggagg aggaggatct 480
aagccaacag agaacaatga ggatttcaac atcgtggctg tggccagcaa ttttgctacc 540
acagacctgg atgccgacag aggcaagctg ccaggcaaga agctgcccct ggaggtgctg 600
aaggagatgg aggctaacgc taggaaggct ggatgtacca ggggatgcct gatctgtctg 660
tctcacatca agtgcacacc taagatgaag aagttcatcc caggccgctg tcatacctac 720
gagggcgata aggagtccgc tcagggagga atcggagagg ccatcgtgga tatccccgag 780
atccctggct tcaaggacct ggagcccatg gagcagttta tcgctcaggt ggatctgtgc 840
gtggactgta ccacaggctg cctgaagggc ctggccaatg tgcagtgttc cgacctgctg 900
aagaagtggc tgcctcagag gtgcgctacc tttgccagca agatccaggg ccaggtggat 960
aagatcaagg gagctggagg cgaccaccat caccatcacc at 1002
<210> 14
<211> 1002
<212> DNA
<213> Artificial sequence
<400> 14
atgggcgtga aggtgctgtt cgccctgatc tgcatcgccg tggctgaggc catggctgag 60
gctggaatca ccggaacatg gtacaaccag ctgggcagca cctttatcgt gacagctgga 120
gctgatggcg ccctgaccgg cacatatgag tccgctgtgg gcaatgctga gagcagatac 180
gtgctgaccg gccggtatga ttctgctcca gctaccgacg gatccggaac agctctggga 240
tggaccgtgg cctggaagaa caattacagg aacgctcact ctgccaccac atggtccgga 300
cagtatgtgg gaggagctga ggctcggatc aacacacagt ggctgctgac cagcggcacc 360
acagaggcta atgcctggaa gtctaccctg gtgggccatg ataccttcac aaaggtgaag 420
ccctccgccg ctagcggagg aggaggatct ggaggaggag gatccggagg aggaggatct 480
aagccaacag agaacaatga ggatttcaac atcgtggctg tggccagcaa ttttgctacc 540
acagacctgg atgccgacag aggcaagctg ccaggcaaga agctgcccct ggaggtgctg 600
aaggagctcg aggctaacgc taggaaggct ggatgtacca ggggatgcct gatctgtctg 660
tctcacatca agtgcacacc taagatgaag aagttcatcc caggccgctg tcatacctac 720
gagggcgata aggagtccgc tcagggagga atcggagagg ccatcgtgga tatccccgag 780
atccctggct tcaaggacct ggagcccctc gagcagttta tcgctcaggt ggatctgtgc 840
gtggactgta ccacaggctg cctgaagggc ctggccaatg tgcagtgttc cgacctgctg 900
aagaagtggc tgcctcagag gtgcgctacc tttgccagca agatccaggg ccaggtggat 960
aagatcaagg gagctggagg cgaccaccat caccatcacc at 1002
<210> 15
<211> 6
<212> PRT
<213> Artificial sequence
<400> 15
Gly Gly Gly Gly Gly Ser
1 5
<210> 16
<211> 11
<212> PRT
<213> Artificial sequence
<400> 16
Gly Gln Gly Gln Gly Gln Gly Gln Gly Gln Gly
1 5 10
<210> 17
<211> 15
<212> PRT
<213> Artificial sequence
<400> 17
Gly Ser Thr Ser Gly Ser Gly Lys Ser Ser Glu Lys Gly Lys Gly
1 5 10 15
<210> 18
<211> 10
<212> PRT
<213> Artificial sequence
<400> 18
Val Pro Gly Val Gly Val Pro Gly Val Gly
1 5 10
<210> 19
<211> 8
<212> PRT
<213> Artificial sequence
<400> 19
Ser Ala Pro Gly Thr Pro Ser Arg
1 5
<210> 20
<211> 14
<212> PRT
<213> Artificial sequence
<400> 20
Glu Gly Lys Ser Ser Gly Ser Gly Ser Glu Ser Lys Glu Phe
1 5 10
<210> 21
<211> 6
<212> PRT
<213> Artificial sequence
<400> 21
Gly Ser Gly Gly Ser Gly
1 5
<210> 22
<211> 8
<212> PRT
<213> Artificial sequence
<400> 22
Gly Ser Gly Gly Ser Gly Gly Gly
1 5

Claims (22)

1. A fusion protein of streptavidin and Gaussian luciferase, which is characterized by comprising a streptavidin full-length protein and a Gaussian luciferase persistent luminescent mutant, wherein the persistent luminescent mutant has a sequence shown in SEQ ID NO: 3, the streptavidin full-length protein is connected with the Gaussian luciferase continuous luminescence mutant through a flexible peptide.
2. The fusion protein of claim 1, wherein the streptavidin full-length protein has the sequence shown in SEQ ID NO: 1 is shown.
3. The fusion protein of claim 1, wherein the flexible peptide sequence is as set forth in SEQ ID NO: 4, respectively.
4. The fusion protein of claim 1, further comprising: a signal peptide for directing secretion of the fusion protein from within the expressing cell to outside.
5. The fusion protein of claim 4, wherein the signal peptide is located at the N-terminus of the fusion protein.
6. The fusion protein of claim 4, wherein the signal peptide sequence is as set forth in SEQ ID NO: 5, respectively.
7. The fusion protein of claim 1, further comprising: a fusion tag for affinity purification of the fusion protein.
8. The fusion protein of claim 7, wherein the fusion tag is a 6 × histidine tag.
9. The fusion protein of claim 1, wherein the fusion protein has the sequence set forth in SEQ ID NO: shown at 7.
10. An isolated nucleic acid encoding the fusion protein of claim 1.
11. The isolated nucleic acid of claim 10, wherein the isolated nucleic acid comprises a streptavidin full-length protein coding sequence and a gauss luciferase luminescence-sustaining mutant coding sequence, wherein the streptavidin full-length protein coding sequence is linked to the gauss luciferase luminescence-sustaining mutant coding sequence by a linker sequence, and wherein the gauss luciferase luminescence-sustaining mutant coding sequence is as set forth in SEQ ID NO: shown at 10.
12. The isolated nucleic acid of claim 11, wherein the streptavidin full-length protein coding sequence is set forth in SEQ ID NO: shown in fig. 8.
13. The isolated nucleic acid of claim 11, wherein the linker sequence is set forth in SEQ ID NO: shown at 11.
14. The isolated nucleic acid of claim 11, further comprising: a signal peptide coding sequence which encodes a signal peptide for directing secretion of the fusion protein from the expressing cell to the outside.
15. The isolated nucleic acid of claim 14, wherein the signal peptide coding sequence is located 5' to the isolated nucleic acid.
16. The isolated nucleic acid of claim 14, wherein the signal peptide coding sequence is set forth in SEQ ID NO: shown at 12.
17. The isolated nucleic acid of claim 11, further comprising: a fusion tag coding sequence encoding a fusion tag for affinity purification of the fusion protein.
18. The isolated nucleic acid of claim 17, wherein the fusion tag coding sequence is a sequence encoding a 6 x histidine tag.
19. The isolated nucleic acid of claim 10, wherein the isolated nucleic acid sequence is as set forth in SEQ ID NO: as shown at 14.
20. An expression vector comprising a nucleic acid sequence encoding the fusion protein of any one of claims 1-9, and a vector backbone sequence.
21. A recombinant host cell comprising the expression vector of claim 20.
22. Use of the fusion protein of any one of claims 1 to 9, the isolated nucleic acid of any one of claims 10-19, the expression vector of claim 20, or the recombinant host cell of claim 21 in a biotin molecule assay.
CN201811473694.5A 2018-12-04 2018-12-04 Fusion protein of streptavidin and Gaussian luciferase and application thereof Active CN111269322B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811473694.5A CN111269322B (en) 2018-12-04 2018-12-04 Fusion protein of streptavidin and Gaussian luciferase and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811473694.5A CN111269322B (en) 2018-12-04 2018-12-04 Fusion protein of streptavidin and Gaussian luciferase and application thereof

Publications (2)

Publication Number Publication Date
CN111269322A CN111269322A (en) 2020-06-12
CN111269322B true CN111269322B (en) 2022-02-18

Family

ID=70994703

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811473694.5A Active CN111269322B (en) 2018-12-04 2018-12-04 Fusion protein of streptavidin and Gaussian luciferase and application thereof

Country Status (1)

Country Link
CN (1) CN111269322B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023123509A1 (en) * 2021-12-31 2023-07-06 深圳华大生命科学研究院 Mutant and uses thereof
CN114478725B (en) * 2022-01-19 2023-07-07 大连博格林生物科技有限公司 Streptavidin mutant and preparation method and application thereof
WO2023109981A2 (en) * 2023-04-11 2023-06-22 深圳华大智造科技股份有限公司 Novel copepod luciferase mutant and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105462939A (en) * 2015-12-30 2016-04-06 北京中科紫鑫科技有限责任公司 Expressing method for biotinylation luciferase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105462939A (en) * 2015-12-30 2016-04-06 北京中科紫鑫科技有限责任公司 Expressing method for biotinylation luciferase

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GenBank:ACI46139.1;Colussi,P.A.等;《GenBank:ACI46139.1》;20160726;第1页 *
GenBank:CAB69589.1;Mueller,R.D.等;《GenBank:CAB69589.1》;20000122;第1页 *
Luciferase-streptavidin fusion proteins: Preparation and properties;Smirnova, DV等;《Moscow University Chemistry Bulletin》;20140331;第69卷(第2期);第56页摘要,第60页右栏第1段第1-8行 *
Recombinant Gaussia luciferase. Overexpression, purification, and analytical application of a bioluminescent reporter for DNA hybridization;Verhaegen, M等;《Analytical Chemistry》;20020901;第74卷(第17期);第4378页左栏第1段,第4385页右栏第1段 *

Also Published As

Publication number Publication date
CN111269322A (en) 2020-06-12

Similar Documents

Publication Publication Date Title
US11390653B2 (en) Amino acid-specific binder and selectively identifying an amino acid
CN111269322B (en) Fusion protein of streptavidin and Gaussian luciferase and application thereof
CN111269323B (en) Fusion protein of monomer streptavidin and gauss luciferase and application thereof
CN105073770A (en) Streptavidin muteins and methods of using them
CN111269324B (en) Fusion protein of Gauss luciferase and digoxin single-chain antibody and application thereof
CN113195521A (en) Mtu Delta I-CM intein variants and uses thereof
CN108796040B (en) Bioluminescence detection probe based on transcription activator-like effector and construction method and application thereof
CN106434699A (en) SUMO and SUMO protease encoding gene and application thereof
CA2474108A1 (en) Novel fluorescent proteins from aequorea coerulscens and methods for using same
US20050142623A1 (en) Sensor protein and use thereof
CN117362447A (en) Bioluminescence enzyme-linked immunoassay method based on protein coupled polymer
Dillard et al. Sortase-mediated fluorescent labeling of CRISPR complexes
KR20210055754A (en) Anti-human myocardial troponin I antibody and its use
CN114057861B (en) bio-PROTAC artificial protein targeting UBE2C
CN113025586B (en) Modified luciferase mutant protein, bioluminescent probe, probe set, preparation method and detection method
KR20100001090A (en) Novel peptides with specific binding to fc domain of igg and their preparation methods
CN109880840B (en) In vivo biotinylation labeling system for recombinant protein escherichia coli
US8163521B2 (en) Self-assembled proteins and related methods and protein structures
US7241864B2 (en) Recombinant photoproteins and their conjugates
CN107629112B (en) High-affinity LC3 protein targeting peptide and application thereof
WO2024000408A1 (en) Luciferase mutant and use thereof
Pal et al. PBP isolation and dd-carboxypeptidase assay
CN110564744A (en) DNA polymerase, preparation method thereof, expression gene, expression vector, host cell and kit
CN110684086B (en) Fluorescent protein for detecting copper ions and coding nucleotide sequence and application thereof
CN115703843A (en) Fusion protein, related nucleic acid molecule thereof and application thereof in high-throughput sequencing

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right

Effective date of registration: 20231018

Address after: 266400 building 4, No. 2, hengyunshan Road, Qingdao area, China (Shandong) pilot Free Trade Zone, Huangdao District, Qingdao City, Shandong Province

Patentee after: Qingdao Huada Zhizao Technology Co.,Ltd.

Address before: 518083 7th floor, building 11, Beishan Industrial Zone, 146 Beishan Road, Yangang community, Yantian street, Yantian District, Shenzhen City, Guangdong Province

Patentee before: Shenzhen Huada zhizaojichuang Technology Co.,Ltd.

TR01 Transfer of patent right