CN112391366B - Dre recombination system activated by light induction - Google Patents

Dre recombination system activated by light induction Download PDF

Info

Publication number
CN112391366B
CN112391366B CN201910759889.4A CN201910759889A CN112391366B CN 112391366 B CN112391366 B CN 112391366B CN 201910759889 A CN201910759889 A CN 201910759889A CN 112391366 B CN112391366 B CN 112391366B
Authority
CN
China
Prior art keywords
dre
fragment
fusion protein
leu
ala
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
CN201910759889.4A
Other languages
Chinese (zh)
Other versions
CN112391366A (en
Inventor
李大力
李慧莹
吴英尹
刘明耀
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.)
East China Normal University
Bioray Laboratories Inc
Original Assignee
East China Normal University
Bioray Laboratories Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by East China Normal University, Bioray Laboratories Inc filed Critical East China Normal University
Priority to CN201910759889.4A priority Critical patent/CN112391366B/en
Publication of CN112391366A publication Critical patent/CN112391366A/en
Application granted granted Critical
Publication of CN112391366B publication Critical patent/CN112391366B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention provides a Dre recombination system activated by light induction. The light-induced activated Dre recombination system has higher space-time specificity, can be applied to multiple fields of lineage tracing, cell clearing and the like, combines the specificity of a promoter, and realizes more accurate gene operation by utilizing local blue light irradiation.

Description

Dre recombination system activated by light induction
Technical Field
The invention relates to the technical field of biology, in particular to a Dre recombination system activated by light induction.
Background
The Dre recombinase is derived from the D6 bacteriophage and belongs to the tyrosine recombinase family like the common Cre recombinase. It can specifically recognize a DNA sequence (rox) with the length of 32bp, so that the gene sequence between rox sites is deleted or recombined.
The inducible recombinase system that is currently used is DreER, which expresses the ligand binding domain of the estrogen receptor fused to Dre recombinase to form a cytoplasmic-localized fusion protein. The Dre recombinase dissociates from the dockerin into the nucleus upon the appearance of the ligand tamoxifen to exert a recombination function. Alternatively, RU 486-induced DrePBD system was used to regulate the expression of the recombinase in a time-specific manner. However, these methods have certain limitations, one of them is that the induction by chemical drugs may disturb the endogenous signal pathway of cells, for example, tamoxifen may bind to endogenous estrogen receptor, thereby affecting the normal function of the body. The induction of two chemical drugs takes a long time, the animals need to be treated by the drugs for at least one week, and the drugs are easy to be metabolized rapidly so as to inhibit the induction effect. The induction range of the three drugs is too large, the space specificity can be realized to a certain extent by virtue of a specific promoter, but the precise operation cannot be realized for cell populations which are distributed in the whole body and lack of tissue characteristics.
Therefore, there is an urgent need in the art to establish a system for inducing expression of Dre recombinase by spatio-temporal specific regulation.
Disclosure of Invention
The invention aims to establish a Dre recombinase expression induction system capable of regulating and controlling the Dre recombinase in a space-time specificity manner.
In a first aspect of the invention there is provided a resolved protein comprising a Z1 element or a Z2 element, wherein the Z1 element is an N-terminal fragment of Dre recombinase; the Z2 element is a C-terminal fragment of Dre recombinase;
and, when Z1 is Dre 1-60 When fragment, Z2 is Dre 61-343 A fragment;
when Z1 is Dre 1-65 When fragment, Z2 is Dre 66-343 A fragment;
when Z1 is Dre 1-66 When fragment, Z2 is Dre 67-343 A fragment;
when Z1 is Dre 1-67 When fragment, Z2 is Dre 68-343 A fragment;
when Z1 is Dre 1-103 When fragment, Z2 is Dre 104-343 A fragment;
when Z1 is Dre 1-104 When fragment, Z2 is Dre 105-343 A fragment;
when Z1 is Dre 1-105 When fragment, Z2 is Dre 106-343 A fragment;
when Z1 is Dre 1-107 When fragment, Z2 is Dre 108-343 A fragment;
when Z1 is Dre 1-108 When fragment, Z2 is Dre 109-343 A fragment;
when Z1 is Dre 1-109 When fragment, Z2 is Dre 110-343 A fragment;
when Z1 is Dre 1-150 When fragment, Z2 is Dre 151-343 A fragment;
when Z1 is Dre 1-151 When fragment, Z2 is Dre 152-343 A fragment;
when Z1 is Dre 1-152 When fragment, Z2 is Dre 153-343 A fragment;
when Z1 is Dre 1-157 When fragment, Z2 is Dre 158-343 A fragment;
when Z1 is Dre 1-190 When fragment, Z2 is Dre 191-343 A fragment;
when Z1 is Dre 1-192 When fragment, Z2 is Dre 193-343 A fragment;
when Z1 is Dre 1-193 When fragment, Z2 is Dre 194-343 A fragment;
when Z1 is Dre 1-194 When fragment, Z2 is Dre 195-343 A fragment;
when Z1 is Dre 1-230 When fragment, Z2 is Dre 231-343 A fragment;
when Z1 is Dre 1-234 When fragment, Z2 is Dre 235-343 A fragment;
when Z1 is Dre 1-237 When fragment, Z2 is Dre 238-343 A fragment;
when Z1 is Dre 1-240 When fragment, Z2 is Dre 241-343 A fragment;
when Z1 is Dre 1-246 When fragment, Z2 is Dre 247-343 And (3) fragment.
In a second aspect, the invention provides a polypeptide which is an active fragment obtained by cutting at the following sites of Dre recombinase shown in SEQ ID NO. 10:
60 th, 65 th, 66 th, 67 th, 103 th, 104 th, 105 th, 107 th, 108 th, 109 th, 150 th, 151 th, 152 th, 157 th, 190 th, 192 th, 193 th, 194 th, 230 th, 234 th, 237 th, 240 th, and/or 246 th bits.
In another preferred embodiment, the polypeptide is an active fragment obtained by cutting the Dre recombinase shown in SEQ ID NO. 10 at the following sites:
bits 60, 150, 151, 152, 157, 190, 192, 193, 194, 234 and/or 246.
In another preferred embodiment, the polypeptide is an active fragment obtained by cleavage of the Dre recombinase shown in SEQ ID No. 10 at the following sites:
60, 150 and/or 246.
In a third aspect, the present invention provides a fusion protein selected from the group consisting of: a first fusion protein, a second fusion protein, or a combination thereof; wherein the structure of the first fusion protein is shown as formula I or formula I':
Z1-L1-A1 (I);
A1-L1-Z1 (I’);
the structure of the second fusion protein is shown as formula II or formula II':
Z2-L2-A2 (II);
A2-L2-Z2 (II’);
in the formula (I), the compound is shown in the specification,
z1 is an N-terminal fragment of Dre recombinase;
l1 is an optional first linker peptide;
a1 is a first element of a light sensitive protein;
z2 is a C-terminal fragment of Dre recombinase;
a2 is a second element of the light sensitive protein;
l2 is an optional second linking peptide;
each "-" is independently a linking peptide or non-peptide bond.
In another preferred embodiment, the N-terminal fragment of Dre recombinase is selected from the group consisting of: dre 1-60 Fragment, dre 1-65 Fragment, dre 1-66 Fragment, dre 1-67 Fragment, dre 1-103 Fragment, dre 1-104 Fragment, dre 1-105 Fragment, dre 1-107 Fragment, dre 1-108 Fragment, dre 1-109 Fragment, dre 1-150 Fragment, dre 1-151 Fragment, dre 1-152 Fragment, dre 1-157 Fragment, dre 1-190 Fragment, dre 1-192 Fragment, dre 1-193 Fragment, dre 1-194 Fragment, dre 1-230 Fragment, dre 1-234 Fragment, dre 1-237 Fragment, dre 1-240 Fragment, dre 1-246 Fragments, or combinations thereof.
In another preferred embodiment, the N-terminal fragment of the Dre recombinase is based on the N-terminal fragment of SEQ ID No. 10.
In another preferred embodiment, the N-terminal fragment of Dre recombinase is selected from the group consisting of: dre 1-60 Fragment, dre 1-150 Fragment, dre 1-151 Fragment, dre 1-152 Fragment, dre 1-157 Fragment, dre 1-190 Fragment, dre 1-192 Fragment, dre 1-193 Fragment, dre 1-194 Fragment, dre 1-234 Fragment, dre 1-246 Fragments, or combinations thereof.
In another preferred embodiment, the N-terminal fragment of Dre recombinase is selected from the group consisting of: dre 1-60 Fragment, dre 1-150 Fragment, dre 1-246 Fragments, or combinations thereof.
In another preferred embodiment, the lengths of L1 and L2 are each independently 1 to 80aa, preferably 5 to 60aa, and more preferably 10 to 50aa.
In another preferred embodiment, L1 and L2 are each independently selected from the group consisting of:
(1) A polypeptide having an amino acid sequence as set forth in any one of SEQ ID No. 1-5;
(2) 1-5 by one or more, preferably 1-20, more preferably 1-15, more preferably 1-10, more preferably 1-8, more preferably 1-3, most preferably 1 amino acid residue, and having (1) the function of the polypeptide, and a polypeptide derived from the polypeptide of any one of the amino acid sequences shown in SEQ ID No. 1-5.
In another preferred embodiment, the light sensitive protein is selected from the group consisting of: a blue light-sensitive protein, a red light-sensitive protein, an ultraviolet light-sensitive protein, or a combination thereof.
In another preferred embodiment, the blue light sensitive protein is selected from the group consisting of: LOV2, VVD, CRY2 cluster, magnet, CRY2/CIB1, or combinations thereof.
In another preferred embodiment, the red light sensitive protein is selected from the group consisting of: bphP1/Q-PAS1, phyB/PIF3& PhyB/PIF6, cph1, or combinations thereof.
In another preferred example, the ultraviolet light sensitive protein comprises UVR8/COP1.
In another preferred embodiment, the first element of the light-sensitive protein comprises nMag.
In another preferred embodiment, the second element of the light sensitive protein comprises pMag.
In another preferred embodiment, the C-terminal fragment of Dre recombinase is selected from the group consisting of: dre 61-343 Fragment, dre 66-343 Fragment, dre 67-343 Fragment, dre 68-343 Fragment, dre 104-343 Fragment, dre 105-343 Fragment, dre 106-343 Fragment, dre 108-343 Fragment, dre 109-343 Fragment, dre 110-343 Fragment, dre 151-343 Fragment, dre 152-343 Fragment, dre 153-343 Fragment, dre 158-343 Fragment, dre 191-343 Fragment, dre 193-343 Fragment, dre 194-343 Fragment, dre 195-343 Fragment, dre 231-343 Fragment, dre 234-343 Fragment, dre 238-343 Fragment, dre 241-343 Fragment, dre 247-343 Fragments, or combinations thereof.
In another preferred embodiment, the C-terminal fragment of the Dre recombinase is based on the C-terminal fragment of SEQ ID NO. 10.
In another preferred embodiment, the C-terminal fragment of Dre recombinase is selected from the group consisting of: dre 61-343 Fragment, dre 151-343 Fragment, dre 152-343 Fragment, dre 153-343 Fragment, dre 158-343 Fragment, dre 191-343 Fragment, dre 193-343 Fragment, dre 194-343 Fragment, dre 195-343 Fragment, dre 235-343 Fragment, dre 247-343 Fragments, or combinations thereof.
In another preferred embodiment, the C-terminal fragment of Dre recombinase is selected from the group consisting of: dre 61-343 Fragment, dre 151-343 Fragment, dre 247-343 Fragments, or combinations thereof.
In another preferred embodiment, the fusion protein further comprises a nuclear localization signal.
In another preferred embodiment, the nuclear localization signal is selected from the group consisting of: NLS sv40 、NLS EGL-13 、NLS NLP 、NLS c-Myc 、PY-NLSs、snRNPs、truncated NLS sv40 、NLS TUS Or a combination thereof.
In another preferred embodiment, the amino acid sequences of the nuclear localization signals are each independently selected from the group consisting of:
(1) A polypeptide having an amino acid sequence as set forth in any one of SEQ ID No. 6-9;
(2) A polypeptide derived from the polypeptide of the amino acid sequence shown in any one of SEQ ID No. 6-9, which has the function of the polypeptide of (1), formed by substituting, deleting or adding one or more, preferably 1-20, more preferably 1-15, more preferably 1-10, more preferably 1-8, more preferably 1-3, and most preferably 1 amino acid residue in the amino acid sequence shown in any one of SEQ ID No. 6-9.
In another preferred embodiment, the N-terminal fragment of Dre recombinase and the C-terminal fragment of Dre recombinase together constitute active Dre recombinase.
In another preferred embodiment, the first element of the light-sensitive protein and the second element of the light-sensitive protein together constitute an active light-sensitive protein.
In another preferred embodiment, the first element of the light sensitive protein binds to the second element of the light sensitive protein when the first fusion protein binds to the second fusion protein.
In another preferred embodiment, the fusion protein has the amino acid sequence as shown in any one of SEQ ID No. 11-12.
In another preferred embodiment, the first fusion protein and the second fusion protein combine to form a complex comprising the first fusion protein and the second fusion protein.
In a fourth aspect, the present invention provides a polynucleotide encoding the resolution protein of the first aspect, the polypeptide of the second aspect, or the fusion protein of the third aspect of the present invention.
In another preferred embodiment, said polynucleotide additionally comprises an auxiliary element selected from the group consisting of: a signal peptide, a secretory peptide, a tag sequence (e.g., 6 His), or a combination thereof.
In another preferred embodiment, the polynucleotide is selected from the group consisting of: a DNA sequence, an RNA sequence, or a combination thereof.
In another preferred embodiment, the polynucleotide further comprises a promoter.
In another preferred embodiment, the promoter is selected from the group consisting of: EF1 α, CMV, PGK, CAG, LP1 (liver specific promoter), ctnt (heart specific promoter), syn (interneuron specific promoter), or combinations thereof.
In another preferred embodiment, the nucleic acid construct has a nucleotide sequence as set forth in any one of SEQ ID No. 13-14.
In a fifth aspect, the present invention provides a vector comprising a polynucleotide according to the fourth aspect of the present invention.
In another preferred embodiment, the vector comprises one or more promoters operably linked to the nucleic acid sequence, enhancer, transcription termination signal, polyadenylation sequence, origin of replication, selectable marker, nucleic acid restriction site, and/or homologous recombination site.
In another preferred embodiment, the vector comprises a plasmid, a viral vector.
In another preferred embodiment, the viral vector is selected from the group consisting of: adeno-associated virus (AAV), adenovirus, lentivirus, retrovirus, herpes virus, SV40, poxvirus, or combinations thereof.
In another preferred embodiment, the vector comprises an expression vector, a shuttle vector and an integration vector.
In a sixth aspect, the invention provides a host cell comprising a vector according to the fifth aspect of the invention or having a polynucleotide according to the fourth aspect of the invention integrated into its genome.
In another preferred embodiment, the host cell includes prokaryotic cells and eukaryotic cells.
In another preferred embodiment, the eukaryotic cell comprises a higher eukaryotic cell.
In another preferred embodiment, the cells are from the following species: human, non-human mammal, poultry, plant.
In another preferred embodiment, the non-human mammal includes a rodent (e.g., mouse, rat, rabbit), cow, pig, sheep, horse, dog, cat, non-human primate (e.g., monkey).
In another preferred embodiment, the cell comprises: somatic cells, stem cells, germ cells, non-dividing cells, or combinations thereof.
In another preferred embodiment, the cell comprises: kidney cells, epithelial cells, endothelial cells, or a combination thereof.
In another preferred embodiment, the genetically engineered cell is selected from the group consisting of: human cells, chinese hamster ovary cells, insect cells, wheat germ cells, rabbit reticulocytes, yeast cells, or a combination thereof.
In another preferred embodiment, the genetically engineered cell is selected from the group consisting of: HEK-293, hela, hep2, SH-SY5Y, or a combination thereof.
In a seventh aspect, the present invention provides a method of producing a split protein according to the first aspect of the invention, a polypeptide according to the second aspect of the invention, or a fusion protein according to the third aspect of the invention, comprising the steps of:
culturing the host cell of the sixth aspect of the invention under conditions suitable for expression, thereby expressing the split protein, polypeptide or fusion protein; and/or isolating the split protein, polypeptide or fusion protein.
The eighth aspect of the present invention provides a gene editing reagent comprising the fusion protein according to the third aspect of the present invention.
The ninth aspect of the present invention provides a kit comprising the gene editing reagent according to the eighth aspect of the present invention.
In another preferred embodiment, the kit further comprises a label or instructions.
In a tenth aspect, the invention provides a use of the fusion protein of the third aspect of the invention for preparing a reagent or a kit for improving gene editing efficiency.
The eleventh aspect of the present invention provides a method for gene editing, comprising the steps of:
(a) Providing a first fusion protein and a second fusion protein, wherein the structures of the first fusion protein and the second fusion protein are as set forth in the third aspect of the invention;
(b) Under light, the first fusion protein and the second fusion protein form a complex, thereby performing gene editing in the presence of the complex.
In another preferred example, the illumination comprises blue illumination.
In another preferred embodiment, the illumination intensity is 0.1-20mw/cm 2 Preferably, 0.2 to 10mw/cm 2 More preferably, 1 to 5mw/cm 2
In another preferred embodiment, the illumination time is 2h to 72h, preferably 10 h to 48h, and more preferably 15 h to 40h.
In another preferred embodiment, the method is non-diagnostic and non-therapeutic.
In another preferred embodiment, the gene editing comprises: site-specific cleavage, site-specific insertion, and site-specific recombination.
In another preferred embodiment, said gene editing is performed in a cell in vitro.
In another preferred embodiment, the cells are from the following species: human, non-human mammal, poultry, plant.
In another preferred embodiment, the non-human mammal includes a rodent (e.g., mouse, rat, rabbit), cow, pig, sheep, horse, dog, cat, non-human primate (e.g., monkey).
In another preferred embodiment, the cell comprises: somatic cells, stem cells, germ cells, non-dividing cells, or combinations thereof.
In another preferred embodiment, the cell comprises: kidney cells, epithelial cells, endothelial cells, or a combination thereof.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1 shows functional verification of different fragment sites of Dre recombinase.
Left panel: schematic of plasmid structure right panel: flow cytometric analysis results
FIG. 2 shows activity screening after segmented Dre recombinase fusion with light sensitive proteins.
Left panel: plasmid structure schematic right panel: results of luciferase Activity detection
FIG. 3 shows the relative positional adjustment of the segmented Dre recombinase to the light-sensitive protein.
Left panel: plasmid structure schematic right panel: results of luciferase Activity detection
FIG. 4 shows a comparison of the activity of the segmented Dre recombinase of different linkers.
FIG. 5 shows a comparison of the activity of the segmented Dre recombinases for different NLSs.
FIG. 6 shows a comparison of recombinase activity for a two plasmid system versus a single plasmid system.
Fig. 7 shows a schematic diagram of the PA-Dre operation.
Fig. 8 shows the illumination time dependence of the PA-Dre system.
Fig. 9 shows the illumination intensity dependence of the PA-Dre system.
FIG. 10 shows that the PA-Dre system is applicable to different promoters.
FIG. 11 shows that the PA-Dre system is suitable for different cell lines.
FIG. 12 shows the detection of luciferase activity in mice at different illumination times when low doses of PA-Dre plasmid were injected.
Left panel: right image of mouse live imaging results: results of luciferase Activity detection
FIG. 13 shows the luciferase activity assay of mice after light irradiation when high doses of PA-Dre plasmid were injected.
Left panel: right image of mouse in vivo imaging results: mouse liver living body imaging result statistics
Detailed Description
The inventor of the invention has conducted extensive and intensive research, and found for the first time, unexpectedly, that the complete Dre recombinase is split into an N-terminal segment and a C-terminal segment, and the N-terminal segment and the C-terminal segment are fused and expressed with photosensitive proteins nMAG and pMag respectively through a linker, so that a light-activated Dre recombinase system, also called PA-Dre, is formed. In the absence of blue light, the segmented Dre recombinase has no catalytic activity and rapidly dimerizes with the light-sensitive protein to form a catalytically active recombinase when the system is stimulated with blue light. Moreover, the applicant also unexpectedly finds that compared with a traditional induction system such as a Dre recombinase induced by tamoxifen, the PA-Dre system of the invention does not affect an endogenous signal path of a cell, has higher space-time specificity, can be applied to multiple fields such as lineage tracing, cell clearing and the like, combines the specificity of a promoter, and realizes more accurate gene operation by local blue light irradiation. On this basis, the present inventors have completed the present invention.
Term(s) for
In order that the disclosure may be more readily understood, certain terms are first defined. As used in this application, each of the following terms shall have the meaning given below, unless explicitly specified otherwise herein. Other definitions are set forth throughout the application.
The term "about" can refer to a value or composition that is within an acceptable error range for the particular value or composition, as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of or" consisting of 823030A ".
Sequence identity (or homology) is determined by comparing two aligned sequences along a predetermined comparison window (which may be 50%, 60%, 70%, 80%, 90%, 95% or 100% of the length of the reference nucleotide sequence or protein) and determining the number of positions at which identical residues occur. Typically, this is expressed as a percentage. The measurement of sequence identity of nucleotide sequences is a method well known to those skilled in the art.
In the present invention, dre is used 1-60 Fragment, dre 61-343 The fragments are exemplified and explained.
Wherein, dre 1-60 Fragment refers to the 1-60 position of the amino acid sequence of Dre recombinase based on SEQ ID NO. 10.
Dre 61-343 The fragment refers to the 61-343 position of the amino acid sequence of Dre recombinase based on SEQ ID NO. 10.
Dre recombinase
The Dre recombinase is derived from the D6 phage, belongs to the family of tyrosine recombinases, and has a molecular weight of about 38kDa. It can specifically recognize rox sites and catalyze the recombination of sequences between two sites, thereby causing the phenomena of DNA insertion, deletion, translocation and the like.
In the present invention, a preferred Dre recombinase is codon-optimized Dre recombinase.
Light-sensitive proteins
The photosensitive protein is mostly from animals, plants, bacteria or fungi, and can rapidly change conformation when receiving light stimulation of specific wavelength, so as to convert light into cell information.
Fusion proteins
As used herein, "fusion protein of the invention", or "polypeptide" both refer to a fusion protein according to the first aspect of the invention. The fusion protein of the invention is selected from the group consisting of: a first fusion protein, a second fusion protein, or a combination thereof;
wherein the structure of the first fusion protein is shown as the following formula I or I':
Z1-L1-A1 (I);
A1-L1-Z1 (I’);
the structure of the second fusion protein is shown as formula II or formula II':
Z2-L2-A2 (II);
A2-L2-Z2 (II’);
in the formula (I), the compound is shown in the specification,
z1 is an N-terminal fragment of Dre recombinase;
l1 is an optional first linker peptide;
a1 is a first element of a light sensitive protein;
z2 is C-terminal fragment of Dre recombinase;
a2 is a second element of the light-sensitive protein;
l2 is an optional second linker peptide;
each "-" is independently a linking peptide or non-peptide bond.
In the present invention, the length of the connecting peptide has an effect on the activity of the fusion protein, and preferably the length of the first connecting peptide and the second connecting peptide is 1 to 80aa, preferably 5 to 60aa, and more preferably 10 to 50aa.
A preferred linker peptide is shown in SEQ ID NO. 1-5.
The term "fusion protein" as used herein also includes variants as shown in any of SEQ ID No. 11-12 having the above-described activity. These variants include (but are not limited to): deletion, insertion and/or substitution of 1 to 3 (usually 1 to 2, more preferably 1) amino acids, and addition or deletion of one or several (usually up to 3, preferably up to 2, more preferably up to 1) amino acids at the C-terminal and/or N-terminal. For example, in the art, substitutions with amino acids of similar or similar properties will not generally alter the function of the protein. Also, for example, the addition or deletion of one or several amino acids at the C-terminus and/or N-terminus does not generally alter the structure and function of the protein. In addition, the term also includes monomeric and multimeric forms of the polypeptides of the invention. The term also includes linear as well as non-linear polypeptides (e.g., cyclic peptides).
The invention also includes active fragments, derivatives and analogs of the above fusion proteins. As used herein, the terms "fragment," "derivative," and "analog" refer to a polypeptide that substantially retains the function or activity of a fusion protein of the invention. The polypeptide fragment, derivative or analogue of the present invention may be (i) a polypeptide in which one or more conserved or non-conserved amino acid residues (preferably conserved amino acid residues) are substituted, or (ii) a polypeptide having a substituent group in one or more amino acid residues, or (iii) a polypeptide in which an antigenic peptide is fused to another compound (such as a compound that increases the half-life of the polypeptide, e.g., polyethylene glycol), or (iv) a polypeptide in which an additional amino acid sequence is fused to the polypeptide sequence (a fusion protein in which a tag sequence such as a leader sequence, a secretory sequence or 6His is fused). Such fragments, derivatives and analogs are within the purview of those skilled in the art in view of the teachings herein.
A preferred class of reactive derivatives refers to polypeptides formed by the replacement of up to 3, preferably up to 2, more preferably up to 1 amino acid with a qualitatively similar or analogous amino acid compared to the amino acid sequence of formula I or formula II or a combination thereof. These conservative variants are preferably produced by amino acid substitutions according to Table A.
TABLE A
Initial residue(s) Representative substitutions Preferred substitutions
Ala(A) Val;Leu;Ile Val
Arg(R) Lys;Gln;Asn Lys
Asn(N) Gln;His;Lys;Arg Gln
Asp(D) Glu Glu
Cys(C) Ser Ser
Gln(Q) Asn Asn
Glu(E) Asp Asp
Gly(G) Pro;Ala Ala
His(H) Asn;Gln;Lys;Arg Arg
Ile(I) Leu;Val;Met;Ala;Phe Leu
Leu(L) Ile;Val;Met;Ala;Phe Ile
Lys(K) Arg;Gln;Asn Arg
Met(M) Leu;Phe;Ile Leu
Phe(F) Leu;Val;Ile;Ala;Tyr Leu
Pro(P) Ala Ala
Ser(S) Thr Thr
Thr(T) Ser Ser
Trp(W) Tyr;Phe Tyr
Tyr(Y) Trp;Phe;Thr;Ser Phe
Val(V) Ile;Leu;Met;Phe;Ala Leu
The invention also provides analogs of the fusion proteins of the invention. These analogs may differ from the polypeptide of any of SEQ ID Nos. 11-12 by amino acid sequence differences, by modifications that do not affect the sequence, or by both. Analogs also include analogs having residues other than the natural L-amino acids (e.g., D-amino acids), as well as analogs having non-naturally occurring or synthetic amino acids (e.g., beta, gamma-amino acids). It is to be understood that the polypeptides of the present invention are not limited to the representative polypeptides exemplified above.
Modified (generally without altering primary structure) forms include: chemically derivatized forms of the polypeptide, such as acetylated or carboxylated, in vivo or in vitro. Modifications also include glycosylation, such as those resulting from glycosylation modifications in the synthesis and processing of the polypeptide or in further processing steps. Such modification may be accomplished by exposing the polypeptide to an enzyme that effects glycosylation, such as a mammalian glycosylase or deglycosylase. Modified forms also include sequences having phosphorylated amino acid residues (e.g., phosphotyrosine, phosphoserine, phosphothreonine). Also included are polypeptides modified to increase their resistance to proteolysis or to optimize solubility.
In a preferred embodiment, the amino acid sequence of the fusion protein of the invention is as shown in any one of SEQ ID No. 11-12.
Expression vectors and host cells
The invention also relates to vectors comprising the polynucleotides of the invention, as well as genetically engineered host cells transformed with the vectors of the invention or the coding sequences of the fusion proteins of the invention, and methods for producing the polypeptides of the invention by recombinant techniques.
The polynucleotide sequences of the present invention may be used to express or produce recombinant fusion proteins by conventional recombinant DNA techniques. Generally, the following steps are provided:
(1) Transforming or transducing a suitable host cell with a polynucleotide (or variant) of the invention encoding a fusion protein of the invention, or with a recombinant expression vector comprising the polynucleotide;
(2) A host cell cultured in a suitable medium;
(3) Isolating and purifying the protein from the culture medium or the cells.
In the present invention, the polynucleotide sequence encoding the fusion protein may be inserted into a recombinant expression vector. The term "recombinant expression vector" refers to a bacterial plasmid, bacteriophage, yeast plasmid, plant cell virus, mammalian cell virus such as adenovirus, retrovirus, or other vectors well known in the art. Any plasmid or vector may be used as long as it can replicate and is stable in the host. An important feature of expression vectors is that they typically contain an origin of replication, a promoter, a marker gene, and translation control elements.
Methods well known to those skilled in the art can be used to construct expression vectors containing a DNA sequence encoding a fusion protein of the invention and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, DNA synthesis techniques, in vivo recombinant techniques, and the like. The DNA sequence may be operably linked to a suitable promoter in an expression vector to direct mRNA synthesis. Representative examples of such promoters are: lac or trp promoter of E.coli; a lambda phage PL promoter; eukaryotic promoters include CMV immediate early promoter, HSV thymidine kinase promoter, early and late SV40 promoter, LTRs of retrovirus, and other known promoters which can control the expression of genes in prokaryotic or eukaryotic cells or viruses. The expression vector also includes a ribosome binding site for translation initiation and a transcription terminator.
In addition, the expression vector preferably contains one or more selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase, neomycin resistance, and Green Fluorescent Protein (GFP) for eukaryotic cell culture, or tetracycline or ampicillin resistance for E.coli.
Vectors comprising the appropriate DNA sequences described above, together with appropriate promoter or control sequences, may be used to transform appropriate host cells to enable expression of the protein.
The host cell may be a prokaryotic cell (e.g., E.coli), or a lower eukaryotic cell, or a higher eukaryotic cell, such as a yeast cell, a plant cell, or a mammalian cell (including human and non-human mammals). Representative examples are: escherichia coli, wheat germ cells, insect cells, SF9, hela, HEK293, CHO, yeast cells, etc. In a preferred embodiment of the present invention, a yeast cell (e.g., pichia pastoris, kluyveromyces lactis, or a combination thereof; preferably, the yeast cell comprises Kluyveromyces lactis, more preferably Kluyveromyces marxianus, and/or Kluyveromyces lactis) is selected as the host cell.
When the polynucleotide of the present invention is expressed in higher eukaryotic cells, transcription will be enhanced if an enhancer sequence is inserted into the vector. Enhancers are cis-acting elements of DNA, usually about 10 to 300 bp in length, that act on a promoter to increase gene transcription. Examples include the SV40 enhancer on the late side of the replication origin at 100 to 270 bp, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
It will be clear to one of ordinary skill in the art how to select appropriate vectors, promoters, enhancers and host cells.
Transformation of a host cell with recombinant DNA can be carried out using conventional techniques well known to those skilled in the art. When the host is prokaryotic, e.g., E.coli, competent cells capable of DNA uptake can be harvested after exponential growth phase using CaCl 2 Methods of treatment, the steps used are well known in the art. Another method is to use MgCl 2 . If desired, transformation can also be carried out by electroporation. When the host is a eukaryote, the following DNA transfection methods may be used: calcium phosphate coprecipitation, conventional mechanical methods such as microinjection, electroporation, liposome encapsulation, etc.
The obtained transformant can be cultured by a conventional method to express the polypeptide encoded by the gene of the present invention. The medium used in the culture may be selected from various conventional media depending on the host cell used. The culturing is performed under conditions suitable for growth of the host cell. After the host cells have been grown to an appropriate cell density, the selected promoter is induced by suitable means (e.g., temperature shift or chemical induction) and the cells are cultured for an additional period of time.
The recombinant polypeptide in the above method may be expressed intracellularly or on the cell membrane, or secreted extracellularly. If necessary, the recombinant protein can be isolated and purified by various separation methods using its physical, chemical and other properties. These methods are well known to those skilled in the art. Examples of such methods include, but are not limited to: conventional renaturation treatment, treatment with a protein precipitant (such as salt precipitation), centrifugation, cell lysis by osmosis, sonication, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, high Performance Liquid Chromatography (HPLC), and other various liquid chromatography techniques, and combinations thereof.
The main advantages of the invention include:
(1) The invention discovers for the first time that the complete Dre recombinase is split into an N-terminal segment and a C-terminal segment, and the N-terminal segment and the C-terminal segment are respectively fused and expressed with photosensitive proteins nMAG and pMag through a linker to form a light-activated Dre recombinase system, also called PA-Dre. In the absence of blue light, the segmented Dre recombinase has no catalytic activity, and when the system is stimulated by blue light, the segmented Dre recombinase is quickly dimerized under the drive of photosensitive protein to form the recombinase with catalytic activity, so that the activity is high, the background is few, and the induction multiple is up to 102 times.
(2) Compared with the traditional induction system such as a Dre recombinase induced by tamoxifen, the PA-Dre system not only does not influence the endogenous signal path of cells, but also has higher space-time specificity, can be applied to multiple fields such as lineage tracing, cell clearing and the like, combines the specificity of a promoter, and realizes more accurate gene operation by using local blue light irradiation.
(3) The PA-Dre system established by the invention can realize space-time specific gene operation by controlling the time and space of illumination, makes up the defects of the traditional induction system and provides a more appropriate tool for subsequent research.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally following conventional conditions, such as Sambrook et al, molecular cloning: the conditions described in the Laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer's recommendations. Unless otherwise indicated, percentages and parts are percentages and parts by weight.
Unless otherwise specified, the materials and reagents used in the examples of the present invention are commercially available products.
Example 1 determination of the segmental sites of the Dre recombinase
First, 23 potential splitting sites are selected (table 1), and the N-terminal and the C-terminal of Dre recombinase are respectively connected with two interacting proteins Cho2 and Docs through a linker to form a fusion protein. Three plasmids were co-transfected into HEK293 cells using CAG-rox-stop-rox-ZsGreen as the reporter plasmid, and 11 of these cleavage sites were found to be working (fig. 1). Then N end and C end of Dre recombinase formed by the 11 splitting sites are respectively fused and expressed with blue light-sensitive proteins pMag and nMAG, CMV-rox-stop-rox-LUC is taken as a report plasmid, three plasmids are co-transferred into HEK293 cells, and the activity of the segmented Dre recombinase formed by 3 splitting sites (R60/Q61, L150/L151 and S246/N247) is found to be dependent on blue light regulation, and the three sites are taken as candidate sites of a PA-Dre system. (FIG. 2).
Figure BDA0002169935480000141
Figure BDA0002169935480000151
Example 2 construction and optimization of PA-Dre System
(1) Adjusting the relative direction
In order to adjust the relative positions of the light sensitive protein and the segmented Dre recombinase, 4 different plasmids were constructed for the three segmented sites, and the plasmids were co-transformed into HEK293 cells by combining a + B, a + D, C + B, and C + D, and a + D was found to be the optimal configuration, when the activity of the segmented Dre recombinase was the highest (fig. 3). Considering that the enzyme activity of the R60/Q61 segmented site is low, the subsequent experiments are optimized on two segmented sites of L150/L151 and S246/N247.
(2) Change different linker
In the process of constructing fusion proteins, linker is important because it is involved in the activity of the protein. We selected 5 different linkers (table 2) and verified that L4 was most effective on HEK293 cells (fig. 4). When the segmented Dre recombinase and the photosensitive protein are connected by the L4, the enzyme activity is higher and the background is greatly reduced. The two segmented Dre recombinase induction folds of L150/L151 and S246/N247 are similar, but the S246/N247 enzyme activity is higher, so this site is taken as the final selection.
Figure BDA0002169935480000152
(3) Changing different NLS
To increase the activity of the segmented Dre recombinase, we selected 4 different nuclear localization signal sequences (table 3) that were linked to the C-terminus of the segmented Dre recombinase, respectively. The best effect of NLP was confirmed on HEK293 cells, and the induction factor of the segmented Dre recombinase was increased from the initial 26-fold to 70-fold (FIG. 5).
Figure BDA0002169935480000161
(4) Construction of Single plasmid System
In order to make the segmented Dre recombinase system more stable and efficient, we used the self-hydrolyzed peptide T2A to fuse and express the C-terminal and the N-terminal of the segmented Dre to form a single plasmid system, and the induction fold of Dre recombinase was increased by about 30-fold (fig. 6). We define this final single particle as PA-Dre (FIG. 7).
Example 3 illustrates the features of the PA-Dre System
(1) The PA-Dre system has illumination time dependency
The PA-Dre and CMV-rox-stop-rox-LUC reporter plasmids were co-transferred into HEK293 cells. After 24 hours, one group was dark and protected from light, and the other group was used at an intensity of 2mw/cm 2 Blue light (30 s on, 3min off), illuminate 4h, 8h, 16h, 24h, 32h, 36h and 48h, respectively. The enzyme activity of the LUC is detected after 48h, and the PA-Dre system is found to have illumination time dependency, and the enzyme activity of the LUC is higher along with the increase of illumination time (figure 8).
(2) The PA-Dre system has the dependence of illumination intensity
The PA-Dre and CMV-rox-stop-rox-LUC reporter plasmids were co-transfected into HEK293 cells. After 24 hours, one group was dark and protected from light, and the other group was irradiated at an intensity of 4mw/cm, respectively 2 、2mw/cm 2 、1mw/cm 2 、0.5mw/cm 2 And 0.05mw/cm 2 Blue light (30 s on, 3min off). The enzyme activity of the LUC is detected after 48h, and the PA-Dre system is found to have illumination intensity dependency, and the enzyme activity of the LUC is higher along with the increase of illumination intensity (figure 9).
(3) The PA-Dre system is adapted to different promoters
To verify whether the PA-Dre system is compatible with different promoters, we constructed plasmids driving expression by EF 1. Alpha., CMV, and PGK. These were separately co-transfected in equimolar amounts with the CMV-rox-stop-rox-LUC reporter plasmid into HEK293 cells. After 24 hours, one group was dark and protected from light, and the other group was used at an intensity of 2mw/cm 2 Blue light (30 s on, 3min off) illumination. After 48h, the enzyme activity of LUC was tested, and the PA-Dre system was found to be adapted to different promoters, among which CAG promoter is expressed most strongly (FIG. 10).
(3) The PA-Dre System is adapted to different cell lines
To verify whether the PA-Dre system is compatible with different cell lines, we performed the validation on HEK-293A, hela, hep2 and SH-SY5Y cells, respectively. The results indicate that the PA-Dre system is adapted to different cell lines, probably affected by transfection efficiency, and works best on HEK-293 cells (FIG. 11).
Example 4 validation of PA-Dre System in WT mice
(1) Injection of Low dose PA-Dre plasmid
We used the hydrodynamic tail vein injection of plasmid to co-inject low doses of PA-Dre and CMV-rox-stop-rox-LUC reporter plasmid into mice. After 8h, one group was dark and protected from light, and the other group was used at an intensity of 20mw/cm 2 Blue light (1 min on, 3min off), 0.5h, 1h, 2h and 4h, respectively. After 24h, live images were taken and the results showed that the PA-Dre system was also light time dependent in mice when low doses of PA-Dre plasmid were injected and that the catalytic efficiency of the recombinant enzyme was highest at 4h (FIG. 12).
(2) Injection of high dose of PA-Dre plasmid
High doses of PA-Dre and CMV-rox-stop-rox-LUC reporter plasmid were co-injected into mice. After 8h, one group was dark and protected from light, and the other group was used at an intensity of 20mw/cm 2 Blue light (1 min on, 3min off) for 1min. After 24h, in vivo imaging was taken and the results showed that PA-Dre had higher catalytic efficiency when injected with high doses of PA-Dre plasmid, even with short exposure times (FIG. 11). The liver of the mouse was taken and imaged in vivo and the LUC signal was found to be indeed very strong, demonstrating that the PA-Dre system works well not only at the cellular level but also in animals (FIG. 13).
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes or modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the appended claims of the present application.
Sequence listing
<110> university of east China
Shanghai Bangyao Biological Technology Co.,Ltd.
<120> Dre recombination System with light-induced activation
<130> P2019-1015
<160> 14
<170> SIPOSequenceListing 1.0
<210> 1
<211> 15
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 1
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Arg
1 5 10 15
<210> 2
<211> 16
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 2
Leu Glu Ala Ser Pro Ser Asn Pro Gly Ala Ser Asn Gly Ser Gly Thr
1 5 10 15
<210> 3
<211> 15
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 3
Glu Ala Ala Ala Lys Glu Ala Ala Ala Lys Glu Ala Ala Ala Lys
1 5 10 15
<210> 4
<211> 32
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 4
Ala Pro Thr Leu Ala Asp Leu Ala Val Asp Leu Ala Ala Leu Arg Pro
1 5 10 15
Leu Glu His Pro Asn Pro Pro Leu Gln Arg Ala Ala Glu Ala Leu Leu
20 25 30
<210> 5
<211> 16
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 5
Val Asp Leu Ala Ala Leu Arg Pro Leu Glu His Pro Asn Pro Pro Leu
1 5 10 15
<210> 6
<211> 7
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 6
Pro Lys Lys Lys Arg Lys Val
1 5
<210> 7
<211> 25
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 7
Met Ser Arg Arg Arg Lys Ala Asn Pro Thr Lys Leu Ser Glu Asn Ala
1 5 10 15
Lys Lys Leu Ala Lys Glu Val Glu Asn
20 25
<210> 8
<211> 20
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 8
Ala Val Lys Arg Pro Ala Ala Thr Lys Lys Ala Gly Gln Ala Lys Lys
1 5 10 15
Lys Lys Leu Asp
20
<210> 9
<211> 9
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 9
Pro Ala Ala Lys Arg Val Lys Leu Asp
1 5
<210> 10
<211> 342
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 10
Met Ser Glu Leu Ile Ile Ser Gly Ser Ser Gly Gly Phe Leu Arg Asn
1 5 10 15
Ile Gly Lys Glu Tyr Gln Glu Ala Ala Glu Asn Phe Met Arg Phe Met
20 25 30
Asn Asp Gln Gly Ala Tyr Ala Pro Asn Thr Leu Arg Asp Leu Arg Leu
35 40 45
Val Phe His Ser Trp Ala Arg Trp Cys His Ala Arg Gln Leu Ala Trp
50 55 60
Phe Pro Ile Ser Pro Glu Met Ala Arg Glu Tyr Phe Leu Gln Leu His
65 70 75 80
Asp Ala Asp Leu Ala Ser Thr Thr Ile Asp Lys His Tyr Ala Met Leu
85 90 95
Asn Met Leu Leu Ser His Cys Gly Leu Pro Pro Leu Ser Asp Asp Lys
100 105 110
Ser Val Ser Leu Ala Met Arg Arg Ile Arg Arg Glu Ala Ala Thr Glu
115 120 125
Lys Gly Glu Arg Thr Gly Gln Ala Ile Pro Leu Arg Trp Asp Asp Leu
130 135 140
Lys Leu Leu Asp Val Leu Leu Ser Arg Ser Glu Arg Leu Val Asp Leu
145 150 155 160
Arg Asn Arg Ala Phe Leu Phe Val Ala Tyr Asn Thr Leu Met Arg Met
165 170 175
Ser Glu Ile Ser Arg Ile Arg Val Gly Asp Leu Asp Gln Thr Gly Asp
180 185 190
Thr Val Thr Leu His Ile Ser His Thr Lys Thr Ile Thr Thr Ala Ala
195 200 205
Gly Leu Asp Lys Val Leu Ser Arg Arg Thr Thr Ala Val Leu Asn Asp
210 215 220
Trp Leu Asp Val Ser Gly Leu Arg Glu His Pro Asp Ala Val Leu Phe
225 230 235 240
Pro Pro Ile His Arg Ser Asn Lys Ala Arg Ile Thr Thr Thr Pro Leu
245 250 255
Thr Ala Pro Ala Met Glu Lys Ile Phe Ser Asp Ala Trp Val Leu Leu
260 265 270
Asn Lys Arg Asp Ala Thr Pro Asn Lys Gly Arg Tyr Arg Thr Trp Thr
275 280 285
Gly His Ser Ala Arg Val Gly Ala Ala Ile Asp Met Ala Glu Lys Gln
290 295 300
Val Ser Met Val Glu Ile Met Gln Glu Gly Thr Trp Lys Lys Pro Glu
305 310 315 320
Thr Leu Met Arg Tyr Leu Arg Arg Gly Gly Val Ser Val Gly Ala Asn
325 330 335
Ser Arg Leu Met Asp Ser
340
<210> 11
<211> 428
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 11
Met Ser Glu Leu Ile Ile Ser Gly Ser Ser Gly Gly Phe Leu Arg Asn
1 5 10 15
Ile Gly Lys Glu Tyr Gln Glu Ala Ala Glu Asn Phe Met Arg Phe Met
20 25 30
Asn Asp Gln Gly Ala Tyr Ala Pro Asn Thr Leu Arg Asp Leu Arg Leu
35 40 45
Val Phe His Ser Trp Ala Arg Trp Cys His Ala Arg Gln Leu Ala Trp
50 55 60
Phe Pro Ile Ser Pro Glu Met Ala Arg Glu Tyr Phe Leu Gln Leu His
65 70 75 80
Asp Ala Asp Leu Ala Ser Thr Thr Ile Asp Lys His Tyr Ala Met Leu
85 90 95
Asn Met Leu Leu Ser His Cys Gly Leu Pro Pro Leu Ser Asp Asp Lys
100 105 110
Ser Val Ser Leu Ala Met Arg Arg Ile Arg Arg Glu Ala Ala Thr Glu
115 120 125
Lys Gly Glu Arg Thr Gly Gln Ala Ile Pro Leu Arg Trp Asp Asp Leu
130 135 140
Lys Leu Leu Asp Val Leu Leu Ser Arg Ser Glu Arg Leu Val Asp Leu
145 150 155 160
Arg Asn Arg Ala Phe Leu Phe Val Ala Tyr Asn Thr Leu Met Arg Met
165 170 175
Ser Glu Ile Ser Arg Ile Arg Val Gly Asp Leu Asp Gln Thr Gly Asp
180 185 190
Thr Val Thr Leu His Ile Ser His Thr Lys Thr Ile Thr Thr Ala Ala
195 200 205
Gly Leu Asp Lys Val Leu Ser Arg Arg Thr Thr Ala Val Leu Asn Asp
210 215 220
Trp Leu Asp Val Ser Gly Leu Arg Glu His Pro Asp Ala Val Leu Phe
225 230 235 240
Pro Pro Ile His Arg Ser Ala Pro Thr Leu Ala Asp Leu Ala Val Asp
245 250 255
Leu Ala Ala Leu Arg Pro Leu Glu His Pro Asn Pro Pro Leu Gln Arg
260 265 270
Ala Ala Glu Ala Leu Leu His Thr Leu Tyr Ala Pro Gly Gly Tyr Asp
275 280 285
Ile Met Gly Tyr Leu Asp Gln Ile Gly Asn Arg Pro Asn Pro Gln Val
290 295 300
Glu Leu Gly Pro Val Asp Thr Ser Cys Ala Leu Ile Leu Cys Asp Leu
305 310 315 320
Lys Gln Lys Asp Thr Pro Ile Val Tyr Ala Ser Glu Ala Phe Leu Tyr
325 330 335
Met Thr Gly Tyr Ser Asn Ala Glu Val Leu Gly Arg Asn Cys Arg Phe
340 345 350
Leu Gln Ser Pro Asp Gly Met Val Lys Pro Lys Ser Thr Arg Lys Tyr
355 360 365
Val Asp Ser Asn Thr Ile Asn Thr Met Arg Lys Ala Ile Asp Arg Asn
370 375 380
Ala Glu Val Gln Val Glu Val Val Asn Phe Lys Lys Asn Gly Gln Arg
385 390 395 400
Phe Val Asn Phe Leu Thr Met Ile Pro Val Arg Asp Glu Thr Gly Glu
405 410 415
Tyr Arg Tyr Ser Met Gly Phe Gln Cys Glu Thr Glu
420 425
<210> 12
<211> 279
<212> PRT
<213> Artificial sequence (artificial sequence)
<400> 12
Met His Thr Leu Tyr Ala Pro Gly Gly Tyr Asp Ile Met Gly Tyr Leu
1 5 10 15
Arg Gln Ile Arg Asn Arg Pro Asn Pro Gln Val Glu Leu Gly Pro Val
20 25 30
Asp Thr Ser Cys Ala Leu Ile Leu Cys Asp Leu Lys Gln Lys Asp Thr
35 40 45
Pro Ile Val Tyr Ala Ser Glu Ala Phe Leu Tyr Met Thr Gly Tyr Ser
50 55 60
Asn Ala Glu Val Leu Gly Arg Asn Cys Arg Phe Leu Gln Ser Pro Asp
65 70 75 80
Gly Met Val Lys Pro Lys Ser Thr Arg Lys Tyr Val Asp Ser Asn Thr
85 90 95
Ile Asn Thr Met Arg Lys Ala Ile Asp Arg Asn Ala Glu Val Gln Val
100 105 110
Glu Val Val Asn Phe Lys Lys Asn Gly Gln Arg Phe Val Asn Phe Leu
115 120 125
Thr Met Ile Pro Val Arg Asp Glu Thr Gly Glu Tyr Arg Tyr Ser Met
130 135 140
Gly Phe Gln Cys Glu Thr Glu Ala Pro Thr Leu Ala Asp Leu Ala Val
145 150 155 160
Asp Leu Ala Ala Leu Arg Pro Leu Glu His Pro Asn Pro Pro Leu Gln
165 170 175
Arg Ala Ala Glu Ala Leu Leu Asn Lys Ala Arg Ile Thr Thr Thr Pro
180 185 190
Leu Thr Ala Pro Ala Met Glu Lys Ile Phe Ser Asp Ala Trp Val Leu
195 200 205
Leu Asn Lys Arg Asp Ala Thr Pro Asn Lys Gly Arg Tyr Arg Thr Trp
210 215 220
Thr Gly His Ser Ala Arg Val Gly Ala Ala Ile Asp Met Ala Glu Lys
225 230 235 240
Gln Val Ser Met Val Glu Ile Met Gln Glu Gly Thr Trp Lys Lys Pro
245 250 255
Glu Thr Leu Met Arg Tyr Leu Arg Arg Gly Gly Val Ser Val Gly Ala
260 265 270
Asn Ser Arg Leu Met Asp Ser
275
<210> 13
<211> 1284
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 13
atgagcgagc tgatcatctc tggctcctct ggaggattcc tgaggaacat cggcaaggag 60
taccaggagg ctgctgagaa cttcatgaga ttcatgaatg accagggagc ctacgcccct 120
aacaccctga gagacctgag gctggtgttc cactcctggg ctagatggtg ccacgctaga 180
cagctggcct ggttccctat ctctcctgag atggctaggg agtacttcct tcagctgcac 240
gatgctgacc tggcctctac caccatcgac aagcactacg ccatgctgaa catgctgctg 300
tcccactgtg gcctgcctcc tctgtctgat gacaagtctg tgagcctggc catgaggaga 360
atccggagag aggctgccac cgagaaggga gagagaaccg gccaggccat ccctctgaga 420
tgggatgacc tgaagctgct ggatgtgctg ctgtctagat ctgagagact ggtggacctg 480
aggaataggg ccttcctgtt tgtggcctac aacaccctga tgaggatgtc tgagatctct 540
aggatcagag tgggagacct ggaccagacc ggagacaccg tgaccctgca catctcccac 600
accaagacca tcaccaccgc tgctggcctg gacaaagtgc tgtctaggag gaccaccgct 660
gtgctgaatg actggctgga tgtgtctggc ctgagagagc accctgacgc tgtgctgttc 720
cctcctatcc accggagcgc accgactctg gcggatctag cggtggatct agcggctctg 780
agacccctgg aacatccgaa tccgccactc cagagagcag cggaggctct tctgcatact 840
ctttatgccc ccggtggata tgacattatg ggatatctgg accagatcgg caaccggcca 900
aacccgcagg tggaactggg ccccgtggat acatcctgcg ccttgattct ttgtgacctg 960
aaacagaaag acaccccgat agtttacgcg agtgaagcct tcctctacat gacaggttac 1020
agcaacgcag aggtgctggg ccggaattgc cggtttctgc aaagccctga cggcatggtg 1080
aagcccaaga gcacccggaa gtacgtggat agtaacacaa tcaatactat gcgcaaggca 1140
atcgacagga atgccgaggt gcaggttgaa gtagtcaatt ttaaaaagaa tggacagcga 1200
tttgttaatt tcctgactat gatacctgtt agggacgaaa caggcgagta tcgatactct 1260
atgggattcc agtgcgaaac agaa 1284
<210> 14
<211> 837
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 14
atgcatactc tttatgcccc cggtggatat gacattatgg gatatctgag gcagatcagg 60
aaccggccaa acccgcaggt ggaactgggc cccgtggata catcctgcgc cttgattctt 120
tgtgacctga aacagaaaga caccccgata gtttacgcga gtgaagcctt cctctacatg 180
acaggttaca gcaacgcaga ggtgctgggc cggaattgcc ggtttctgca aagccctgac 240
ggcatggtga agcccaagag cacccggaag tacgtggata gtaacacaat caatactatg 300
cgcaaggcaa tcgacaggaa tgccgaggtg caggttgaag tagtcaattt taaaaagaat 360
ggacagcgat ttgttaattt cctgactatg atacctgtta gggacgaaac aggcgagtat 420
cgatactcta tgggattcca gtgcgaaaca gaagcaccga ctctggcgga tctagcggtg 480
gatctagcgg ctctgagacc cctggaacat ccgaatccgc cactccagag agcagcggag 540
gctcttctga acaaggctag gatcaccacc acccctctga ccgcccctgc catggagaag 600
atttttagcg atgcctgggt gctgctgaac aagagggatg ccacccctaa caagggccgc 660
taccggacct ggaccggcca ctctgctaga gtgggagctg ccatcgacat ggctgagaag 720
caagtgtcca tggtggagat catgcaggag ggcacctgga aaaagcctga gacactgatg 780
agatacctga ggaggggagg agtgtctgtg ggagccaact ctaggctgat ggactcc 837

Claims (14)

1. A fusion protein, wherein the fusion protein is a combination of a first fusion protein and a second fusion protein; wherein the first fusion protein has a structure shown in formula I or formula I':
Z1-L1-A1 (I);
A1-L1-Z1 (I’);
the structure of the second fusion protein is shown as formula II or formula II':
Z2-L2-A2 (II);
A2-L2-Z2(II’);
in the formula (I), the compound is shown in the specification,
z1 is an N-terminal fragment of Dre recombinase;
l1 is an optional first linker peptide;
a1 is a first element of a light sensitive protein;
z2 is a C-terminal fragment of Dre recombinase;
a2 is a second element of the light sensitive protein;
l2 is an optional second linking peptide;
each "-" is independently a linking peptide or a peptide or non-peptide bond;
the N-terminal fragment of the Dre recombinase is based on SEQ ID No.:10, the C-terminal fragment of the Dre recombinase is based on SEQ ID No.:10, and the N-terminal fragment of the Dre recombinase is Dre 1-60 A fragment, wherein the C-terminal fragment of the Dre recombinase is Dre 61-342 A fragment; or the N-terminal fragment of the Dre recombinase is Dre 1-150 A fragment, wherein the C-terminal fragment of the Dre recombinase is Dre 151-342 A fragment; or the N-terminal fragment of the Dre recombinase is Dre 1-246 A fragment, wherein the C-terminal fragment of the Dre recombinase is Dre 247-342 And (3) fragment.
2. The fusion protein of claim 1, wherein the fusion protein further comprises a nuclear localization signal.
3. The fusion protein of claim 2, wherein the nuclear localization signal is selected from the group consisting of: NLS sv40 、NLS EGL-13 、NLS NLP 、NLS c-Myc 、PY-NLSs、snRNPs、truncated NLS sv40 、NLS TUS Or a combination thereof.
4. The fusion protein of claim 1, wherein the fusion protein has an amino acid sequence as set forth in any one of SEQ ID No. 11-12.
5. A polynucleotide encoding the fusion protein of claim 1.
6. A vector comprising the polynucleotide of claim 5.
7. A host cell comprising the vector of claim 6, or having the polynucleotide of claim 5 integrated into its genome.
8. A method of producing the fusion protein of claim 1, comprising the steps of:
culturing the host cell of claim 7 under conditions suitable for expression, thereby expressing the fusion protein; and/or isolating the fusion protein.
9. A gene editing reagent comprising the fusion protein of claim 1.
10. A kit comprising the gene editing reagent according to claim 9.
11. Use of the fusion protein of claim 1 for the preparation of a reagent or kit for increasing gene editing efficiency.
12. A method of performing gene editing comprising the steps of:
(a) Providing a first fusion protein and a second fusion protein, wherein the structures of the first fusion protein and the second fusion protein are as set forth in claim 1;
(b) Under illumination, the first fusion protein and the second fusion protein form a complex, thereby performing gene editing in the presence of the complex.
13. The method of claim 12, wherein the illumination intensity is from 0.1 to 20mw/cm 2
14. The method of claim 12, wherein the gene editing is performed in a cell in vitro.
CN201910759889.4A 2019-08-16 2019-08-16 Dre recombination system activated by light induction Active CN112391366B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910759889.4A CN112391366B (en) 2019-08-16 2019-08-16 Dre recombination system activated by light induction

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910759889.4A CN112391366B (en) 2019-08-16 2019-08-16 Dre recombination system activated by light induction

Publications (2)

Publication Number Publication Date
CN112391366A CN112391366A (en) 2021-02-23
CN112391366B true CN112391366B (en) 2022-10-14

Family

ID=74602950

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910759889.4A Active CN112391366B (en) 2019-08-16 2019-08-16 Dre recombination system activated by light induction

Country Status (1)

Country Link
CN (1) CN112391366B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107779462A (en) * 2016-08-29 2018-03-09 中国科学院上海生命科学研究院 Double homologous recombination pedigree tracer techniques
CN108070035A (en) * 2017-10-12 2018-05-25 中国科学院上海生命科学研究院 Inducibility Genetic Recombination enzyme system CrexER

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7422889B2 (en) * 2004-10-29 2008-09-09 Stowers Institute For Medical Research Dre recombinase and recombinase systems employing Dre recombinase
DK1815001T3 (en) * 2004-11-26 2011-05-16 Helmholtz Zentrum Muenchen Fallback cartridges for random and targeted conditioned gene inactivation
WO2018111838A1 (en) * 2016-12-12 2018-06-21 Trustees Of Boston University Inducible dimerization of recombinases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107779462A (en) * 2016-08-29 2018-03-09 中国科学院上海生命科学研究院 Double homologous recombination pedigree tracer techniques
CN108070035A (en) * 2017-10-12 2018-05-25 中国科学院上海生命科学研究院 Inducibility Genetic Recombination enzyme system CrexER

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
A photoactivatable Cre-loxP recombination system for optogenetic genome engineering;Fuun Kawano等;《Nat Chem Biol》;20161231;第12卷(第12期);第1059-1064页 *
A practical summary of site-specific recombination, conditional mutagenesis, and tamoxifen induction of CreERT2;Konstantinos Anastassiadis等;《Methods Enzymol》;20101231;第477卷;第109-123页 *
Binary recombinase systems for high-resolution conditional mutagenesis;Mario Hermann 等;《Nucleic Acids Res》;20140430;第42卷(第6期);第3894-3907页 *
Rapid blue light induction of protein interactions in living cells;Matthew J. Kennedy等;《Nat Methods》;20101231;第7卷(第12期);第973-975页 *
基于T7 RNA聚合酶构建光诱导的基因开关;韩悌云等;《中国生物工程学会第二届青年科技论坛暨首届青年工作委员会学术年会论文集》;20171231;第105页 *

Also Published As

Publication number Publication date
CN112391366A (en) 2021-02-23

Similar Documents

Publication Publication Date Title
Caterina et al. Cloning and functional characterization of LCR-F1: a bZIP transcription factor that activates erythroid-specific, human globin gene expression
JPH08506586A (en) Stable protein product with improved bactericidal / penetration and pharmaceutical composition containing the same
US10669531B2 (en) Cell-permeable Cre (iCP-Cre) recombinant protein and use thereof
NO331814B1 (en) Purified or Isolated Nucleic Acid, Isolated Polypeptide, Cloning and / or Expression Vector comprising the Nucleic Acid, Host Cell, Differentiated ES Cell, Animals comprising the Cells, Use of the Nucleic Acid for Displacement and / or Amplification of Nucleic Acid Sequences, as Sens or Antisense Oligonucleotide Oligonucleotide polypeptide, monoclonal or polyclonal antibody that selectively binds the polypeptide, method for assaying the polypeptide, assay for use in the method, assay for pluripotent nature of ES bird cell, bird classification method, DNA chip, protein chip method the polypeptide in food sample, method for analyzing whether compound or medium can induce differentiation of pluripotent cell, or restore pluripotent nature, and use of the nucleic acid that promotes gene for expression of the gene in pluripotent bird cell.
CN101484462A (en) Chimeric zinc finger recombinases optimized for catalysis by directed evolution
JPH09505474A (en) Translation enhancer DNA
US8993742B2 (en) Tubulo-vesicular structure localization signals
KR102584789B1 (en) ADH protein family mutations and their uses
US20040146889A1 (en) Inducible regulatory system and use thereof
TR201816314T4 (en) Expression vectors containing the chimeric cytomegalovirus promoter and enhancer sequences.
US20220195416A1 (en) Rna site-directed editing using artificially constructed rna editing enzymes and related uses
WO2003076561A2 (en) Recombinase fusion protein with enhanced cellular uptake
CN110996658A (en) Non-human animals comprising a humanized ASGR1 locus
CN112391366B (en) Dre recombination system activated by light induction
WO2021110119A1 (en) Highly active transposase and application thereof
WO2020235692A1 (en) Mass production system of recombinant bagworm silk thread protein
CN103361342B (en) A kind of method and its application of transgenosis positioning integration
CN106978416B (en) Gene positioning integration expression system and application thereof
CN109971729B (en) Enzyme composition
CN111088251A (en) Gene expression cassette and application thereof in Cre-lox recombination efficiency detection
CN112921052B (en) In vivo cell proliferation marker and tracer system and application thereof
US9611486B2 (en) Constructs and method for regulating gene expression or for detecting and controlling a DNA locus in eukaryotes
KR102195740B1 (en) Cell-penetrating peptide dimers, method for preparing the same, and cargo delivery system using the same
JP2003518947A (en) Transduction of recombinase for inducible gene targeting
JP4811765B2 (en) An expression vector that can control the inducible expression of foreign genes

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