[go: up one dir, main page]

WO2018121602A1 - Protein function switch system controlled by small molecule drug - Google Patents

Protein function switch system controlled by small molecule drug Download PDF

Info

Publication number
WO2018121602A1
WO2018121602A1 PCT/CN2017/118970 CN2017118970W WO2018121602A1 WO 2018121602 A1 WO2018121602 A1 WO 2018121602A1 CN 2017118970 W CN2017118970 W CN 2017118970W WO 2018121602 A1 WO2018121602 A1 WO 2018121602A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
protease
switch system
protein function
target protein
Prior art date
Application number
PCT/CN2017/118970
Other languages
French (fr)
Chinese (zh)
Inventor
邹庆剑
翟珍珍
高斌
Original Assignee
天津天锐生物科技有限公司
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 天津天锐生物科技有限公司 filed Critical 天津天锐生物科技有限公司
Publication of WO2018121602A1 publication Critical patent/WO2018121602A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • 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
    • 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
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/503Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from viruses
    • C12N9/506Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from viruses derived from RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/95Fusion polypeptide containing a motif/fusion for degradation (ubiquitin fusions, PEST sequence)
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Definitions

  • the invention relates to the field of biotechnology, in particular to a protein function switching system controlled by a small molecule drug.
  • protein function regulation methods are mainly divided into three categories: 1 using small molecules or light to regulate the binding or dissociation of two heterodimers; 2 using small molecules to regulate protein structure changes; 3 using small molecules to regulate protein stability Sex.
  • the techniques involved in the regulation of each type of protein function are described below:
  • CID chemical inducers of dimerization
  • FK506 the immunosuppressive drug FK506.
  • FKBP12 FK506 binding protein
  • calcineurin FK506 binding protein
  • FK1012 FK506 binding protein
  • rapamycin a macrocyclic natural product that mediates the interaction between FKBP12 and the FRB domain of mTOR (Brown et al., 1994). If the two proteins of interest are fused to FKBP and FRB, respectively, they can be close to each other in the presence of rapamycin, and rapamycin can rapidly achieve FKBP-FRB binding at low doses. However, cellular endogenous FKBP and mTOR compete with FKBP and FRB fusion proteins for binding to rapamycin, resulting in ineffective interactions. Binding of rapamycin to mTOR also leads to cell cycle arrest (Haruki et al., 2008).
  • the optimized method to obtain Rimiducid-regulated FKBP-FRB dimerization can effectively reduce the CID off-target effect by site-directed mutagenesis of rapamycin derivatization and FKBP or FRB domain (Patent No.: US2015/065629) .
  • the dexamethasone-methotrexate (Dex-Mtx) complex induces an interaction between the glucocorticoid receptor (GR) and dihydrofolate reductase (DHFR).
  • GR glucocorticoid receptor
  • DHFR dihydrofolate reductase
  • Mtx is an inhibitor of DHFR, which limits its use.
  • trimethoprim-SLF (TMP-SLF) compound triggers the interaction between the DHFR-FKBP12 fusions (Czlapinski et al., 2008). This compound does not bind endogenous proteins, but these small molecules are expensive and require multiple steps of synthesis, which limits their widespread use.
  • Abscisic acid is one of the hormones that stimulate plant development. It inhibits protein phosphatase (PP2Cs) by binding to pyribactin resistance (PYR)/PYR1-like (PYL) (Cutler et al., 2010; patent number: US2012043121). After merging the PYL and PP2C domains with the DNA binding domain Gal4 and the transcriptional activation domain VP64, respectively, Gal4 transcriptional activity can be reconstituted.
  • Another phytohormone, gibberellin (GA3) can also be used as CID. GA3 binds to GID1 and promotes its interaction with GAI (Hirano et al., 2008; Miyamoto et al., 2012). Plant hormones are relatively economical, safe and responsive, making them useful.
  • the protein of interest is split into two inactive fragments, and small molecules of specific compounds can induce association of these fragments, thereby restoring the structure and function of the target protein. This process is also called fragment complementation.
  • Muir and colleagues developed an intein-based conditional protein splicing system (Mootz and Muir, 2002). The intein is divided into N- and C-terminal halves. When present alone, both inteins are inactive, but once they are close to each other, an active domain is formed, and the proteins at both ends are spliced together to form an activity. protein. For example, two former fusions are between FKBP and maltose binding protein (MBP), which is fused between the FRB and His tags.
  • MBP maltose binding protein
  • fusions dimerize under rapamycin induction and the intein domain is close, which restores intein splicing activity and leads to the formation of His-tagged MBP.
  • Intein splicing is an irreversible process, and the major disadvantage of intein-based fragment-complementing systems is the lack of reversibility.
  • conditional protein degradation is accomplished using the ubiquitin-proteasome system, so the regulation of protein stability can be achieved by using small molecules to regulate various aspects of protein degradation.
  • Degradants degrons
  • Most degradation-mediated degradation is accomplished by the ubiquitin-proteasome degradation pathway, or by autophagy.
  • a number of degrading sequences have been identified, for example: ddFKBP is an unstable domain (DD domain) that is rapidly degraded after expression.
  • DD domain unstable domain
  • the chemical small molecule shield-1 can bind this structure to make it stable without rapid degradation.
  • the target protein is bound to ddFKBP, and the stability and degradation process of the protein can be effectively regulated by shield-1 (Banaszynski et al., 2006).
  • ecDHFR can also regulate its own stability by binding to trimethoprim (Iwamoto et al., 2010; patent number: 09487787).
  • the plant hormone binding domain (AID) IAA17 binds to TIR1 mediated by indoleacetic acid and is recognized by ubiquitin ligase and is degraded by ubiquitination in one step (Nishimura et al., 2009; :US20120115232).
  • the existing protein activity control technologies mainly include: 1. Using rapamycin to regulate the polymerization and dissociation of FKBP and FRB; 2. Using the interaction between abscisic acid regulation (PYR) and PYR1-like (PYL) 3. Using gibberellin to regulate the interaction between CID and GAI; 4. Using intein to achieve splicing of two-stage protein; 5. Using regulatable degradants to achieve regulation of fusion protein activity, such as shield-1 regulation of ddFKBP fusion The activity of the protein, TMP regulates the activity of the DHFR fusion protein, and the acetic acid regulates the activity of the AID fusion protein.
  • the technical problem to be solved by the present invention is how to regulate the target protein.
  • the present invention first provides a small molecule drug controlled protein function switching system.
  • the switch system provided by the invention consists of an operon original and a control sub-unit, the control is a single viral protease, or a protease-degradant formed by fusion of a viral protease and a protein degradation agent; the operon is located inside the target protein Or located between the target protein and the protease-degrading structure, and consists of a ligation peptide of the protease; the whole system regulates the target protein through the inhibitor compound of the small molecule of the protease as a switch.
  • the regulation of the protein of interest involves air conditioning control of the structure and/or function and/or location of the protein of interest.
  • the regulation of the structure and function includes regulating the primary structure, the tertiary structure, the expression of the protein, the stability, the functional activity, and the intracellular position of the target protein.
  • the switching system further comprises an inhibitor compound of a small molecule of a protease.
  • the operon element is a linker peptide recognizable and cleaved by the protease; the number of the operon elements is one or two or more.
  • the protease may be a viral protease.
  • the viral protease may be a hepatitis C virus protease (NS3pro/4A) or a protease having 70% or more homology with the hepatitis C virus protease NS3pro/4A. Mutant or isomerase.
  • the amino acid sequence of the NS3pro/4A protease is SEQ ID No. 7.
  • the invention mutates the NS3pro and 4A structural linking portions of the N34d protein to obtain the NS3pro/4A protease, and the NS3pro/4A protease has stable structure and loses the function of the degradation, but retains the complete protease function.
  • the N34d protein is an unstable and easily degradable protein formed by removing the NS3 RNA helicase region from the NS3/4A structure.
  • the viral protease can also be ligated into a degrading domain (degron).
  • the degrading domain may be an NS4A degrader which, after being fused to the corresponding protein, degrades in a very short period of time, thereby affecting protein function.
  • the protease-degrading protein is an N34d protein having an amino acid sequence of SEQ ID No. 6.
  • the degrading domain can also be a small molecule-regulated degrader, such as ddFKBP or exDHFR.
  • the two domains can be stabilized for a long time without being degradation. After removal of the ligand, the two structures are extremely unstable and, after expression, degrade in a very short time.
  • linker peptide is one of the following sequences:
  • 4B5A Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
  • 5A5B Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
  • the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir (CAS Number: 402957-28-2); Boceprevir (CAS Number: 394730-60-0); Simeprevir (CAS Number: 923604- 59-5); Faldaprevir (CAS Number: 801283-95-4); Asunaprevir (CAS Number: 630420-16-5); Furaprevir (CAS Number: 1435923-88-8).
  • the degradation agent is an NS3/4A unstable region, a ddFKBP unstable region, or an ecDHFR unstable region.
  • the target protein is a single protein or a fusion protein.
  • Each protein may be a protein having one or several domains, or a fusion protein composed of a plurality of functional proteins.
  • the operon element may be located inside the target protein (such as between domains) or between the fusion moieties.
  • the target protein may be any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
  • the target protein is a single protein, the target protein, the control subunit, and the operon element constitute a fusion protein, the operon element is used to link the target protein and the control sub-assembly; the control The sub-origin is a protease-degrading agent;
  • the target protein is a fusion protein having several domains or several functional proteins
  • a viral protease recognition sequence can be inserted between each domain or a functional protein.
  • the target protein, the control subunit, and the operon element constitute a fusion protein, the operon element is used to link the target protein and the control subunit, or to link a domain of the target protein Or functional protein.
  • the protein of interest may be a fusion protein of two different fluorescent proteins, the linker peptide being located between two different fluorescent proteins, joining two different fluorescent proteins, and fluorescent protein fusion expression in the case where protease activity is inhibited It will co-localize in the cell. Under the protease active state, the fluorescent protein expressed by fusion will be cleaved into two proteins by protease and expressed separately. Or a chimeric antigen receptor (CAR) having multiple domains, the linker peptides being located between different domains, linking different domains, allowing the various domains of the chimeric antigen receptor to be cascaded, and the protease activity is inhibited.
  • CAR chimeric antigen receptor
  • the chimeric antigen receptor is expressed in T cells and has the function of recognizing a tumor antigen delivery signal and activating T cell function. In the protease active state, each domain of the chimeric antigen receptor is cleaved into two or more Independent structure that affects T cells to recognize tumor antigens and transmit signals.
  • the target protein can be expressed in fusion with viral proteases or separately. Fusion expression allows the viral protease to be located closer to the target protein and function faster. Separate expression reduces the effect on the structure of the target protein and more precisely regulates protein activity.
  • protein functional switching system is used in eukaryotic mammalian cells.
  • the present invention further provides a nucleic acid molecule, a plasmid, a cell line, and a kit comprising the above elements for expressing a protein function switching system.
  • the nucleic acid molecule is a DNA molecule represented by SEQ ID No. 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4 or SEQ ID No. 5;
  • An expression vector for a nucleic acid molecule which may specifically be a lentiviral vector Pltr vector;
  • the cell line is a cell line containing the plasmid, and the cell line may specifically be a 293T cell or a 3T3 cell.
  • the present invention also provides a novel use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit.
  • the invention provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the structure and/or function of a protein of interest.
  • the invention also provides the use of a protein functional switch system or nucleic acid molecule, plasmid, cell line or kit as described above for the preparation of a product that modulates the structure and/or function of a protein of interest.
  • the invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the activity of a target protein.
  • the invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the activity of a target protein.
  • the invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the stability of a target protein.
  • the invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the stability of a target protein.
  • the invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the spatiotemporal position of a target protein.
  • the invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the spatiotemporal position of a target protein.
  • the present invention finally provides a method of regulating a target protein.
  • the method for regulating a target protein comprises the steps of: expressing a fusion protein comprising a promoter subgenus, an operon element and a target protein in a recipient cell, and then using an inhibitor compound of a protease small molecule to regulate the controller The activity of the original, thereby achieving the regulation of the target protein;
  • the control element is a single viral protease, or a protease-degradant fused by a viral protease and a protein degrader; the operon element is a linker peptide that the protease recognizes and cleaves.
  • the number of the operon originals is one or two or more.
  • the viral protease is a hepatitis C virus protease.
  • the hepatitis C virus protease is specifically a hepatitis C virus protease NS3pro/4A or a protease mutant or isomerase having 70% or more homology with the hepatitis C virus protease NS3pro/4A.
  • the linker peptide is one of the following sequences:
  • 4B5A Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
  • 5A5B Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
  • the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir (CAS Number: 402957-28-2); Boceprevir (CAS Number: 394730-60-0); Simeprevir (CAS Number: 923604) -59-5); Faldaprevir (CAS Number: 801283-95-4); Asunaprevir (CAS Number: 630420-16-5); Furaprevir (CAS Number: 1435923-88-8).
  • the proton is an NS3/4A unstable region, a ddFKBP unstable region, or an ecDHFR unstable region.
  • the protease-degrading agent is an N43d protein.
  • the target protein is a single protein or a fusion protein.
  • Each protein may be a protein having one or several domains, or a fusion protein composed of a plurality of functional proteins.
  • the operon element may be located inside the target protein (such as between domains) or between the fusion moieties.
  • the target protein may be any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
  • the method of expressing the fusion protein comprising the promoter element, the operon element and the target protein in the recipient cell, and then using the inhibitor compound of the protease small molecule to regulate the activity of the control element is a vector expressing the control element, the operon element, and the target protein is introduced into the recipient cell to obtain a recombinant cell; the recombinant cell is cultured in a culture system to obtain the fusion protein; Addition or removal of the inhibitor compound in the culture system can effect modulation of the activity of the control element.
  • the specific method can be as follows (1) or (2):
  • (1-1) co-transfecting a receptor-derived cell with a protease-degradant expressing a target protein, a protease and a proteosome, and a vector of the protease-recognizing and cleavable linker peptide together with the small molecule inhibitor of the protease Recombinant cells are obtained; the recombinant cells are cultured in a recombinant cell culture system, and a fusion protein is expressed, wherein the inhibitor inhibits the recognition and cleavage activity of the protease in the fusion protein, and the degradant converts the fusion protein Degrading, the target protein in the fusion protein is inactivated;
  • the fusion protein consists of the target protein, the protease-degradant, and the linker peptide for ligation;
  • the vector expressing the control subunit, the operon original, and the target protein is a coding gene containing the control subunit, a coding gene of the operon original, and the target protein.
  • a fragment of the coding gene is inserted into the multiple cloning site of the expression vector.
  • the coding gene containing the control subunit, the coding gene of the operon original, and the fragment of the coding gene of the target protein may specifically be SEQ ID No. 1 or SEQ ID No. 2 or SEQ ID No. 3. Or the DNA molecule shown in SEQ ID No. 4 or SEQ ID No. 5.
  • the expression vector may specifically be a lentiviral vector Pltr vector.
  • the recipient cell may specifically be a 293T cell or a 3T3 cell.
  • the vector expressing the target protein including: functional protein, fusion protein or genetic engineering protein
  • viral protease or engineered viral protease
  • viral protease recognition and cleavable linker peptide is co-transformed.
  • the activity of the protease can be regulated by the addition and removal of small molecule inhibitors of the corresponding proteases, and the expression of the protease affects the function of the target protein, thereby indirectly controlling the function of the target protein.
  • the viral protease of the present invention may be a natural protease, a protease that is genetically engineered to reduce the molecular weight of the protease, or a protease-degradant fused to a degrading domain (degron).
  • Viral protease inhibitors are primarily drugs that are commercially available for treatment or for clinical trials.
  • the invention prepares a switching system of a virus protease-dependent small molecule drug for regulating the function of a target protein, and proposes a protein function regulation method which is fast, efficient, reversible, controllable, simple, economical and has good application prospect.
  • FIG. 1 is a diagram showing the SwichOFF system-green fluorescent protein expression vector in Example 1 of the present invention.
  • Fig. 2 is a diagram showing the expression of green fluorescent protein by small molecule ASV in Example 1 of the present invention. Above: Green fluorescent protein is stably expressed when no ASV is added; the following figure: Green fluorescent protein is degraded and not expressed after adding ASV.
  • Figure 3 is a rapid switching of fluorescent protein expression in Example 1 of the present invention. Above: 24 hours after the addition of ASV, green fluorescence rapidly diminished. Bottom: After removal of ASV, fluorescence is rapidly and stably expressed.
  • FIG. 4 is a diagram showing the SwichON system-fusion fluorescent protein expression vector in Example 2 of the present invention.
  • Figure 5 is a diagram showing the localization of a fluorescent protein in a cell by the SwichON system in Example 2 of the present invention.
  • the first column is red fluorescence
  • the second column is green fluorescence
  • the third column is fluorescence overlay.
  • the first row no ARV, green fluorescence is expressed in the cytoplasm; the second column: adding ASV, green fluorescence is expressed in mitochondria.
  • Figure 6 is a diagram showing the localization of the fast-switching fusion fluorescent protein in the SwichON system of Example 2 of the present invention.
  • the first column is red fluorescence
  • the second column is green fluorescence
  • the third column is fluorescence overlay. 24 hours after the addition of ASV, green fluorescence rapidly localized from the cytoplasm to the mitochondria.
  • Figure 7 is a diagram showing the localization of the fast-switching fusion fluorescent protein in the SwichON system of Example 2 of the present invention.
  • the first column is red fluorescence
  • the second column is green fluorescence
  • the third column is fluorescence overlay. 24 hours after the removal of ASV, green fluorescence was rapidly released from the mitochondria to the cytoplasm.
  • Figure 8 is a map of the SwichOFF system CAR regulation carrier in the third embodiment of the present invention.
  • Fig. 9 is a schematic diagram showing the regulation of CAR expression by the SwichOFF system in the third embodiment of the present invention.
  • Figure 10 is a map of the SwichON system CAR regulatory vector in Example 4 of the present invention.
  • Fig. 11 is a schematic diagram showing the function of regulating the CAR of the SwichON system in the fourth embodiment of the present invention.
  • Figure 12 is a map of the SwichON system CAR regulatory vector in Example 5 of the present invention.
  • Figure 13 is a schematic diagram showing the function of the SwichON system for regulating CAR in the fifth embodiment of the present invention.
  • Figure 14 is a Western blot analysis of the Switch protein regulatory system in Example 6 of the present invention.
  • fCAR-V1 The first column: a section of 63kd Flag-CAR fragment is expressed without ASV. Second column: After the addition of ASV, the fusion protein rapidly degraded.
  • fCAR-V2 group First column: Flag-CAR1 protein of 45 kd in size was detected without ASV. Second column: After adding ASV, the anti-flag antibody detected 97kd of intact protein.
  • fCAR-V3 group The first column: the Flag-CAR1 protein with a size of 33 kd was expressed without ASV. Second column: After adding ASV, the anti-flag antibody detected 97kd of intact protein. Since the hinge site of CAR itself may naturally break, a group of 33kd protein can be detected in all groups.
  • Figure 15 shows the results of the chimeric antigen receptor killing function test.
  • Example 1 Small molecule drug modulates the activity of green fluorescent protein (SwichOFF system)
  • the green fluorescent protein is expressed by fusion with the self-degrading HCV protease, and the expression of the green fluorescent protein is regulated by the HCV protease inhibitor ASV by inhibiting the protease activity.
  • the fusion expression gene represented by SEQ ID No. 1 was inserted into a lentiviral vector Pltr vector (addgen, 25870) to obtain a recombinant vector Pltr-sfGFP-N3N4, and positions 1-714 of SEQ ID No. 1 were sfGFP gene, 721-
  • the 753 is the coding gene of the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), and the 842-1635 is the N34d gene (with the NS3/4A sequence and a stretcher sequence).
  • the amino acid sequence of the N34d protein expressed by the N34d gene is SEQ ID No. 6.
  • the NS3/4A structure contains NS3pro (protease region), NS3 RNA helicase region and 4A region.
  • the N34d protein is an unstable and easily degradable protein formed by removing the NS3 RNA helicase region from the NS3/4A structure.
  • Fig. 1 The structure of the recombinant vector Pltr-sfGFP-N3N4 is shown in Fig. 1.
  • ASV 0uM group one well was added to the medium without ASV (DMEM (GBICO) containing 10% serum (BI));
  • ASV 1uM group One well was added to the medium containing 1 uM ASV.
  • the specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 50uL respectively. Medium, then add 3.0uL Lipofectamine to one of the tubes 3000 reagent (Thermo Fisher), 2 ug DNA plasmid (recombinant vector Pltr-sfGFP-N3N4) was added to the other tube, and mixed separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 100 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 50 uL of the mixture to the corresponding wells of the ASV 0uM group and the ASV 1uM group, respectively, and change the solution after 6 hours.
  • the cells were observed to express fluorescence.
  • the result is shown in Figure 2. It can be seen from the figure: the first row: the fusion protein sfGFP-4A4B-NS3/4A-Degron expressed without ASV (ASV 0uM group). The protease has a cleavage function and the fusion protein is cleaved at 4A4B. Degron then mediates rapid degradation of the NS3/4A protease without affecting the function of sfGFP.
  • ASV ASV 1uM group
  • was added and the 3T3 cells also expressed the fusion protein sfGFP-4A4B-NS3/4A-Degron.
  • ASV inhibited the recognition and cleavage of 4A4B by the NS3/4A protease, allowing Degron to bring the entire fusion protein into the ubiquitin-proteasome pathway for rapid degradation.
  • sfGFP is rapidly degraded, rendering cells unable to express green fluorescent protein. This indicates that ASV can regulate the expression of target proteins.
  • the cell culture medium of ASV 0uM group was aspirated, and fresh medium containing 1 uM of ASV was added;
  • the ASV 1uM cell culture medium was aspirated, washed twice with PBS, and then fresh ASV-free medium was added.
  • the cells were observed to express fluorescence after adding ASV and removing ASV for 24 h.
  • the result is shown in Figure 3. It can be seen from the figure: The first row: without the addition of ASV, the expression of green fluorescent protein is very high in the cells, and the expression of green fluorescent protein drops sharply at 24 h after the addition of ASV, to a very weak level. The second row: under the condition of adding ASV, the cells could not express or only a few cells expressed very weak green fluorescent protein. After 24 hours of ASV, the expression of green fluorescent protein in the cells recovered to a high level. This indicates that ASV can quickly switch the expression of the target protein.
  • the green fluorescent protein, the red fluorescent protein and the self-degrading HCV protease are fused and expressed, and the HCV protease inhibitor ASV inhibits the localization of the green fluorescent protein and the red fluorescent protein by inhibiting the protease activity.
  • the fusion protein gene represented by SEQ ID No. 2 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-tomDsRed-N3N4-GFP V2, and the 1-10th position of SEQ ID No. 2 was the mitochondrial membrane localization signal of the Tom20 gene.
  • the coding gene, the 118th-789th position is the DsRed ex gene, the 808-840 is the coding gene of the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), the 880-1590 is the sfGFP gene, and the 1606-1629 bits are the flag tag sequence.
  • the 1660-2352 position is the NS3pro/4A protease gene.
  • the amino acid sequence of the NS3pro/4A protease encoded by the NS3pro/4A protease gene is SEQ ID No. 7.
  • the NS3pro/4A protease is a protease obtained by mutating the NS3pro and 4A structural linking portions of the N34d protein.
  • the NS3pro/4A protease is structurally stable and loses its proteolytic function, but retains intact protease function.
  • the mitochondrial membrane localization signal sequence of the Tom20 gene is as follows: Met Val Gly Arg Asn Ser Ala Ile Ala Ala Gly Val Cys Gly Ala Leu Phe Ile Gly Tyr Cys Ile Tyr Phe Asp Arg Lys Arg Arg Ser Asp Pro Asn Phe Lys.
  • the flag tag sequence is as follows: Asp Tyr Lys Asp Asp Asp Asp Lys.
  • ASV 0uM group one well was added to the medium without ASV;
  • ASV 1uM group One well was added to the medium containing 1 uM ASV.
  • the specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 50uL respectively. Medium, then add 3.0uL Lipofectamine to one of the tubes 3000 reagent (Thermo Fisher), 2 ug DNA plasmid (recombinant vector Pltr-sfGFP-N3N4) was added to the other tube, and mixed separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 100 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 50 uL of the mixture to the corresponding wells of the ASV 0uM group and the ASV 1uM group, respectively, and change the solution after 6 hours.
  • the cells were observed to express fluorescence.
  • the result is shown in Figure 5. It can be seen from the figure that the first row: the fusion protein MTS-DsRed-4A4B-sfGFP-NS3/4A was expressed without ASV (ASV 0uM group).
  • the protease is active and can cleave the fusion protein at 4A4B to form two proteins, MTS-DsRed and sfGFP-NS3/4A. Due to the mitochondrial localization signal, MTS-DsRed expression was localized on the mitochondrial outer membrane, while the sfGFP-NS3/4A fusion protein was widely distributed in the cytoplasm.
  • the cell culture medium of ASV 0uM group was aspirated, and fresh medium containing 1 uM of ASV was added;
  • the ASV 1uM cell culture medium was aspirated, washed twice with PBS, and then fresh ASV-free medium was added.
  • the cells expressed fluorescence after 24 hours of ASV addition The result is shown in Figure 6. As can be seen from the figure: The first row: green fluorescent protein and red fluorescent protein are expressed separately without ASV. Second row: After 24 hours of ASV addition, the localization of green fluorescent protein in the cells was gradually transferred to the mitochondrial surface.
  • the chimeric antigen receptor (CAR) is expressed by fusion with a self-degrading HCV protease, and the chimeric antigen receptor activity is regulated by the HCV protease inhibitor ASV by inhibiting protease activity.
  • the fusion protein gene represented by SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N34d, the 1st to 27th of SEQ ID No. 3 was the Flag tag sequence, and the 28th to 1491th were CAR Genes, positions 1498-1530 are the genes encoding the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), and positions 1588-2388 are the N34d gene (with the NS3/4A sequence and a stretcher sequence).
  • the structure of the recombinant vector Pltr-CAR19-N34d is shown in Fig. 8.
  • the antigen recognized by the chimeric antigen receptor is human CD19, and the CAR is expressed by fusion with the self-degrading HCV protease, and the protease recognition site 4A4B is located between the two proteins.
  • the HCV protease inhibitor ASV regulates CAR expression by inhibiting protease activity.
  • Figure 9 Left: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, CAR is separated from protease, protease is degraded by degron into the ubiquitin-proteasome system, and CAR can express and recognize antigen And the function of activating T cells internal signals. At this point, the switch is in the on state.
  • CAR chimeric antigen receptor
  • the fusion protein gene represented by SEQ ID No. 4 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N3V4V2, the 1st to 27th of SEQ ID No. 4 was the Flag tag sequence, and the positions 28 to 1530 were CAR.
  • the structure of the recombinant vector Pltr-CAR19-N34d is shown in FIG.
  • the chimeric antigen receptor recognizes an antigen that is human CD19, and fuses CAR and HCV protease, and the protease recognition site 4A4B is located between the 4-1BB and CD3zata domains of the CAR.
  • the structure and function of CAR is controlled by HCV protease by ASV.
  • Figure 11 Left panel: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, and the 4-1BB in the CAR protein is separated from the CD3zeta domain and cannot transmit cell proliferation signals. Even if CAR-T cells recognize tumor antigens, they cannot effectively kill the corresponding tumor cells because they cannot proliferate. At this point, the switch is in the off state.
  • NS3/4A is inhibited and can not cleave 4A4B sequence, CAR, protease fusion expression, CAR has a complete structure, and functions to recognize antigen and activate T cell internal signals. At this point, the switch is in the on state.
  • the fusion of the chimeric antigen receptor (CAR) with the HCV protease is different from that of the second embodiment in that the protease recognition region is increased within the CAR protein by three, which are located between the anti-CD19 scFv of the CAR and the transmembrane region of the CD8, Between the 4-1BB and CD3zata domains, between the CD8 transmembrane region and the 4-1BB domain.
  • the structure and function of CAR is controlled by HCV protease by ASV.
  • the fusion protein gene represented by SEQ ID No. 5 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N3V4V3, the 1st to 28th of SEQ ID No. 5 was the Flag tag sequence, and the 28th to the 1296th was CAR
  • the gene encoding the HCV viral protease recognition sequence 4A4B is located between the scFv and CD8 transmembrane regions, the CD8 transmembrane region and the 4-1BB domain, and the HCV viral protease recognition sequence 5A5B is located between the 4-1BB and CD3zata domains, 1648.
  • the -2334 position is the NS3pro/4A protease gene.
  • the structure of the recombinant vector Pltr-CAR19-N34d is shown in FIG.
  • the chimeric antigen receptor recognizes the antigen as human CD19, and fuses the expression of CAR and HCV protease.
  • the protease recognition sites are located between the anti-CD19scFv of CAR and the transmembrane region of CD8, respectively.
  • Figure 13 Left: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, and the CAR protein is cleaved into four separate domains, causing the corresponding T cells to fail to recognize the antigen and also fail to deliver cell activation and Proliferation signals cannot kill the corresponding tumor cells.
  • the switch is in the off state.
  • NS3/4A is inhibited and can not cleave 4A4B sequence, CAR and protease fusion expression, CAR has a complete structure, and functions to recognize antigen and activate T cell internal signal.
  • the switch is in the on state.
  • 293T cells (Clontech: 632180) were resuscitated and cultured, and fCAR-V1, fCAR-V2 and fCAR-V3 were transfected into 293T cells, respectively.
  • fCAR-V1, fCAR-V2 and fCAR-V3 were transfected into 293T cells, respectively.
  • One day before transfection 5 x 10 5 cells were added to each well in a 12-well plate.
  • the liquid exchange before transfection was divided into the following two groups according to whether or not the small molecule inhibitor Asunaprevir (ASV) was added:
  • ASV 0uM group one well was added to the medium without ASV;
  • ASV 1uM group One well was added to the medium containing 1 uM ASV.
  • the specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 100uL respectively. Medium, then add 6.0uL Lipofectamine to one of the tubes For the 3000 reagent, add 4ug DNA plasmid (fCAR-V1, fCAR-V2 or fCAR-V3) to the other tube and mix separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 200 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 100 uL of the mixture to each of the two wells, and change the solution after 6 hours.
  • fCAR-V1 group the first column: the fusion protein Flag-CAR-4A4B-NS3/4A-Degron expressed without ASV.
  • the protease has a cleavage function and the fusion protein is cleaved at 4A4B.
  • Degron mediates the rapid degradation of NS3/4A protease to form a 63kd size Flag-CAR fragment.
  • Second column After adding ASV, 293 cells also expressed the fusion protein Flag-CAR-4A4B-NS3/4A-Degron.
  • fCAR-V2 group The first column: the fusion protein Flag-CAR1-4A4B-CAR2-NS3/4A was expressed without ASV. The protease is active and can cleave the fusion protein at 4A4B to form a 45 kd Flag-CAR1 and a 52 kd CAR2-NS3/4A protein. 45 kd Flag-CAR1 was detected by anti-flag antibody.
  • Second column After addition of ASV, 293 cells expressed the fusion protein Flag-CAR-4A4B-CAR2-NS3/4A. Since ASV inhibits the recognition and cleavage of 4A4B by NS3/4A protease, the entire fusion protein can be stably present, and the anti-flag antibody detects a protein size of 97 kd.
  • fCAR-V3 group First column: The fusion protein Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A was expressed without ASV.
  • the protease is active and can cleave the fusion protein at 4A4B and 5A5B to form four proteins of Flag-CAR1, 33kd, CAR2, 5kd CAR3 and 52kd CAR4-NS3/4A.
  • the 33 kd Flag-CAR1 was detected by an anti-flag antibody.
  • Second column After addition of ASV, 293 cells expressed the fusion protein Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A.
  • the entire fusion protein can be stably present, and the anti-flag antibody detects a protein size of 97 kd. Since the hinge site of CAR itself may naturally break, a group of 33kd protein can be detected in all groups.
  • the fusion protein gene represented by SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector to obtain a lentiviral vector Pltr-CAR19-N34d expressing the chimeric antigen receptor (CAR) and self-degradation of the Pltr-CAR19-N34d vector. Fusion protein of HCV protease.
  • the CAR gene shown in positions 28-1491 of SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector, and the resulting lentiviral vector PLTR-CAR19 was obtained.
  • the lentiviral vector PLTR-CAR19 expresses a chimeric antigen receptor (CAR).
  • Lentiviral PLTR-CAR19-N34d and lentiviral vector PLTR-CAR19 were co-transfected into 293T cells with viral packaging plasmids to obtain viral particles. Two days later, the cell culture supernatant was collected and PEG-precipitated for virus concentration. The virus solution CAR19-N34d and the virus solution CAR19 were respectively obtained and stored frozen at -80 ° C for use.
  • T cells are cultured with X-vivo15 containing 0.5% HSA and 300U IL2).
  • Lonza cultured cells obtained three days after PBMC cells (Tianjin blood center), and regulatable CAR-T cells and common CAR-T cells were obtained, respectively.
  • the CAR-T cells expressing the chimeric antigen receptor are co-cultured with the raji target cells in a culture system containing the viral protease inhibitor ASV or DPV (Aladdin), and the virus protease inhibitor is not included as a control (Control) ).
  • Elisport was used to detect the expression of IFN-gamma in CAR-T cells. Specific steps are as follows:
  • Fig. 15 The results are shown in Fig. 15. As can be seen from the figure: the upper row: normal CAR-T cells release a large amount of IFN-gamma (black spots in the figure) after binding to raji cells, and the addition of the viral protease inhibitors ASV or DPV does not Inhibition of release of IFN-gamma by CAR-T cells. Lower row: Regulatory CAR-T cells also release large amounts of IFN-gamma after binding to raji cells, but the expression of IFN-gamma is greatly reduced by the addition of the viral protease inhibitors ASV or DPV to the culture system.
  • the invention discloses a small molecule drug controlled protein function switching system.
  • the system comprises a control element original and a operon original, the control element is a protease or a protease-degradant fused by a protease and a proton; the operon element is a linker peptide recognizable and cleaved by the protease;
  • the system also includes a small molecule inhibitor of the protease; the entire system modulates the activity of the protease or protease-degradant by a small molecule inhibitor of the protease, thereby regulating the activity of the protein of interest.
  • the small molecule drug can be used to precisely regulate the biological activity of the target protein.
  • the viral protease of the present invention may be a natural protease, a protease that is genetically engineered to reduce the molecular weight of the protease, or a protease-degradant fused to a degrading domain (degron).
  • Viral protease inhibitors are primarily drugs that are commercially available for treatment or for clinical trials.
  • the invention prepares a switching system of a virus protease-dependent small molecule drug for regulating the function of a target protein, and provides a protein function regulation method which is fast, efficient, reversible, controllable, simple, economical and has good application prospect.

Landscapes

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

Abstract

Disclosed is a protein function switch system controlled by a small molecule drug, consisting of an expression target protein, a protease or a protease-degron, a carrier of a linker peptide able to be recognized and cleaved by the protease, and a small molecule inhibitor of the protease. The biological activity of the target protein can be precisely regulated using the small molecule drug. The viral protease can be a natural protease, and can also be a protease genetically engineered, with the protease having reduced molecular weight, or can be a protease-degron formed by fusion with a degron domain. The viral protease inhibitor is mainly a drug for use in treatment or entering a clinical experiment on the market. Prepared is a switch system using a viral protease-dependent small molecule drug for regulating a target protein function. Provided is a rapid, efficient, reversible, controllable, simple and economic protein function regulation method with good application prospects.

Description

一种小分子药物控制的蛋白功能开关系统Small molecule drug controlled protein function switch system 技术领域Technical field
本发明涉及生物技术领域,具体涉及一种小分子药物控制的蛋白功能开关系统。The invention relates to the field of biotechnology, in particular to a protein function switching system controlled by a small molecule drug.
背景技术Background technique
细胞内不同的蛋白行使着不同的功能,而要研究其功能,通常是用某些方法干扰其行使功能,观察其反应。目前常用的方法是在基因组或转录水平操纵细胞内特定基因的表达,但是直接在蛋白水平的调控研究相对较少。直接对胞内蛋白的功能进行操作具有灵敏、快速、可逆、可控性、简便等方面的优点。结合遗传操作的特异性和基于小分子药物调控的方法,可在时间和空间上对细胞或组织特定蛋白进行精确控制,从而减少不必要的副作用。如蛋白在细胞内的定位、蛋白分子的激活或抑制、稳定或降解,细胞内特定信号通路的激活或阻断等。目前,蛋白功能调控方式主要分为三类:①利用小分子或光调控两个异源二聚体的结合或解离;②利用小分子调控蛋白结构变化;③利用小分子调节控制蛋白的稳定性。下面分别介绍各个类型蛋白功能调控所涉及到的技术:Different proteins in the cell perform different functions, and to study their function, they usually interfere with their function by some methods and observe the reaction. At present, the commonly used method is to manipulate the expression of specific genes in cells at the genomic or transcriptional level, but there are relatively few studies directly on the regulation of protein levels. Direct manipulation of the function of intracellular proteins is advantageous in terms of sensitivity, rapidity, reversibility, controllability, and simplicity. Combining the specificity of genetic manipulation with a method based on small molecule drug regulation allows precise control of cell or tissue specific proteins in time and space, thereby reducing unnecessary side effects. Such as the localization of proteins in cells, activation or inhibition of protein molecules, stabilization or degradation, activation or blockade of specific signaling pathways within cells. At present, protein function regulation methods are mainly divided into three categories: 1 using small molecules or light to regulate the binding or dissociation of two heterodimers; 2 using small molecules to regulate protein structure changes; 3 using small molecules to regulate protein stability Sex. The techniques involved in the regulation of each type of protein function are described below:
(1)利用小分子控制两个异源二聚体的结合或解离(1) Using small molecules to control the binding or dissociation of two heterodimers
在过去二十年中,化学小分子已经作为调控蛋白质-蛋白质相互作用的有效手段。这些化合物被称为二聚化的化学诱导物(chemical inducers of dimerization,CID),具有同时结合两个蛋白质结构域的能力,从而诱导它们的相互接近。CID已经用于:①促进或抑制特定基因的转录活性;②将靶蛋白募集到细胞内特定区域;③调控蛋白质聚集和解聚。In the past two decades, chemical small molecules have been used as an effective means of regulating protein-protein interactions. These compounds are known as chemical inducers of dimerization (CID) and have the ability to simultaneously bind two protein domains, thereby inducing their proximity. CID has been used to: 1 promote or inhibit the transcriptional activity of specific genes; 2 recruit target proteins to specific regions within the cell; 3 regulate protein aggregation and depolymerization.
1991年Liu等人发现了第一个CID——免疫抑制剂药物FK506。这个小分子通过同时结合FK506结合蛋白(FKBP12)和钙依赖磷酸酶,抑制T细胞受体介导的信号传导。在此基础上,将FK506合成二聚体(命名为FK1012),其能够使FKBP12二聚化。FKBP12与T细胞受体的ζ链的融合可实现快速的,可调节的和可逆的FK1012依赖的信号转导调节(Spencer et al.,1993)。而目前研究最充分的CID为雷帕霉素(rapamycin),这是一种大环内酯天然产物,介导FKBP12与mTOR的FRB结构域之间的相互作用(Brown et al.,1994)。如果将两种感兴趣的蛋白质分别与FKBP和FRB融合表达,可在雷帕霉素存在的情况下相互靠近,雷帕霉素在低剂量条件下就可快速实现FKBP-FRB的结合。然而,细胞内源性FKBP和mTOR会与FKBP和FRB融合蛋白竞争结合雷帕霉素,导致无效的相互作用。雷帕霉素与mTOR的结合还会导致细胞周期的停滞(Haruki et al.,2008)。为了减轻其脱靶效应,通过将雷帕霉素衍生化以及FKBP或FRB结构域进行定点突变,优化获得Rimiducid调控FKBP-FRB二聚化的方法可有效降低CID脱靶效应(专利号:US2015/065629)。In 1991, Liu et al. discovered the first CID, the immunosuppressive drug FK506. This small molecule inhibits T cell receptor-mediated signaling by simultaneously binding to FK506 binding protein (FKBP12) and calcineurin. On this basis, FK506 was synthesized as a dimer (designated FK1012) which was capable of dimerizing FKBP12. Fusion of FKBP12 to the ζ chain of T cell receptors enables rapid, regulatable and reversible FK1012-dependent signal transduction regulation (Spencer et al., 1993). The most well-studied CID currently studied is rapamycin, a macrocyclic natural product that mediates the interaction between FKBP12 and the FRB domain of mTOR (Brown et al., 1994). If the two proteins of interest are fused to FKBP and FRB, respectively, they can be close to each other in the presence of rapamycin, and rapamycin can rapidly achieve FKBP-FRB binding at low doses. However, cellular endogenous FKBP and mTOR compete with FKBP and FRB fusion proteins for binding to rapamycin, resulting in ineffective interactions. Binding of rapamycin to mTOR also leads to cell cycle arrest (Haruki et al., 2008). In order to alleviate its off-target effect, the optimized method to obtain Rimiducid-regulated FKBP-FRB dimerization can effectively reduce the CID off-target effect by site-directed mutagenesis of rapamycin derivatization and FKBP or FRB domain (Patent No.: US2015/065629) .
最近,一些新的蛋白质-配体对的发现促进了CID系统的发展。例如,地塞米松-甲氨蝶呤(Dex-Mtx)复合物可诱导糖皮质激素受体(GR)和二氢叶酸还原酶 (DHFR)之间的相互作用。令人遗憾的是,Mtx是DHFR的抑制剂,这限制其应用。经过改进后,甲氧苄啶-SLF(TMP-SLF)化合物可触发DHFR-FKBP12融合体之间的相互作用(Czlapinski et al.,2008)。该化合物不结合内源性蛋白质,但这些小分子是昂贵的并且需要多步合成,这限制了它们的广泛应用。近年来,科学家发现一些植物激素可有效调控目标蛋白的结合与解离。脱落酸(ABA)是刺激植物发育的激素之一。它通过结合pyribactin resistance(PYR)/PYR1-like(PYL),抑制蛋白磷酸酶(PP2Cs)(Cutler et al.,2010;专利号:US2012043121)。将PYL和PP2C结构域分别与DNA结合结构域Gal4和转录激活结构域VP64融合后,可以重建Gal4转录活性。另一种植物激素——赤霉素(GA3)也可以用作CID。GA3可结合GID1,促进其和GAI之间的相互作用(Hirano et al.,2008;Miyamoto et al.,2012)。植物激素相对经济,安全和反应灵敏,使其具有一定的应用价值。Recently, the discovery of some new protein-ligand pairs has facilitated the development of CID systems. For example, the dexamethasone-methotrexate (Dex-Mtx) complex induces an interaction between the glucocorticoid receptor (GR) and dihydrofolate reductase (DHFR). Unfortunately, Mtx is an inhibitor of DHFR, which limits its use. After modification, the trimethoprim-SLF (TMP-SLF) compound triggers the interaction between the DHFR-FKBP12 fusions (Czlapinski et al., 2008). This compound does not bind endogenous proteins, but these small molecules are expensive and require multiple steps of synthesis, which limits their widespread use. In recent years, scientists have discovered that some plant hormones can effectively regulate the binding and dissociation of target proteins. Abscisic acid (ABA) is one of the hormones that stimulate plant development. It inhibits protein phosphatase (PP2Cs) by binding to pyribactin resistance (PYR)/PYR1-like (PYL) (Cutler et al., 2010; patent number: US2012043121). After merging the PYL and PP2C domains with the DNA binding domain Gal4 and the transcriptional activation domain VP64, respectively, Gal4 transcriptional activity can be reconstituted. Another phytohormone, gibberellin (GA3), can also be used as CID. GA3 binds to GID1 and promotes its interaction with GAI (Hirano et al., 2008; Miyamoto et al., 2012). Plant hormones are relatively economical, safe and responsive, making them useful.
(2)利用小分子调控蛋白片段互补(2) Using small molecules to regulate protein fragmentation
将感兴趣的蛋白质分裂成两个无活性片段,特定化合物小分子可以诱导这些片段缔合,从而恢复靶蛋白的结构和功能,这个过程也称为片段互补。Muir和同事开发了一个基于内含肽的条件性蛋白剪接系统(Mootz and Muir,2002)。将内含肽分成N-和C-末端两半,单独存在时,两个内含肽都没有活性,但一旦两者接近就形成有活性的结构域,将两端的蛋白剪接在一起,形成活性蛋白。例如:两段前者融合在FKBP和麦芽糖结合蛋白(MBP)之间,后者融合在FRB和His标签之间。这些融合物在雷帕霉素诱导下二聚化,内含肽结构域接近,其恢复内含肽剪接活性并导致His-标记的MBP的形成。内含肽剪接是一个不可逆的过程,基于内含肽的片段互补系统的主要缺点是缺乏可逆性。The protein of interest is split into two inactive fragments, and small molecules of specific compounds can induce association of these fragments, thereby restoring the structure and function of the target protein. This process is also called fragment complementation. Muir and colleagues developed an intein-based conditional protein splicing system (Mootz and Muir, 2002). The intein is divided into N- and C-terminal halves. When present alone, both inteins are inactive, but once they are close to each other, an active domain is formed, and the proteins at both ends are spliced together to form an activity. protein. For example, two former fusions are between FKBP and maltose binding protein (MBP), which is fused between the FRB and His tags. These fusions dimerize under rapamycin induction and the intein domain is close, which restores intein splicing activity and leads to the formation of His-tagged MBP. Intein splicing is an irreversible process, and the major disadvantage of intein-based fragment-complementing systems is the lack of reversibility.
(3)利用小分子控制蛋白的稳定性(3) Using small molecules to control protein stability
大多数条件蛋白降解是利用泛素-蛋白酶体系统完成的,所以利用小分子调控蛋白降解过程中的各个环节,可实现蛋白稳定性调控。降解子(degron)是蛋白质的一部分,在调节蛋白质降解速率中起着重要作用。多数降解子介导的降解是通过泛素-蛋白酶体降解途径完成的,也有的是通过自噬途径完成的。目前已经鉴定出不少降解子序列,例如:ddFKBP是个不稳定结构域(DD domain),表达后很快被降解。但化学小分子shield-1可结合这个结构,使其稳定而不会被快速降解。将目标蛋白与ddFKBP结合,通过shield-1可有效调控蛋白的稳定和降解过程(Banaszynski et al.,2006)。类似地,ecDHFR也可以通过与trimethoprim的结合来调控自身的稳定性(Iwamoto et al.,2010;专利号:09487787)。Most conditional protein degradation is accomplished using the ubiquitin-proteasome system, so the regulation of protein stability can be achieved by using small molecules to regulate various aspects of protein degradation. Degradants (degrons) are part of proteins and play an important role in regulating the rate of protein degradation. Most degradation-mediated degradation is accomplished by the ubiquitin-proteasome degradation pathway, or by autophagy. A number of degrading sequences have been identified, for example: ddFKBP is an unstable domain (DD domain) that is rapidly degraded after expression. However, the chemical small molecule shield-1 can bind this structure to make it stable without rapid degradation. The target protein is bound to ddFKBP, and the stability and degradation process of the protein can be effectively regulated by shield-1 (Banaszynski et al., 2006). Similarly, ecDHFR can also regulate its own stability by binding to trimethoprim (Iwamoto et al., 2010; patent number: 09487787).
相反,有些结构域本身比较稳定,但在特定小分子作用下则被快速泛素化而降解。例如:植物激素结合结构域(AID)IAA17,可在吲哚乙酸的介导下结合TIR1,并被泛素连接酶识别,经一步被泛素化而降解(Nishimura et al.,2009;专利号:US20120115232)。Conversely, some domains are inherently stable, but are rapidly ubiquitinated and degraded by the action of specific small molecules. For example, the plant hormone binding domain (AID) IAA17 binds to TIR1 mediated by indoleacetic acid and is recognized by ubiquitin ligase and is degraded by ubiquitination in one step (Nishimura et al., 2009; :US20120115232).
综上所述,现有蛋白活性调控技术主要有:1.利用雷帕霉素调控FKBP与FRB的聚合与解离;2.利用脱落酸调控(PYR)与PYR1-like(PYL)的相互作用;3. 利用赤霉素调控CID与GAI的相互作用;4.利用内含肽实现两段蛋白的拼接;5.利用可调控的降解子实现融合蛋白的活性调控,如shield-1调控ddFKBP融合蛋白的活性,TMP调控DHFR融合蛋白的活性,吲哚乙酸调控AID融合蛋白的活性。In summary, the existing protein activity control technologies mainly include: 1. Using rapamycin to regulate the polymerization and dissociation of FKBP and FRB; 2. Using the interaction between abscisic acid regulation (PYR) and PYR1-like (PYL) 3. Using gibberellin to regulate the interaction between CID and GAI; 4. Using intein to achieve splicing of two-stage protein; 5. Using regulatable degradants to achieve regulation of fusion protein activity, such as shield-1 regulation of ddFKBP fusion The activity of the protein, TMP regulates the activity of the DHFR fusion protein, and the acetic acid regulates the activity of the AID fusion protein.
发明公开Invention disclosure
本发明要解决的技术问题是如何调控目标蛋白。The technical problem to be solved by the present invention is how to regulate the target protein.
为了解决该技术问题,本发明首先提供了一种小分子药物控制的蛋白功能开关系统。In order to solve this technical problem, the present invention first provides a small molecule drug controlled protein function switching system.
本发明提供的开关系统由操纵子原件和控制子原件构成,所述控制子为单个病毒蛋白酶,或由病毒蛋白酶与蛋白降解子融合而成的蛋白酶-降解子;所述操纵子位于目标蛋白内部,或位于目标蛋白与蛋白酶-降解子结构之间,由蛋白酶的一段连接肽构成;整个系统通过蛋白酶小分子的抑制剂化合物作为开关,实现对目标蛋白的调控。The switch system provided by the invention consists of an operon original and a control sub-unit, the control is a single viral protease, or a protease-degradant formed by fusion of a viral protease and a protein degradation agent; the operon is located inside the target protein Or located between the target protein and the protease-degrading structure, and consists of a ligation peptide of the protease; the whole system regulates the target protein through the inhibitor compound of the small molecule of the protease as a switch.
本发明所述的对目标蛋白的调控包括对目标蛋白的结构和/或功能和/或位置进行时空调控。所述结构和功能的调控又包括对目标蛋白的一级结构、三级结构、蛋白的表达、稳定性、功能活性、细胞内位置等方面进行调控。The regulation of the protein of interest according to the present invention involves air conditioning control of the structure and/or function and/or location of the protein of interest. The regulation of the structure and function includes regulating the primary structure, the tertiary structure, the expression of the protein, the stability, the functional activity, and the intracellular position of the target protein.
进一步的,所述开关系统还包括蛋白酶小分子的抑制剂化合物。Further, the switching system further comprises an inhibitor compound of a small molecule of a protease.
上述系统中,所述操纵子原件为可被所述蛋白酶识别并切割的连接肽;所述操纵子原件的个数为一个或两个或多个。In the above system, the operon element is a linker peptide recognizable and cleaved by the protease; the number of the operon elements is one or two or more.
进一步的,所述蛋白酶可为病毒蛋白酶,具体的,所述病毒蛋白酶可为丙肝病毒蛋白酶(NS3pro/4A)或与所述丙肝病毒蛋白酶NS3pro/4A具有70%或70%以上同源性的蛋白酶突变体或异构酶。NS3pro/4A蛋白酶的氨基酸序列为SEQ ID No.7。本发明将N34d蛋白中NS3pro和4A结构连接部分进行突变,得到NS3pro/4A蛋白酶,NS3pro/4A蛋白酶结构稳定,失去了降解子功能,但保留了完整的蛋白酶功能。N34d蛋白是将NS3/4A结构中去掉NS3RNA解旋酶区后,形成的不稳定易降解蛋白。Further, the protease may be a viral protease. Specifically, the viral protease may be a hepatitis C virus protease (NS3pro/4A) or a protease having 70% or more homology with the hepatitis C virus protease NS3pro/4A. Mutant or isomerase. The amino acid sequence of the NS3pro/4A protease is SEQ ID No. 7. The invention mutates the NS3pro and 4A structural linking portions of the N34d protein to obtain the NS3pro/4A protease, and the NS3pro/4A protease has stable structure and loses the function of the degradation, but retains the complete protease function. The N34d protein is an unstable and easily degradable protein formed by removing the NS3 RNA helicase region from the NS3/4A structure.
所述病毒蛋白酶还可以连入一段降解子结构域(degron),在两者共表达的情况下,融合蛋白会被快速降解,而无法发挥功能。降解子结构域可以是NS4A降解子,在与相应蛋白融合表达后,在极短的时间内降解,从而影响蛋白功能。所述蛋白酶-降解子为N34d蛋白,其氨基酸序列为SEQ ID No.6。降解子结构域也可以是由小分子调控的降解子,如ddFKBP或exDHFR,在结合相应小分子配体Shield-1或trimethoprim的条件下,两个结构域能被稳定,保持较长时间不被降解。在撤去配体后,这两个结构极不稳定,表达后,在极短的时间内降解。The viral protease can also be ligated into a degrading domain (degron). In the case of co-expression of the two, the fusion protein is rapidly degraded and cannot function. The degrading domain may be an NS4A degrader which, after being fused to the corresponding protein, degrades in a very short period of time, thereby affecting protein function. The protease-degrading protein is an N34d protein having an amino acid sequence of SEQ ID No. 6. The degrading domain can also be a small molecule-regulated degrader, such as ddFKBP or exDHFR. Under the conditions of binding the corresponding small molecule ligand Shield-1 or trimethoprim, the two domains can be stabilized for a long time without being degradation. After removal of the ligand, the two structures are extremely unstable and, after expression, degrade in a very short time.
进一步的,所述连接肽为下述序列之一:Further, the linker peptide is one of the following sequences:
TRIF:Pro Ser Ser Thr Pro Cys Ser Ala His Leu;TRIF: Pro Ser Ser Thr Pro Cys Ser Ala His Leu;
MAVS:Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;MAVS: Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;
3-4A:Asp Leu Glu Val Val Thr Ser Thr Trp Val;3-4A: Asp Leu Glu Val Val Thr Ser Thr Trp Val;
4A4B:Asp Glu Met Glu Glu Cys Ser Gln His Leu;4A4B: Asp Glu Met Glu Glu Cys Ser Gln His Leu;
4B5A:Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;4B5A: Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
5A5B:Glu Asp Val Val Cys Cys Ser Met Ser Tyr。5A5B: Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
进一步的,所述蛋白酶小分子的抑制剂化合物为下述化合物之一:Telaprevir(CAS Number:402957-28-2);Boceprevir(CAS Number:394730-60-0);Simeprevir(CAS Number:923604-59-5);Faldaprevir(CAS Number:801283-95-4);Asunaprevir(CAS Number:630420-16-5);Furaprevir(CAS Number:1435923-88-8)。Further, the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir (CAS Number: 402957-28-2); Boceprevir (CAS Number: 394730-60-0); Simeprevir (CAS Number: 923604- 59-5); Faldaprevir (CAS Number: 801283-95-4); Asunaprevir (CAS Number: 630420-16-5); Furaprevir (CAS Number: 1435923-88-8).
进一步的,所述降解子为NS3/4A不稳定区域、ddFKBP不稳定区域或ecDHFR不稳定区域。Further, the degradation agent is an NS3/4A unstable region, a ddFKBP unstable region, or an ecDHFR unstable region.
进一步的,所述目标蛋白为单个蛋白或融合蛋白。每个蛋白可以为具有1个或若干结构域的蛋白,或由多个功能蛋白融合而成的融合蛋白。所述操纵子原件可以位于目标蛋白内部(如结构域之间)、也可以位于融合部分之间。具体的,所述目标蛋白可为荧光蛋白、耐药基因、抗体、细胞因子和抗原识别分子中的任意一种。Further, the target protein is a single protein or a fusion protein. Each protein may be a protein having one or several domains, or a fusion protein composed of a plurality of functional proteins. The operon element may be located inside the target protein (such as between domains) or between the fusion moieties. Specifically, the target protein may be any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
若目标蛋白为单个蛋白时,所述目标蛋白、所述控制子原件和所述操纵子原件构成融合蛋白,所述操纵子原件用于连接所述目标蛋白和所述控制子原件;所述控制子原件为蛋白酶-降解子;If the target protein is a single protein, the target protein, the control subunit, and the operon element constitute a fusion protein, the operon element is used to link the target protein and the control sub-assembly; the control The sub-origin is a protease-degrading agent;
若目标蛋白为具有若干结构域或若干功能蛋白构成的融合蛋白时,可在各个结构域或功能蛋白之间插入病毒蛋白酶识别序列。所述目标蛋白、所述控制子原件和所述操纵子原件构成融合蛋白,所述操纵子原件用于连接所述目标蛋白和所述控制子原件,或用于连接所述目标蛋白的结构域或功能蛋白。If the target protein is a fusion protein having several domains or several functional proteins, a viral protease recognition sequence can be inserted between each domain or a functional protein. The target protein, the control subunit, and the operon element constitute a fusion protein, the operon element is used to link the target protein and the control subunit, or to link a domain of the target protein Or functional protein.
在一些实施方案中,目标蛋白可以是两个不同荧光蛋白的融合蛋白,连接肽位于两个不同荧光蛋白之间,连接两个不同荧光蛋白,在蛋白酶活性被抑制的情况下,荧光蛋白融合表达,会在细胞内共定位,在蛋白酶活性状态下,融合表达的荧光蛋白,被蛋白酶剪切为两个蛋白,分开表达。或是具有多个结构域的嵌合抗原受体(CAR),连接肽位于不同结构域之间,连接不同的结构域,使嵌合抗原受体各个结构域串联,在蛋白酶活性被抑制的情况下,嵌合抗原受体在T细胞中表达,并具有识别肿瘤抗原传递信号,激活T细胞功能的作用,在蛋白酶活性状态下,嵌合抗原受体各个结构域被切割为两个或多个独立结构,影响T细胞识别肿瘤抗原和传递信号。In some embodiments, the protein of interest may be a fusion protein of two different fluorescent proteins, the linker peptide being located between two different fluorescent proteins, joining two different fluorescent proteins, and fluorescent protein fusion expression in the case where protease activity is inhibited It will co-localize in the cell. Under the protease active state, the fluorescent protein expressed by fusion will be cleaved into two proteins by protease and expressed separately. Or a chimeric antigen receptor (CAR) having multiple domains, the linker peptides being located between different domains, linking different domains, allowing the various domains of the chimeric antigen receptor to be cascaded, and the protease activity is inhibited. The chimeric antigen receptor is expressed in T cells and has the function of recognizing a tumor antigen delivery signal and activating T cell function. In the protease active state, each domain of the chimeric antigen receptor is cleaved into two or more Independent structure that affects T cells to recognize tumor antigens and transmit signals.
目标蛋白可以和病毒蛋白酶融合表达,也可以分别表达。融合表达可以使病毒蛋白酶与目标蛋白空间位置更近,更快地行使功能。分开表达可减少对目标蛋白结构的影响,更准确地调控蛋白活性。The target protein can be expressed in fusion with viral proteases or separately. Fusion expression allows the viral protease to be located closer to the target protein and function faster. Separate expression reduces the effect on the structure of the target protein and more precisely regulates protein activity.
进一步的,所述蛋白功能开关系统用于真核哺乳动物细胞中。Further, the protein functional switching system is used in eukaryotic mammalian cells.
为了解决上述技术问题,本发明又提供了用于表达蛋白功能开关系统的核酸分子、质粒、细胞系以及包含上述元素装配而成的试剂盒。In order to solve the above technical problems, the present invention further provides a nucleic acid molecule, a plasmid, a cell line, and a kit comprising the above elements for expressing a protein function switching system.
进一步的,所述核酸分子为SEQ ID No.1、SEQ ID No.2、SEQ ID No.3、SEQ ID  No.4或SEQ ID No.5所示的DNA分子;所述质粒为含有所述核酸分子的表达载体,所述表达载体具体可为慢病毒载体Pltr载体;所述细胞系为含有所述质粒的细胞系,所述细胞系具体可为293T细胞或3T3细胞。Further, the nucleic acid molecule is a DNA molecule represented by SEQ ID No. 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4 or SEQ ID No. 5; An expression vector for a nucleic acid molecule, which may specifically be a lentiviral vector Pltr vector; the cell line is a cell line containing the plasmid, and the cell line may specifically be a 293T cell or a 3T3 cell.
为了解决上述技术问题,本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒的新用途。In order to solve the above technical problems, the present invention also provides a novel use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit.
本发明提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在调控目标蛋白结构和/或功能中的应用。The invention provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the structure and/or function of a protein of interest.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白结构和/或功能的产品中的应用。The invention also provides the use of a protein functional switch system or nucleic acid molecule, plasmid, cell line or kit as described above for the preparation of a product that modulates the structure and/or function of a protein of interest.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在调控目标蛋白活性中的应用。The invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the activity of a target protein.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白活性的产品中的应用。The invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the activity of a target protein.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在调控目标蛋白稳定性中的应用。The invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the stability of a target protein.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白稳定性的产品中的应用。The invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the stability of a target protein.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在调控目标蛋白的时空位置中的应用。The invention also provides the use of the above protein function switch system or nucleic acid molecule, plasmid, cell line or kit for regulating the spatiotemporal position of a target protein.
本发明还提供了上述蛋白功能开关系统或核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白的时空位置的产品中的应用。The invention also provides the use of the above protein functional switch system or nucleic acid molecule, plasmid, cell line or kit for the preparation of a product for regulating the spatiotemporal position of a target protein.
为了解决上述技术问题,本发明最后提供了一种调控目标蛋白的方法。In order to solve the above technical problems, the present invention finally provides a method of regulating a target protein.
本发明提供的调控目标蛋白的方法包括如下步骤:在受体细胞中表达出包含控制子原件、操纵子原件和目标蛋白的融合蛋白,再利用蛋白酶小分子的抑制剂化合物来调控所述控制子原件的活性,进而实现对目标蛋白的调控;The method for regulating a target protein provided by the present invention comprises the steps of: expressing a fusion protein comprising a promoter subgenus, an operon element and a target protein in a recipient cell, and then using an inhibitor compound of a protease small molecule to regulate the controller The activity of the original, thereby achieving the regulation of the target protein;
所述控制子原件为单个病毒蛋白酶,或由病毒蛋白酶与蛋白降解子融合而成的蛋白酶-降解子;所述操纵子原件为所述蛋白酶可识别并切割的连接肽。The control element is a single viral protease, or a protease-degradant fused by a viral protease and a protein degrader; the operon element is a linker peptide that the protease recognizes and cleaves.
上述方法中,所述操纵子原件的个数为一个或两个或多个。In the above method, the number of the operon originals is one or two or more.
上述方法中,所述病毒蛋白酶为丙肝病毒蛋白酶。所述丙肝病毒蛋白酶具体为丙肝病毒蛋白酶NS3pro/4A或与丙肝病毒蛋白酶NS3pro/4A具有70%或70%以上同源性的蛋白酶突变体或异构酶。In the above method, the viral protease is a hepatitis C virus protease. The hepatitis C virus protease is specifically a hepatitis C virus protease NS3pro/4A or a protease mutant or isomerase having 70% or more homology with the hepatitis C virus protease NS3pro/4A.
上述方法中,所述连接肽为下述序列之一:In the above method, the linker peptide is one of the following sequences:
TRIF:Pro Ser Ser Thr Pro Cys Ser Ala His Leu;TRIF: Pro Ser Ser Thr Pro Cys Ser Ala His Leu;
MAVS:Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;MAVS: Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;
3-4A:Asp Leu Glu Val Val Thr Ser Thr Trp Val;3-4A: Asp Leu Glu Val Val Thr Ser Thr Trp Val;
4A4B:Asp Glu Met Glu Glu Cys Ser Gln His Leu;4A4B: Asp Glu Met Glu Glu Cys Ser Gln His Leu;
4B5A:Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;4B5A: Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
5A5B:Glu Asp Val Val Cys Cys Ser Met Ser Tyr。5A5B: Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
上述方法中,所述蛋白酶小分子的抑制剂化合物为下述化合物之一:Telaprevir(CAS Number:402957-28-2);Boceprevir(CAS Number:394730-60-0);Simeprevir(CAS Number:923604-59-5);Faldaprevir(CAS Number:801283-95-4);Asunaprevir(CAS Number:630420-16-5);Furaprevir(CAS Number:1435923-88-8)。In the above method, the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir (CAS Number: 402957-28-2); Boceprevir (CAS Number: 394730-60-0); Simeprevir (CAS Number: 923604) -59-5); Faldaprevir (CAS Number: 801283-95-4); Asunaprevir (CAS Number: 630420-16-5); Furaprevir (CAS Number: 1435923-88-8).
上述方法中,所述降解子为NS3/4A不稳定区域、ddFKBP不稳定区域或ecDHFR不稳定区域。所述蛋白酶-降解子为N43d蛋白。In the above method, the proton is an NS3/4A unstable region, a ddFKBP unstable region, or an ecDHFR unstable region. The protease-degrading agent is an N43d protein.
上述方法中,所述目标蛋白为单个蛋白或融合蛋白。每个蛋白可以为具有1个或若干结构域的蛋白,或由多个功能蛋白融合而成的融合蛋白。所述操纵子原件可以位于目标蛋白内部(如结构域之间)、也可以位于融合部分之间。具体的,所述目标蛋白可为荧光蛋白、耐药基因、抗体、细胞因子和抗原识别分子中的任意一种。In the above method, the target protein is a single protein or a fusion protein. Each protein may be a protein having one or several domains, or a fusion protein composed of a plurality of functional proteins. The operon element may be located inside the target protein (such as between domains) or between the fusion moieties. Specifically, the target protein may be any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
上述方法中,所述在受体细胞中表达出包含控制子原件、操纵子原件和目标蛋白的融合蛋白,再利用蛋白酶小分子的抑制剂化合物来调控所述控制子原件的活性的方法为将表达所述控制子原件、所述操纵子原件和所述目标蛋白的载体导入所述受体细胞中,得到重组细胞;在培养体系中培养所述重组细胞,得到所述融合蛋白;通过在所述培养体系中的添加或去除所述抑制剂化合物可实现调控所述控制子原件的活性。具体方法可为如下(1)或(2):In the above method, the method of expressing the fusion protein comprising the promoter element, the operon element and the target protein in the recipient cell, and then using the inhibitor compound of the protease small molecule to regulate the activity of the control element is a vector expressing the control element, the operon element, and the target protein is introduced into the recipient cell to obtain a recombinant cell; the recombinant cell is cultured in a culture system to obtain the fusion protein; Addition or removal of the inhibitor compound in the culture system can effect modulation of the activity of the control element. The specific method can be as follows (1) or (2):
(1)包括如下步骤:(1) Includes the following steps:
(1-1)将表达目标蛋白、蛋白酶与降解子融合而成的蛋白酶-降解子和所述蛋白酶识别和可切割的连接肽的载体与所述蛋白酶的小分子抑制剂共同转染受体细胞中,得到重组细胞;在重组细胞培养体系中培养所述重组细胞,表达得到融合蛋白,由于所述抑制剂抑制所述融合蛋白中蛋白酶的识别和切割活性,此时降解子将所述融合蛋白降解,所述融合蛋白中的目标蛋白失去活性;(1-1) co-transfecting a receptor-derived cell with a protease-degradant expressing a target protein, a protease and a proteosome, and a vector of the protease-recognizing and cleavable linker peptide together with the small molecule inhibitor of the protease Recombinant cells are obtained; the recombinant cells are cultured in a recombinant cell culture system, and a fusion protein is expressed, wherein the inhibitor inhibits the recognition and cleavage activity of the protease in the fusion protein, and the degradant converts the fusion protein Degrading, the target protein in the fusion protein is inactivated;
(1-2)去除所述重组细胞培养体系中的小分子抑制剂,培养所述重组细胞,表达得到融合蛋白,由于所述蛋白酶具有识别并切割所述连接肽的活性,将所述融合蛋白中连接肽切割,所述目标蛋白与所述蛋白酶-降解子分开,所述目标蛋白正常表达,具有活性;(1-2) removing a small molecule inhibitor in the recombinant cell culture system, culturing the recombinant cell, expressing a fusion protein, and the fusion protein is obtained by the protease having the activity of recognizing and cleaving the linker peptide The ligated peptide is cleaved, the target protein is separated from the protease-degrading agent, and the target protein is normally expressed and active;
所述融合蛋白由所述目标蛋白、所述蛋白酶-降解子和用于连接二者的所述连接肽组成;The fusion protein consists of the target protein, the protease-degradant, and the linker peptide for ligation;
(2)包括如下步骤:(2) Includes the following steps:
(2-1)将表达目标蛋白、蛋白酶和所述蛋白酶识别和可切割的连接肽的载体与所述蛋白酶的小分子抑制剂共同转染受体细胞中,在重组细胞培养体系中培养所述重组细胞,表达得到融合蛋白,由于所述抑制剂抑制所述融合蛋白中蛋白酶的识别和切割活性,所述目标蛋白正常表达,具有活性;(2-1) co-transfecting a vector expressing a target protein, a protease, and the protease-recognizing and cleavable linker peptide with a small molecule inhibitor of the protease into a recipient cell, and culturing the recombinant cell culture system Recombinant cells, expressing a fusion protein, wherein the target protein is normally expressed and active because the inhibitor inhibits the recognition and cleavage activity of the protease in the fusion protein;
(2-2)去除所述重组细胞培养体系中的小分子抑制剂,培养所述重组细胞, 表达得到融合蛋白,由于所述蛋白酶具有识别并切割所述连接肽的活性,将所述目标蛋白中连接肽切割,所述目标蛋白失去活性。(2-2) removing the small molecule inhibitor in the recombinant cell culture system, culturing the recombinant cell, expressing a fusion protein, and the target protein is obtained by the protease having the activity of recognizing and cleaving the linker peptide The linker peptide is cleaved and the target protein is inactivated.
上述方法中,所述表达所述控制子原件、所述操纵子原件和所述目标蛋白的载体为将含有所述控制子原件的编码基因、所述操纵子原件的编码基因和所述目标蛋白的编码基因的片段插入表达载体的多克隆位点得到的。所述含有所述控制子原件的编码基因、所述操纵子原件的编码基因和所述目标蛋白的编码基因的片段具体可为SEQ ID No.1或SEQ ID No.2或SEQ ID No.3或SEQ ID No.4或SEQ ID No.5所示的DNA分子。所述表达载体具体可为慢病毒载体Pltr载体。所述受体细胞具体可为293T细胞或3T3细胞。In the above method, the vector expressing the control subunit, the operon original, and the target protein is a coding gene containing the control subunit, a coding gene of the operon original, and the target protein. A fragment of the coding gene is inserted into the multiple cloning site of the expression vector. The coding gene containing the control subunit, the coding gene of the operon original, and the fragment of the coding gene of the target protein may specifically be SEQ ID No. 1 or SEQ ID No. 2 or SEQ ID No. 3. Or the DNA molecule shown in SEQ ID No. 4 or SEQ ID No. 5. The expression vector may specifically be a lentiviral vector Pltr vector. The recipient cell may specifically be a 293T cell or a 3T3 cell.
经过本发明研究和实验表明:将表达目标蛋白(包括:功能蛋白、融合蛋白或基因工程蛋白)、病毒蛋白酶(或经过改造的病毒蛋白酶)和病毒蛋白酶识别和可切割的连接肽的载体共转染到哺乳动物细胞中,通过添加和去除相应蛋白酶的小分子抑制剂可调控蛋白酶的活性,而蛋白酶的表达会影响目标蛋白的功能,从而间接控制目标蛋白的功能。本发明的病毒蛋白酶可以是天然的蛋白酶,也可以经过遗传改造,减小蛋白酶分子量的蛋白酶,或是与降解子结构域(degron)融合而成的蛋白酶-降解子。病毒蛋白酶抑制剂主要为市面上用于治疗或进入临床实验的药物。本发明制备了病毒蛋白酶依赖的小分子药物用来调控目标蛋白功能的开关系统,提出了一种快速、高效、可逆、可控、简单、经济和具有良好应用前景的蛋白功能调控方法。Through the research and experiments of the present invention, it is shown that the vector expressing the target protein (including: functional protein, fusion protein or genetic engineering protein), viral protease (or engineered viral protease) and viral protease recognition and cleavable linker peptide is co-transformed. In mammalian cells, the activity of the protease can be regulated by the addition and removal of small molecule inhibitors of the corresponding proteases, and the expression of the protease affects the function of the target protein, thereby indirectly controlling the function of the target protein. The viral protease of the present invention may be a natural protease, a protease that is genetically engineered to reduce the molecular weight of the protease, or a protease-degradant fused to a degrading domain (degron). Viral protease inhibitors are primarily drugs that are commercially available for treatment or for clinical trials. The invention prepares a switching system of a virus protease-dependent small molecule drug for regulating the function of a target protein, and proposes a protein function regulation method which is fast, efficient, reversible, controllable, simple, economical and has good application prospect.
附图说明DRAWINGS
图1是本发明实施例1中的SwichOFF系统--绿色荧光蛋白表达载体图谱。BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a diagram showing the SwichOFF system-green fluorescent protein expression vector in Example 1 of the present invention.
图2是本发明实施例1中的小分子ASV调控绿色荧光蛋白表达。上图:不加ASV时,绿色荧光蛋白稳定表达;下图:加入ASV后绿色荧光蛋白被降解,而不表达。Fig. 2 is a diagram showing the expression of green fluorescent protein by small molecule ASV in Example 1 of the present invention. Above: Green fluorescent protein is stably expressed when no ASV is added; the following figure: Green fluorescent protein is degraded and not expressed after adding ASV.
图3是本发明实施例1中的荧光蛋白表达的快速切换。上图:加入ASV后24小时,绿色荧光迅速减弱。下图:去除ASV后,荧光快速稳定表达。Figure 3 is a rapid switching of fluorescent protein expression in Example 1 of the present invention. Above: 24 hours after the addition of ASV, green fluorescence rapidly diminished. Bottom: After removal of ASV, fluorescence is rapidly and stably expressed.
图4是本发明实施例2中的SwichON系统--融合荧光蛋白表达载体图谱。Figure 4 is a diagram showing the SwichON system-fusion fluorescent protein expression vector in Example 2 of the present invention.
图5是本发明实施例2中的SwichON系统调控荧光蛋白在细胞内的定位。第一列为红色荧光,第二列为绿色荧光,第三列为荧光重叠图。第一排:不加ASV,绿色荧光表达于细胞质;第二列:加入ASV,绿色荧光表达于线粒体。Figure 5 is a diagram showing the localization of a fluorescent protein in a cell by the SwichON system in Example 2 of the present invention. The first column is red fluorescence, the second column is green fluorescence, and the third column is fluorescence overlay. The first row: no ARV, green fluorescence is expressed in the cytoplasm; the second column: adding ASV, green fluorescence is expressed in mitochondria.
图6是本发明实施例2中的SwichON系统调控快速切换融合荧光蛋白在细胞内的定位。第一列为红色荧光,第二列为绿色荧光,第三列为荧光重叠图。加入ASV后24小时,绿色荧光迅速从细胞质定位到线粒体。Figure 6 is a diagram showing the localization of the fast-switching fusion fluorescent protein in the SwichON system of Example 2 of the present invention. The first column is red fluorescence, the second column is green fluorescence, and the third column is fluorescence overlay. 24 hours after the addition of ASV, green fluorescence rapidly localized from the cytoplasm to the mitochondria.
图7是本发明实施例2中的SwichON系统调控快速切换融合荧光蛋白在细胞内的定位。第一列为红色荧光,第二列为绿色荧光,第三列为荧光重叠图。去除ASV后24小时,绿色荧光迅速从线粒体释放到细胞质。Figure 7 is a diagram showing the localization of the fast-switching fusion fluorescent protein in the SwichON system of Example 2 of the present invention. The first column is red fluorescence, the second column is green fluorescence, and the third column is fluorescence overlay. 24 hours after the removal of ASV, green fluorescence was rapidly released from the mitochondria to the cytoplasm.
图8是本发明实施例3中的SwichOFF系统CAR调控载体图谱。Figure 8 is a map of the SwichOFF system CAR regulation carrier in the third embodiment of the present invention.
图9是本发明实施例3中的SwichOFF系统调控CAR表达原理图。Fig. 9 is a schematic diagram showing the regulation of CAR expression by the SwichOFF system in the third embodiment of the present invention.
图10是本发明实施例4中的SwichON系统CAR调控载体图谱。Figure 10 is a map of the SwichON system CAR regulatory vector in Example 4 of the present invention.
图11是本发明实施例4中的SwichON系统调控CAR功能原理图。Fig. 11 is a schematic diagram showing the function of regulating the CAR of the SwichON system in the fourth embodiment of the present invention.
图12是本发明实施例5中的SwichON系统CAR调控载体图谱。Figure 12 is a map of the SwichON system CAR regulatory vector in Example 5 of the present invention.
图13是本发明实施例5中的SwichON系统调控CAR功能原理图。Figure 13 is a schematic diagram showing the function of the SwichON system for regulating CAR in the fifth embodiment of the present invention.
图14是本发明实施例6中的western blot检测Switch蛋白调控系统。上图:fCAR-V1、fCAR-V2、fCAR-V3结构示意图;下图:western blot检测flag标签融合蛋白大小。fCAR-V1组:第一列:不加ASV时,表达一段63kd大小的Flag-CAR片段。第二列:加入ASV后,融合蛋白快速降解。fCAR-V2组:第一列:不加ASV时,检测到大小45kd的Flag-CAR1蛋白。第二列:加入ASV后,anti-flag抗体检测到97kd的完整蛋白。fCAR-V3组:第一列:不加ASV时,表达大小33kd的Flag-CAR1蛋白。第二列:加入ASV后,anti-flag抗体检测到97kd的完整蛋白。由于CAR的hinge部位自身可能自然断裂,所以所有组都能检测到一条大小33kd的蛋白。Figure 14 is a Western blot analysis of the Switch protein regulatory system in Example 6 of the present invention. Above: Schematic diagram of fCAR-V1, fCAR-V2, and fCAR-V3; bottom panel: Western blot analysis of flag tag fusion protein size. fCAR-V1 group: The first column: a section of 63kd Flag-CAR fragment is expressed without ASV. Second column: After the addition of ASV, the fusion protein rapidly degraded. fCAR-V2 group: First column: Flag-CAR1 protein of 45 kd in size was detected without ASV. Second column: After adding ASV, the anti-flag antibody detected 97kd of intact protein. fCAR-V3 group: The first column: the Flag-CAR1 protein with a size of 33 kd was expressed without ASV. Second column: After adding ASV, the anti-flag antibody detected 97kd of intact protein. Since the hinge site of CAR itself may naturally break, a group of 33kd protein can be detected in all groups.
图15为嵌合抗原受体杀伤功能测试结果。Figure 15 shows the results of the chimeric antigen receptor killing function test.
实施发明的最佳方式The best way to implement the invention
下述实施例中所使用的实验方法如无特殊说明,均为常规方法。The experimental methods used in the following examples are conventional methods unless otherwise specified.
下述实施例中所用的材料、试剂等,如无特殊说明,均可从商业途径得到。The materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
实施例1:小分子药物调控绿色荧光蛋白的活性(SwichOFF系统)Example 1: Small molecule drug modulates the activity of green fluorescent protein (SwichOFF system)
本实施例中,将绿色荧光蛋白与自降解的HCV蛋白酶融合表达,由HCV蛋白酶抑制剂ASV通过抑制蛋白酶活性,来调控绿色荧光蛋白的表达。In this embodiment, the green fluorescent protein is expressed by fusion with the self-degrading HCV protease, and the expression of the green fluorescent protein is regulated by the HCV protease inhibitor ASV by inhibiting the protease activity.
①载体构建:1 carrier construction:
将SEQ ID No.1所示的融合表达基因插入慢病毒载体Pltr载体(addgen,25870),得到重组载体Pltr-sfGFP-N3N4,SEQ ID No.1第1-714位为sfGFP基因,第721-753位为HCV病毒蛋白酶识别序列4A4B(DEMEECSQHLP)的编码基因,第842-1635位为N34d基因(带有NS3/4A序列和一段降解子序列)。N34d基因表达的N34d蛋白的氨基酸序列为SEQ ID No.6。NS3/4A结构中包含NS3pro(蛋白酶区)、NS3RNA解旋酶区和4A区。N34d蛋白是将NS3/4A结构中去掉NS3RNA解旋酶区后,形成的不稳定易降解蛋白。The fusion expression gene represented by SEQ ID No. 1 was inserted into a lentiviral vector Pltr vector (addgen, 25870) to obtain a recombinant vector Pltr-sfGFP-N3N4, and positions 1-714 of SEQ ID No. 1 were sfGFP gene, 721- The 753 is the coding gene of the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), and the 842-1635 is the N34d gene (with the NS3/4A sequence and a stretcher sequence). The amino acid sequence of the N34d protein expressed by the N34d gene is SEQ ID No. 6. The NS3/4A structure contains NS3pro (protease region), NS3 RNA helicase region and 4A region. The N34d protein is an unstable and easily degradable protein formed by removing the NS3 RNA helicase region from the NS3/4A structure.
重组载体Pltr-sfGFP-N3N4的结构如图1所示。The structure of the recombinant vector Pltr-sfGFP-N3N4 is shown in Fig. 1.
②细胞转染:2 cell transfection:
复苏并培养3T3细胞(北纳生物,BNCC339681)。转染前一天,在24孔板中每孔加入5×10 5个细胞。转染前换液,按照是否加入小分子抑制剂Asunaprevir(ASV)(阿拉丁,A126103)分为如下两组: Resuscitation and culture of 3T3 cells (Northern Biology, BNCC339681). One day before transfection, 5 x 10 5 cells were added to each well in a 24-well plate. The transfusion before transfection was divided into the following two groups according to whether or not the small molecule inhibitor Asunaprevir (ASV) (Aladdin, A126103) was added:
ASV 0uM组:一个孔加入不含ASV的培养基(含10%血清(BI)的DMEM(GBICO));ASV 0uM group: one well was added to the medium without ASV (DMEM (GBICO) containing 10% serum (BI));
ASV 1uM组:一个孔加入含1uM ASV的培养基。ASV 1uM group: One well was added to the medium containing 1 uM ASV.
具体转染步骤如下:1)取两支1.5ml EP管,分别加入50uL  培养 基,然后在其中一支管中加入3.0uL Lipofectamine
Figure PCTCN2017118970-appb-000002
3000试剂(Thermo Fisher),另一支管中加入2ug DNA质粒(重组载体Pltr-sfGFP-N3N4),分别混匀。2)将两个EP管中的液体混合为一管,共得到100uL的混合物,轻轻吹打混匀15次,室温静置5分钟。3)分别将50uL的混合物加到ASV 0uM组和ASV 1uM组对应的孔中,6h后换液。
The specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 50uL respectively. Medium, then add 3.0uL Lipofectamine to one of the tubes
Figure PCTCN2017118970-appb-000002
3000 reagent (Thermo Fisher), 2 ug DNA plasmid (recombinant vector Pltr-sfGFP-N3N4) was added to the other tube, and mixed separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 100 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 50 uL of the mixture to the corresponding wells of the ASV 0uM group and the ASV 1uM group, respectively, and change the solution after 6 hours.
转染后24h,观察细胞表达荧光的情况。结果如图2所示。从图中可以看出:第一排:不加ASV(ASV 0uM组),表达的融合蛋白sfGFP-4A4B-NS3/4A-Degron。蛋白酶具有切割功能,在4A4B处将融合蛋白切断。然后,Degron介导NS3/4A蛋白酶快速降解,而不影响sfGFP的功能。第二排:加入ASV(ASV 1uM组),3T3细胞同样表达融合蛋白sfGFP-4A4B-NS3/4A-Degron。但加入ASV抑制了NS3/4A蛋白酶对4A4B的识别和切割,使得Degron将整个融合蛋白带入泛素-蛋白酶体途径,进行快速降解。sfGFP被快速降解,使细胞不能表达绿色荧光蛋白。说明ASV可调控目标蛋白的表达。At 24 hours after transfection, the cells were observed to express fluorescence. The result is shown in Figure 2. It can be seen from the figure: the first row: the fusion protein sfGFP-4A4B-NS3/4A-Degron expressed without ASV (ASV 0uM group). The protease has a cleavage function and the fusion protein is cleaved at 4A4B. Degron then mediates rapid degradation of the NS3/4A protease without affecting the function of sfGFP. The second row: ASV (ASV 1uM group) was added, and the 3T3 cells also expressed the fusion protein sfGFP-4A4B-NS3/4A-Degron. However, the addition of ASV inhibited the recognition and cleavage of 4A4B by the NS3/4A protease, allowing Degron to bring the entire fusion protein into the ubiquitin-proteasome pathway for rapid degradation. sfGFP is rapidly degraded, rendering cells unable to express green fluorescent protein. This indicates that ASV can regulate the expression of target proteins.
③小分子药物调控:3 small molecule drug regulation:
转染24h后,将ASV 0uM组细胞培养基吸去,加入新鲜的含1uM的ASV的培养基;After transfection for 24 h, the cell culture medium of ASV 0uM group was aspirated, and fresh medium containing 1 uM of ASV was added;
转染24h后,将ASV 1uM组细胞培养基吸去,用PBS清洗两遍,然后加入新鲜的不含ASV的培养基。After 24 h of transfection, the ASV 1uM cell culture medium was aspirated, washed twice with PBS, and then fresh ASV-free medium was added.
加入ASV和去除ASV 24h后观察细胞表达荧光的情况。结果如图3所示。从图中可以看出:第一排:不加ASV,绿色荧光蛋白在细胞中表达非常高,加入ASV后24h,绿色荧光蛋白的表达量急剧下降,到了非常微弱的水平。第二排:在加ASV的条件下,细胞不能表达或仅少量细胞表达极弱的绿色荧光蛋白,去ASV后24h,绿色荧光蛋白在细胞中表达又恢复到高水平。说明ASV可快速切换目标蛋白的表达。The cells were observed to express fluorescence after adding ASV and removing ASV for 24 h. The result is shown in Figure 3. It can be seen from the figure: The first row: without the addition of ASV, the expression of green fluorescent protein is very high in the cells, and the expression of green fluorescent protein drops sharply at 24 h after the addition of ASV, to a very weak level. The second row: under the condition of adding ASV, the cells could not express or only a few cells expressed very weak green fluorescent protein. After 24 hours of ASV, the expression of green fluorescent protein in the cells recovered to a high level. This indicates that ASV can quickly switch the expression of the target protein.
实施例2:小分子药物调控融合荧光蛋白的定位(SwichON系统)Example 2: Localization of Fusion Proteins by Small Molecule Drugs (SwichON System)
本实施例中,将绿色荧光蛋白、红色荧光蛋白与自降解的HCV蛋白酶融合表达,由HCV蛋白酶抑制剂ASV通过抑制蛋白酶活性,来调控绿色荧光蛋白和红色荧光蛋白在细胞中的定位。In this embodiment, the green fluorescent protein, the red fluorescent protein and the self-degrading HCV protease are fused and expressed, and the HCV protease inhibitor ASV inhibits the localization of the green fluorescent protein and the red fluorescent protein by inhibiting the protease activity.
①载体构建:1 carrier construction:
将SEQ ID No.2所示的融合蛋白基因插入慢病毒载体Pltr载体,得到重组载体Pltr-tomDsRed-N3N4-GFP V2,SEQ ID No.2第1-105位为Tom20基因的线粒体膜定位信号的编码基因,第118-789位为DsRed ex基因,第808-840位为HCV病毒蛋白酶识别序列4A4B(DEMEECSQHLP)的编码基因,第880-1590位为sfGFP基因,第1606-1629位为flag标签序列,第1660-2352位为NS3pro/4A蛋白酶基因。The fusion protein gene represented by SEQ ID No. 2 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-tomDsRed-N3N4-GFP V2, and the 1-10th position of SEQ ID No. 2 was the mitochondrial membrane localization signal of the Tom20 gene. The coding gene, the 118th-789th position is the DsRed ex gene, the 808-840 is the coding gene of the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), the 880-1590 is the sfGFP gene, and the 1606-1629 bits are the flag tag sequence. The 1660-2352 position is the NS3pro/4A protease gene.
NS3pro/4A蛋白酶基因编码的NS3pro/4A蛋白酶的氨基酸序列为SEQ ID No.7。NS3pro/4A蛋白酶是将N34d蛋白中NS3pro和4A结构连接部分进行突变后得到的蛋白酶。NS3pro/4A蛋白酶结构稳定,失去了降解子功能,但保留了完整的蛋白酶 功能。The amino acid sequence of the NS3pro/4A protease encoded by the NS3pro/4A protease gene is SEQ ID No. 7. The NS3pro/4A protease is a protease obtained by mutating the NS3pro and 4A structural linking portions of the N34d protein. The NS3pro/4A protease is structurally stable and loses its proteolytic function, but retains intact protease function.
Tom20基因的线粒体膜定位信号序列如下:Met Val Gly Arg Asn Ser Ala Ile Ala Ala Gly Val Cys Gly Ala Leu Phe Ile Gly Tyr Cys Ile Tyr Phe Asp Arg Lys Arg Arg Ser Asp Pro Asn Phe Lys。The mitochondrial membrane localization signal sequence of the Tom20 gene is as follows: Met Val Gly Arg Asn Ser Ala Ile Ala Ala Gly Val Cys Gly Ala Leu Phe Ile Gly Tyr Cys Ile Tyr Phe Asp Arg Lys Arg Arg Ser Asp Pro Asn Phe Lys.
flag标签序列如下:Asp Tyr Lys Asp Asp Asp Asp Lys。The flag tag sequence is as follows: Asp Tyr Lys Asp Asp Asp Asp Lys.
重组载体Pltr-tomDsRed-N3N4-GFP V2的结构如图4所示。The structure of the recombinant vector Pltr-tomDsRed-N3N4-GFP V2 is shown in Fig. 4.
②细胞转染:2 cell transfection:
复苏并培养3T3细胞。转染前一天,在24孔板每孔中加入5×10 5个细胞。转染前换液,按照是否加入小分子抑制剂Asunaprevir(ASV)分为如下两组: Resuscitate and culture 3T3 cells. One day before transfection, 5 x 10 5 cells were added to each well of a 24-well plate. The liquid exchange before transfection was divided into the following two groups according to whether or not the small molecule inhibitor Asunaprevir (ASV) was added:
ASV 0uM组:一个孔加入不含ASV的培养基;ASV 0uM group: one well was added to the medium without ASV;
ASV 1uM组:一个孔加入含1uM ASV的培养基。ASV 1uM group: One well was added to the medium containing 1 uM ASV.
具体转染步骤如下:1)取两支1.5ml EP管,分别加入50uL 
Figure PCTCN2017118970-appb-000003
培养基,然后在其中一支管中加入3.0uL Lipofectamine
Figure PCTCN2017118970-appb-000004
3000试剂(Thermo Fisher),另一支管中加入2ug DNA质粒(重组载体Pltr-sfGFP-N3N4),分别混匀。2)将两个EP管中的液体混合为一管,共得到100uL的混合物,轻轻吹打混匀15次,室温静置5分钟。3)分别将50uL的混合物加到ASV 0uM组和ASV 1uM组对应的孔中,6h后换液。
The specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 50uL respectively.
Figure PCTCN2017118970-appb-000003
Medium, then add 3.0uL Lipofectamine to one of the tubes
Figure PCTCN2017118970-appb-000004
3000 reagent (Thermo Fisher), 2 ug DNA plasmid (recombinant vector Pltr-sfGFP-N3N4) was added to the other tube, and mixed separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 100 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 50 uL of the mixture to the corresponding wells of the ASV 0uM group and the ASV 1uM group, respectively, and change the solution after 6 hours.
转染后24h,观察细胞表达荧光的情况。结果如图5所示。从图中可以看出:第一排:不加ASV(ASV 0uM组),表达的融合蛋白MTS-DsRed-4A4B-sfGFP-NS3/4A。蛋白酶具有活性,能在4A4B处将融合蛋白切断,形成MTS-DsRed和sfGFP-NS3/4A两个蛋白。由于有线粒体定位信号,MTS-DsRed表达定位在线粒体外膜上,而sfGFP-NS3/4A融合蛋白则广泛分布于细胞质中。第二排:加入ASV(ASV 1uM组),3T3细胞表达融合蛋白MTS-DsRed-4A4B-sfGFP-NS3/4A。由于ASV抑制了NS3/4A蛋白酶对4A4B的识别和切割,使整个融合蛋白能稳定存在。绿色荧光和红色荧光都受到线粒体定位信号的影响而共定位于线粒体表面。说明ASV可调控目标蛋白在细胞内的定位。At 24 hours after transfection, the cells were observed to express fluorescence. The result is shown in Figure 5. It can be seen from the figure that the first row: the fusion protein MTS-DsRed-4A4B-sfGFP-NS3/4A was expressed without ASV (ASV 0uM group). The protease is active and can cleave the fusion protein at 4A4B to form two proteins, MTS-DsRed and sfGFP-NS3/4A. Due to the mitochondrial localization signal, MTS-DsRed expression was localized on the mitochondrial outer membrane, while the sfGFP-NS3/4A fusion protein was widely distributed in the cytoplasm. The second row: ASV (ASV 1uM group) was added, and the 3T3 cells expressed the fusion protein MTS-DsRed-4A4B-sfGFP-NS3/4A. Since ASV inhibits the recognition and cleavage of 4A4B by NS3/4A protease, the entire fusion protein can be stably present. Both green and red fluorescence are co-localized to the mitochondrial surface due to mitochondrial localization signals. This indicates that ASV can regulate the localization of the target protein in cells.
③小分子药物调控:3 small molecule drug regulation:
转染24h后,将ASV 0uM组细胞培养基吸去,加入新鲜的含1uM的ASV的培养基;After transfection for 24 h, the cell culture medium of ASV 0uM group was aspirated, and fresh medium containing 1 uM of ASV was added;
转染24h后,将ASV 1uM组细胞培养基吸去,用PBS清洗两遍,然后加入新鲜的不含ASV的培养基。After 24 h of transfection, the ASV 1uM cell culture medium was aspirated, washed twice with PBS, and then fresh ASV-free medium was added.
加入ASV 24h后观察细胞表达荧光的情况。结果如图6所示。从图中可以看出:第一排:在没有ASV的条件下,绿色荧光蛋白和红色荧光蛋白分开表达。第二排:在加入ASV 24h后,绿色荧光蛋白在细胞中的定位逐步转移到线粒体表面。The cells expressed fluorescence after 24 hours of ASV addition. The result is shown in Figure 6. As can be seen from the figure: The first row: green fluorescent protein and red fluorescent protein are expressed separately without ASV. Second row: After 24 hours of ASV addition, the localization of green fluorescent protein in the cells was gradually transferred to the mitochondrial surface.
去除ASV 24h后观察细胞表达荧光的情况。结果如图7所示。从图中可以看出:第一排:在加入ASV的条件下,绿色荧光蛋白和红色荧光蛋白共定位表达于线粒体表面。第二排:去除ASV 24h后,红色荧光蛋白仍然定位于线粒体表面, 但绿色荧光蛋白逐步弥散分布于整个细胞质。说明ASV可快速切换目标蛋白在细胞内的定位。The cells were observed for fluorescence after removal of ASV for 24 h. The result is shown in Figure 7. It can be seen from the figure: First row: Under the condition of adding ASV, green fluorescent protein and red fluorescent protein are co-localized on the surface of mitochondria. The second row: After removing ASV for 24h, the red fluorescent protein is still located on the mitochondrial surface, but the green fluorescent protein is gradually dispersed throughout the cytoplasm. This indicates that ASV can quickly switch the localization of the target protein in the cell.
实施例3:嵌合抗原受体活性调控Example 3: Regulation of chimeric antigen receptor activity
一、嵌合抗原受体活性调控方案一I. Chimeric antigen receptor activity regulation program one
将嵌合抗原受体(CAR)与自降解的HCV蛋白酶融合表达,由HCV蛋白酶抑制剂ASV通过抑制蛋白酶活性,来调控嵌合抗原受体活性。The chimeric antigen receptor (CAR) is expressed by fusion with a self-degrading HCV protease, and the chimeric antigen receptor activity is regulated by the HCV protease inhibitor ASV by inhibiting protease activity.
方案一中的载体构建:Carrier construction in scenario 1:
将SEQ ID No.3所示的融合蛋白基因插入慢病毒载体Pltr载体,得到重组载体Pltr-CAR19-N34d,SEQ ID No.3第1-27位为Flag标签序列,第28-1491位为CAR基因,第1498-1530位为HCV病毒蛋白酶识别序列4A4B(DEMEECSQHLP)的编码基因,第1588-2388位为N34d基因(带有NS3/4A序列和一段降解子序列)。重组载体Pltr-CAR19-N34d的结构如图8所示。The fusion protein gene represented by SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N34d, the 1st to 27th of SEQ ID No. 3 was the Flag tag sequence, and the 28th to 1491th were CAR Genes, positions 1498-1530 are the genes encoding the HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), and positions 1588-2388 are the N34d gene (with the NS3/4A sequence and a stretcher sequence). The structure of the recombinant vector Pltr-CAR19-N34d is shown in Fig. 8.
本方案中,嵌合抗原受体(CAR)识别的抗原为人源CD19,将CAR与自降解的HCV蛋白酶融合表达,蛋白酶识别位点4A4B位于两个蛋白之间。HCV蛋白酶抑制剂ASV通过抑制蛋白酶活性,来调控CAR的表达。如图9所示:左图:在无ASV的条件下,NS3/4A切割4A4B序列,CAR与蛋白酶分开,蛋白酶被degron带入泛素-蛋白酶体系统降解,而CAR可完整表达并行使识别抗原和激活T细胞内部信号的功能。此时,开关处于on状态。右图:在有ASV的条件下,NS3/4A被抑制而不能切割4A4B序列,CAR、蛋白酶和degron融合表达,整个融合蛋白被degron带入泛素-蛋白酶体系统降解,CAR被降解,T细胞无法通过CAR识别抗原,不能被激活。此时,开关处于off状态。In the present scheme, the antigen recognized by the chimeric antigen receptor (CAR) is human CD19, and the CAR is expressed by fusion with the self-degrading HCV protease, and the protease recognition site 4A4B is located between the two proteins. The HCV protease inhibitor ASV regulates CAR expression by inhibiting protease activity. As shown in Figure 9: Left: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, CAR is separated from protease, protease is degraded by degron into the ubiquitin-proteasome system, and CAR can express and recognize antigen And the function of activating T cells internal signals. At this point, the switch is in the on state. Right: In the presence of ASV, NS3/4A is inhibited and the 4A4B sequence cannot be cleaved. CAR, protease and degron are fused, and the entire fusion protein is degraded by degron into the ubiquitin-proteasome system, and CAR is degraded. Unable to recognize antigen by CAR and cannot be activated. At this point, the switch is in the off state.
二、嵌合抗原受体活性调控方案二Second, chimeric antigen receptor activity regulation program two
将嵌合抗原受体(CAR)与HCV蛋白酶融合表达,与方案一不同之处在于,4A4B序列不在两个融合蛋白之间,而在CAR蛋白内部,位于4-1BB和CD3zata结构域之间,由HCV蛋白酶通过ASV来控制CAR的结构和功能。Fusion of chimeric antigen receptor (CAR) with HCV protease, which differs from protocol 1 in that the 4A4B sequence is not between the two fusion proteins, but within the CAR protein, between the 4-1BB and CD3zata domains, The structure and function of CAR is controlled by HCV protease by ASV.
方案二中的载体构建:The carrier construction in scenario 2:
将SEQ ID No.4所示的融合蛋白基因插入慢病毒载体Pltr载体,得到重组载体Pltr-CAR19-N3V4V2,SEQ ID No.4第1-27位为Flag标签序列,第28-1530位为CAR基因,HCV病毒蛋白酶识别序列4A4B(DEMEECSQHLP)的编码基因位于4-1BB和CD3zata结构域之间,第1579-2265位为NS3pro/4A蛋白酶基因。重组载体Pltr-CAR19-N34d的结构如图10所示。The fusion protein gene represented by SEQ ID No. 4 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N3V4V2, the 1st to 27th of SEQ ID No. 4 was the Flag tag sequence, and the positions 28 to 1530 were CAR. The gene, HCV viral protease recognition sequence 4A4B (DEMEECSQHLP), encodes the gene between the 4-1BB and CD3zata domains, and the 1579-2265 is the NS3pro/4A protease gene. The structure of the recombinant vector Pltr-CAR19-N34d is shown in FIG.
本方案中,嵌合抗原受体(CAR)识别的抗原为人源CD19,将CAR与HCV蛋白酶融合表达,蛋白酶识别位点4A4B位于CAR的4-1BB和CD3zata结构域之间。由HCV蛋白酶通过ASV来控制CAR的结构和功能。如图11所示:左图:在无ASV的条件下,NS3/4A切割4A4B序列,CAR蛋白中的4-1BB与CD3zeta结构域分开,无法传递细胞增殖信号。即使CAR-T细胞能识别肿瘤抗原,但由于无法增殖,不能有效杀灭相应肿瘤细胞。此时,开关处于off状态。右图:在有ASV的条件下, NS3/4A被抑制而不能切割4A4B序列,CAR、蛋白酶融合表达,CAR具有完整的结构,并行使识别抗原和激活T细胞内部信号的功能。此时,开关处于on状态。In the present scheme, the chimeric antigen receptor (CAR) recognizes an antigen that is human CD19, and fuses CAR and HCV protease, and the protease recognition site 4A4B is located between the 4-1BB and CD3zata domains of the CAR. The structure and function of CAR is controlled by HCV protease by ASV. As shown in Figure 11: Left panel: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, and the 4-1BB in the CAR protein is separated from the CD3zeta domain and cannot transmit cell proliferation signals. Even if CAR-T cells recognize tumor antigens, they cannot effectively kill the corresponding tumor cells because they cannot proliferate. At this point, the switch is in the off state. Right: Under the condition of ASV, NS3/4A is inhibited and can not cleave 4A4B sequence, CAR, protease fusion expression, CAR has a complete structure, and functions to recognize antigen and activate T cell internal signals. At this point, the switch is in the on state.
三、嵌合抗原受体活性调控方案三Third, chimeric antigen receptor activity regulation scheme three
将嵌合抗原受体(CAR)与HCV蛋白酶融合表达,与方案二不同之处在于,蛋白酶识别区在CAR蛋白内部增加为3个,分别位于CAR的anti-CD19scFv与CD8跨膜区之间、4-1BB与CD3zata结构域之间、CD8跨膜区与4-1BB结构域之间。由HCV蛋白酶通过ASV来控制CAR的结构和功能。The fusion of the chimeric antigen receptor (CAR) with the HCV protease is different from that of the second embodiment in that the protease recognition region is increased within the CAR protein by three, which are located between the anti-CD19 scFv of the CAR and the transmembrane region of the CD8, Between the 4-1BB and CD3zata domains, between the CD8 transmembrane region and the 4-1BB domain. The structure and function of CAR is controlled by HCV protease by ASV.
方案三中的载体构建:Carrier construction in scenario three:
将SEQ ID No.5所示的融合蛋白基因插入慢病毒载体Pltr载体,得到重组载体Pltr-CAR19-N3V4V3,SEQ ID No.5第1-28位为Flag标签序列,第28-1596位为CAR基因,HCV病毒蛋白酶识别序列4A4B的编码基因分别位于scFv与CD8跨膜区、CD8跨膜区与4-1BB结构域、HCV病毒蛋白酶识别序列5A5B位于4-1BB与CD3zata结构域之间,第1648-2334位为NS3pro/4A蛋白酶基因。重组载体Pltr-CAR19-N34d的结构如图12所示。The fusion protein gene represented by SEQ ID No. 5 was inserted into the lentiviral vector Pltr vector to obtain the recombinant vector Pltr-CAR19-N3V4V3, the 1st to 28th of SEQ ID No. 5 was the Flag tag sequence, and the 28th to the 1296th was CAR The gene encoding the HCV viral protease recognition sequence 4A4B is located between the scFv and CD8 transmembrane regions, the CD8 transmembrane region and the 4-1BB domain, and the HCV viral protease recognition sequence 5A5B is located between the 4-1BB and CD3zata domains, 1648. The -2334 position is the NS3pro/4A protease gene. The structure of the recombinant vector Pltr-CAR19-N34d is shown in FIG.
本方案中,嵌合抗原受体(CAR)识别的抗原为人源CD19,将CAR与HCV蛋白酶融合表达,蛋白酶识别位点分别位于CAR的anti-CD19scFv与CD8跨膜区之间、4-1BB与CD3zata结构域之间、CD8跨膜区与4-1BB结构域之间。如图13所示:左图:在无ASV的条件下,NS3/4A切割4A4B序列,CAR蛋白被切断为四个单独的结构域,引起相应的T细胞无法识别抗原,也不能传递细胞激活和增殖信号,不能杀灭相应肿瘤细胞。此时,开关处于off状态。右图:在有ASV的条件下,NS3/4A被抑制而不能切割4A4B序列,CAR、蛋白酶融合表达,CAR具有完整的结构,并行使识别抗原和激活T细胞内部信号的功能。此时,开关处于on状态。In this protocol, the chimeric antigen receptor (CAR) recognizes the antigen as human CD19, and fuses the expression of CAR and HCV protease. The protease recognition sites are located between the anti-CD19scFv of CAR and the transmembrane region of CD8, respectively. Between the CD3zata domains, between the CD8 transmembrane region and the 4-1BB domain. As shown in Figure 13: Left: In the absence of ASV, NS3/4A cleaves the 4A4B sequence, and the CAR protein is cleaved into four separate domains, causing the corresponding T cells to fail to recognize the antigen and also fail to deliver cell activation and Proliferation signals cannot kill the corresponding tumor cells. At this point, the switch is in the off state. Right: Under the condition of ASV, NS3/4A is inhibited and can not cleave 4A4B sequence, CAR and protease fusion expression, CAR has a complete structure, and functions to recognize antigen and activate T cell internal signal. At this point, the switch is in the on state.
四、在293细胞上调控嵌合抗原受体活性4. Regulation of chimeric antigen receptor activity on 293 cells
①载体构建:1 carrier construction:
分别将方案一、二和三中构建的载体命名为fCAR-V1、fCAR-V2和fCAR-V3。其结构图如图14所示(上图)。The vectors constructed in Schemes 1, 2, and 3 were named fCAR-V1, fCAR-V2, and fCAR-V3, respectively. Its structure is shown in Figure 14 (above).
②细胞转染:2 cell transfection:
复苏并培养293T细胞(Clontech:632180),分别将fCAR-V1、fCAR-V2和fCAR-V3转染293T细胞。转染前一天,在12孔板中每孔加入5×10 5个细胞。转染前换液,按照是否加入小分子抑制剂Asunaprevir(ASV)分为如下两组: 293T cells (Clontech: 632180) were resuscitated and cultured, and fCAR-V1, fCAR-V2 and fCAR-V3 were transfected into 293T cells, respectively. One day before transfection, 5 x 10 5 cells were added to each well in a 12-well plate. The liquid exchange before transfection was divided into the following two groups according to whether or not the small molecule inhibitor Asunaprevir (ASV) was added:
ASV 0uM组:一个孔加入不含ASV的培养基;ASV 0uM group: one well was added to the medium without ASV;
ASV 1uM组:一个孔加入含1uM ASV的培养基。ASV 1uM group: One well was added to the medium containing 1 uM ASV.
具体转染步骤如下:1)取两支1.5ml EP管,分别加入100uL 
Figure PCTCN2017118970-appb-000005
培养基,然后在其中一支管中加入6.0uL Lipofectamine
Figure PCTCN2017118970-appb-000006
3000试剂,另一支管中加入4ug DNA质粒(fCAR-V1、fCAR-V2或fCAR-V3),分别混匀。2)将两个EP管中的液体混合为一管,共得到200uL的混合物,轻轻吹打混匀15次,室温静置5分钟。3)分别取100uL混合物加到两个孔中,6h后换液。
The specific transfection steps are as follows: 1) Take two 1.5ml EP tubes and add 100uL respectively.
Figure PCTCN2017118970-appb-000005
Medium, then add 6.0uL Lipofectamine to one of the tubes
Figure PCTCN2017118970-appb-000006
For the 3000 reagent, add 4ug DNA plasmid (fCAR-V1, fCAR-V2 or fCAR-V3) to the other tube and mix separately. 2) The liquids in the two EP tubes were mixed into one tube, and a total of 200 uL of the mixture was obtained, and the mixture was gently blown and mixed for 15 times, and allowed to stand at room temperature for 5 minutes. 3) Add 100 uL of the mixture to each of the two wells, and change the solution after 6 hours.
转染24h后,弃去培养液,加入细胞裂解液(碧云天,P0013C)裂解细胞,收集蛋白。并利用western blot技术用Flag抗体(碧云天,AF519)检测含flag标签蛋白的大小。After transfection for 24 hours, the culture solution was discarded, and the cells were lysed by adding cell lysate (Biyuntian, P0013C) to collect proteins. The size of the flag-containing protein was detected by Western blot using the Flag antibody (Biyuntian, AF519).
western blot检测flag标签融合蛋白大小的结果如图14所示(下图)。从图中可以看出:fCAR-V1组:第一列:不加ASV时,表达的融合蛋白Flag-CAR-4A4B-NS3/4A-Degron。蛋白酶具有切割功能,在4A4B处将融合蛋白切断。然后,Degron介导NS3/4A蛋白酶快速降解,形成一段63kd大小的Flag-CAR片段。第二列:加入ASV后,293细胞同样表达融合蛋白Flag-CAR-4A4B-NS3/4A-Degron。但加入ASV抑制了NS3/4A蛋白酶对4A4B的识别和切割,使得Degron将整个融合蛋白带入泛素-蛋白酶体途径,进行快速降解。fCAR-V2组:第一列:不加ASV时,表达的融合蛋白Flag-CAR1-4A4B-CAR2-NS3/4A。蛋白酶具有活性,能在4A4B处将融合蛋白切断,形成大小45kd的Flag-CAR1和52kd的CAR2-NS3/4A两个蛋白。45kd的Flag-CAR1通过anti-flag抗体检测到。第二列:加入ASV后,293细胞表达融合蛋白Flag-CAR-4A4B-CAR2-NS3/4A。由于ASV抑制了NS3/4A蛋白酶对4A4B的识别和切割,使整个融合蛋白能稳定存在,anti-flag抗体检测到蛋白的大小为97kd。fCAR-V3组:第一列:不加ASV时,表达的融合蛋白Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A。蛋白酶具有活性,能在4A4B和5A5B处将融合蛋白切断,形成大小33kd的Flag-CAR1、7kd的CAR2、5kd的CAR3和52kd的CAR4-NS3/4A四个蛋白。33kd的Flag-CAR1通过anti-flag抗体检测到。第二列:加入ASV后,293细胞表达融合蛋白Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A。由于ASV抑制了NS3/4A蛋白酶对4A4B的识别和切割,使整个融合蛋白能稳定存在,anti-flag抗体检测到蛋白的大小为97kd。由于CAR的hinge部位自身可能自然断裂,所以所有组都能检测到一条大小33kd的蛋白。The results of western blot detection of the size of the flag tag fusion protein are shown in Figure 14 (below). It can be seen from the figure: fCAR-V1 group: the first column: the fusion protein Flag-CAR-4A4B-NS3/4A-Degron expressed without ASV. The protease has a cleavage function and the fusion protein is cleaved at 4A4B. Then, Degron mediates the rapid degradation of NS3/4A protease to form a 63kd size Flag-CAR fragment. Second column: After adding ASV, 293 cells also expressed the fusion protein Flag-CAR-4A4B-NS3/4A-Degron. However, the addition of ASV inhibited the recognition and cleavage of 4A4B by the NS3/4A protease, allowing Degron to bring the entire fusion protein into the ubiquitin-proteasome pathway for rapid degradation. fCAR-V2 group: The first column: the fusion protein Flag-CAR1-4A4B-CAR2-NS3/4A was expressed without ASV. The protease is active and can cleave the fusion protein at 4A4B to form a 45 kd Flag-CAR1 and a 52 kd CAR2-NS3/4A protein. 45 kd Flag-CAR1 was detected by anti-flag antibody. Second column: After addition of ASV, 293 cells expressed the fusion protein Flag-CAR-4A4B-CAR2-NS3/4A. Since ASV inhibits the recognition and cleavage of 4A4B by NS3/4A protease, the entire fusion protein can be stably present, and the anti-flag antibody detects a protein size of 97 kd. fCAR-V3 group: First column: The fusion protein Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A was expressed without ASV. The protease is active and can cleave the fusion protein at 4A4B and 5A5B to form four proteins of Flag-CAR1, 33kd, CAR2, 5kd CAR3 and 52kd CAR4-NS3/4A. The 33 kd Flag-CAR1 was detected by an anti-flag antibody. Second column: After addition of ASV, 293 cells expressed the fusion protein Flag-CAR1-4A4B-CAR2-5A5B-CAR3-4A4B-CAR4-NS3/4A. Since ASV inhibits the recognition and cleavage of 4A4B by NS3/4A protease, the entire fusion protein can be stably present, and the anti-flag antibody detects a protein size of 97 kd. Since the hinge site of CAR itself may naturally break, a group of 33kd protein can be detected in all groups.
实施例4:嵌合抗原受体杀伤功能测试Example 4: Chimeric antigen receptor killing function test
1、慢病毒载体的构建1. Construction of lentiviral vector
将SEQ ID No.3所示的融合蛋白基因插入慢病毒载体Pltr载体,得到慢病毒载体Pltr-CAR19-N34d,该慢病毒载体Pltr-CAR19-N34d表达嵌合抗原受体(CAR)和自降解的HCV蛋白酶的融合蛋白。The fusion protein gene represented by SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector to obtain a lentiviral vector Pltr-CAR19-N34d expressing the chimeric antigen receptor (CAR) and self-degradation of the Pltr-CAR19-N34d vector. Fusion protein of HCV protease.
将SEQ ID No.3第28-1491位所示的CAR基因插入慢病毒载体Pltr载体,得到的慢病毒载体PLTR-CAR19。该慢病毒载体PLTR-CAR19表达嵌合抗原受体(CAR)。The CAR gene shown in positions 28-1491 of SEQ ID No. 3 was inserted into the lentiviral vector Pltr vector, and the resulting lentiviral vector PLTR-CAR19 was obtained. The lentiviral vector PLTR-CAR19 expresses a chimeric antigen receptor (CAR).
2、慢病毒载体的包装2. Packaging of lentiviral vectors
分别将慢病毒PLTR-CAR19-N34d和慢病毒载体PLTR-CAR19与病毒包装质粒共转染到293T细胞中进行包装,得到病毒颗粒;两天后,收集细胞培养上清,利用PEG沉淀方法进行病毒浓缩,分别得到病毒液CAR19-N34d和病毒液CAR19,并冻存于-80℃备用。Lentiviral PLTR-CAR19-N34d and lentiviral vector PLTR-CAR19 were co-transfected into 293T cells with viral packaging plasmids to obtain viral particles. Two days later, the cell culture supernatant was collected and PEG-precipitated for virus concentration. The virus solution CAR19-N34d and the virus solution CAR19 were respectively obtained and stored frozen at -80 ° C for use.
3、感染T细胞3. Infected T cells
计算步骤2获得的病毒液CAR19-N34d和病毒液CAR19的病毒滴度,按照感染系数为10:1的比例感染T细胞(T细胞是用含0.5%的HSA和300U的IL2的X-vivo15培养基(Lonza)培养PBMC细胞(天津血液中心)三天后得到的细胞),分别得到可调控的CAR-T细胞和普通的CAR-T细胞。Calculate the virus titer of virus solution CAR19-N34d and virus solution CAR19 obtained in step 2, and infect T cells according to the infection coefficient of 10:1 (T cells are cultured with X-vivo15 containing 0.5% HSA and 300U IL2). Lonza cultured cells obtained three days after PBMC cells (Tianjin blood center), and regulatable CAR-T cells and common CAR-T cells were obtained, respectively.
T细胞感染一周后,分别测试可调控的CAR-T细胞和普通的CAR-T细胞的CAR19表达情况。结果表明:可调控的CAR-T细胞和普通的CAR-T细胞均有20%左右的细胞表达CAR19蛋白。One week after T cell infection, the expression of CAR19 in regulatable CAR-T cells and common CAR-T cells was tested. The results showed that about 20% of cells in regulated CAR-T cells and common CAR-T cells expressed CAR19 protein.
4、嵌合抗原受体杀伤功能测试4, chimeric antigen receptor killing function test
将表达嵌合抗原受体的CAR-T细胞与raji靶细胞在培养体系中共培养,培养体系中含有病毒蛋白酶抑制剂ASV或DPV(阿拉丁),并以不含有病毒蛋白酶抑制剂作为对照(Control)。采用Elisport检测CAR-T细胞的IFN-gamma的表达情况。具体步骤如下:The CAR-T cells expressing the chimeric antigen receptor are co-cultured with the raji target cells in a culture system containing the viral protease inhibitor ASV or DPV (Aladdin), and the virus protease inhibitor is not included as a control (Control) ). Elisport was used to detect the expression of IFN-gamma in CAR-T cells. Specific steps are as follows:
1)取3万个感染7天后的CAR-T细胞和15万个raji靶细胞(购自ADCC),置于100μl的含10%FBS(Biowest)的1640培养基(GBICO)中,然后加入到包被干扰素抗体(南通表源生物技术有限公司)的板子中,置于培养箱16小时。1) Take 30,000 CAR-T cells and 150,000 raji target cells (purchased from ADCC) after 7 days of infection, and place them in 100 μl of 1640 medium (GBICO) containing 10% FBS (Biowest), and then add to The plate coated with interferon antibody (Nantong Table Source Biotechnology Co., Ltd.) was placed in an incubator for 16 hours.
2)弃去培养基,用PBS洗三次,加入检测抗体(南通表源生物技术有限公司)室温避光孵育1h。2) Discard the medium, wash it three times with PBS, add the detection antibody (Nantong Table Source Biotechnology Co., Ltd.) and incubate at room temperature for 1 h in the dark.
3)再用PBS洗三次,加入AP酶标记的链霉亲和素抗体(南通表源生物技术有限公司),室温避光孵育45min。3) Wash three times with PBS, add AP-labeled streptavidin antibody (Nantong Table Source Biotechnology Co., Ltd.), incubate at room temperature for 45 min in the dark.
4)PBS洗四次,加入100μl显色液显色20min后,观察拍照。4) Wash the PBS four times, add 100 μl of the color developing solution for 20 minutes, and observe the photograph.
结果如图15所示,从图中可以看出:上排:普通CAR-T细胞结合raji细胞后释放大量IFN-gamma(图中的黑点),加入病毒蛋白酶抑制剂ASV或DPV,并不能抑制CAR-T细胞释放IFN-gamma。下排:可调控的CAR-T细胞在结合raji细胞后同样释放大量IFN-gamma,但在培养体系中加入病毒蛋白酶抑制剂ASV或DPV,IFN-gamma的表达大大减少。说明加入ASV或DPV后,病毒蛋白酶被抑制而不能切割CAR19-N34d,导致这个蛋白结构不稳定而降解。CAR19被降解后,无法行使识别抗原和激活T细胞内部信号的功能,T细胞无法和raji细胞结合,从而不能刺激自身IFN-gamma的表达。The results are shown in Fig. 15. As can be seen from the figure: the upper row: normal CAR-T cells release a large amount of IFN-gamma (black spots in the figure) after binding to raji cells, and the addition of the viral protease inhibitors ASV or DPV does not Inhibition of release of IFN-gamma by CAR-T cells. Lower row: Regulatory CAR-T cells also release large amounts of IFN-gamma after binding to raji cells, but the expression of IFN-gamma is greatly reduced by the addition of the viral protease inhibitors ASV or DPV to the culture system. It indicated that after addition of ASV or DPV, the viral protease was inhibited and could not cleave CAR19-N34d, resulting in unstable and degraded structure of this protein. After CAR19 is degraded, it cannot function to recognize antigens and activate internal signals of T cells. T cells cannot bind to raji cells, and thus cannot stimulate the expression of IFN-gamma.
以上述依据本发明的理想实施例为启示,通过上述的说明内容,相关工作人员完全可以在不偏离本项发明技术思想的范围内,进行多样的变更以及修改。本项发明的技术性范围并不局限于说明书上的内容,必须要根据权利要求范围来确定其技术性范围。In view of the above-described embodiments of the present invention, various changes and modifications may be made by those skilled in the art without departing from the scope of the invention. The technical scope of the present invention is not limited to the contents of the specification, and the technical scope thereof must be determined according to the scope of the claims.
工业应用Industrial application
本发明公开了一种小分子药物控制的蛋白功能开关系统。该系统包括控制子原件和操纵子原件,控制子原件为蛋白酶或由蛋白酶与降解子融合而成的蛋白酶-降解子;所述操纵子原件为可被所述蛋白酶识别并切割的连接肽;The invention discloses a small molecule drug controlled protein function switching system. The system comprises a control element original and a operon original, the control element is a protease or a protease-degradant fused by a protease and a proton; the operon element is a linker peptide recognizable and cleaved by the protease;
该系统还包括所述蛋白酶的小分子抑制剂;整个系统通过蛋白酶的小分子抑制剂 调控蛋白酶或蛋白酶-降解子的活性,进而调控目标蛋白的活性。利用此小分子药物可精确调节目标蛋白的生物学活性。本发明的病毒蛋白酶可以是天然的蛋白酶,也可以经过遗传改造,减小蛋白酶分子量的蛋白酶,或是与降解子结构域(degron)融合而成的蛋白酶-降解子。病毒蛋白酶抑制剂主要为市面上用于治疗或进入临床实验的药物。本发明制备了病毒蛋白酶依赖的小分子药物用来调控目标蛋白功能的开关系统,提供了一种快速、高效、可逆、可控、简单、经济和具有良好应用前景的蛋白功能调控方法。The system also includes a small molecule inhibitor of the protease; the entire system modulates the activity of the protease or protease-degradant by a small molecule inhibitor of the protease, thereby regulating the activity of the protein of interest. The small molecule drug can be used to precisely regulate the biological activity of the target protein. The viral protease of the present invention may be a natural protease, a protease that is genetically engineered to reduce the molecular weight of the protease, or a protease-degradant fused to a degrading domain (degron). Viral protease inhibitors are primarily drugs that are commercially available for treatment or for clinical trials. The invention prepares a switching system of a virus protease-dependent small molecule drug for regulating the function of a target protein, and provides a protein function regulation method which is fast, efficient, reversible, controllable, simple, economical and has good application prospect.

Claims (34)

  1. 一种小分子药物控制的蛋白功能开关系统,所述开关系统由操纵子原件和控制子原件构成,所述控制子为单个病毒蛋白酶,或由病毒蛋白酶与蛋白降解子融合而成的蛋白酶-降解子;所述操纵子位于目标蛋白内部,或位于目标蛋白与蛋白酶-降解子结构之间,由蛋白酶的一段连接肽构成;整个系统通过蛋白酶小分子的抑制剂化合物作为开关,实现对目标蛋白的调控。A small molecule drug controlled protein function switching system consisting of an operon element and a control subunit, the controller being a single viral protease or a protease-degraded fusion of a viral protease and a protein degradant The operon is located inside the target protein or between the target protein and the protease-degrading structure, and is composed of a ligation peptide of the protease; the whole system realizes the target protein by using the inhibitor compound of the small molecule of the protease as a switch. Regulation.
  2. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述开关系统还包括蛋白酶小分子的抑制剂化合物。The protein function switching system according to claim 1, wherein said switching system further comprises an inhibitor compound of a protease small molecule.
  3. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述操纵子原件的个数为一个或两个或多个。The protein function switch system according to claim 1, wherein the number of the operon originals is one or two or more.
  4. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述病毒蛋白酶为丙肝病毒蛋白酶。The protein function switch system according to claim 1, wherein the viral protease is a hepatitis C virus protease.
  5. 根据权利要求4所述的蛋白功能开关系统,其特征在于,所述丙肝病毒蛋白酶为丙肝病毒蛋白酶NS3pro/4A。The protein function switch system according to claim 4, wherein the hepatitis C virus protease is hepatitis C virus protease NS3pro/4A.
  6. 根据权利要求4所述的蛋白功能开关系统,其特征在于,所述丙肝病毒蛋白酶为与丙肝病毒蛋白酶NS3pro/4A具有70%或70%以上同源性的蛋白酶突变体或异构酶。The protein function switch system according to claim 4, wherein the hepatitis C virus protease is a protease mutant or isomerase having 70% or more homology with the hepatitis C virus protease NS3pro/4A.
  7. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述连接肽为下述序列之一:The protein function switch system according to claim 1, wherein the linker peptide is one of the following sequences:
    TRIF:Pro Ser Ser Thr Pro Cys Ser Ala His Leu;TRIF: Pro Ser Ser Thr Pro Cys Ser Ala His Leu;
    MAVS:Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;MAVS: Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;
    3-4A:Asp Leu Glu Val Val Thr Ser Thr Trp Val;3-4A: Asp Leu Glu Val Val Thr Ser Thr Trp Val;
    4A4B:Asp Glu Met Glu Glu Cys Ser Gln His Leu;4A4B: Asp Glu Met Glu Glu Cys Ser Gln His Leu;
    4B5A:Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;4B5A: Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
    5A5B:Glu Asp Val Val Cys Cys Ser Met Ser Tyr。5A5B: Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
  8. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述蛋白酶小分子的抑制剂化合物为下述化合物之一:Telaprevir、Boceprevir、Simeprevir、Faldaprevir、Asunaprevir和Furaprevir。The protein function switch system according to claim 1, wherein the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir, Boceprevir, Simeprevir, Faldaprevir, Asunaprevir, and Furaprevir.
  9. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述降解子为NS3/4A不稳定区域、ddFKBP不稳定区域或ecDHFR不稳定区域。The protein function switch system according to claim 1, wherein the proton is an NS3/4A unstable region, a ddFKBP unstable region, or an ecDHFR unstable region.
  10. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述蛋白酶-降解子为N43d蛋白。The protein function switching system according to claim 1, wherein the protease-degrading agent is an N43d protein.
  11. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述目标蛋白为单个蛋白或融合蛋白。The protein function switch system according to claim 1, wherein the target protein is a single protein or a fusion protein.
  12. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述目标蛋白为荧光蛋白、耐药基因、抗体、细胞因子和抗原识别分子中的任意一种。The protein function switch system according to claim 1, wherein the target protein is any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
  13. 根据权利要求1所述的蛋白功能开关系统,其特征在于,所述蛋白功能开关系统用于真核哺乳动物细胞中。The protein function switching system according to claim 1, wherein said protein function switching system is used in eukaryotic mammalian cells.
  14. 权利要求1中的表达蛋白功能开关系统的核酸分子、质粒、细胞系以及包含上述元素装配而成的试剂盒;A nucleic acid molecule, a plasmid, a cell line, and a kit comprising the above-described elements, which express a protein function switching system according to claim 1;
    或,用于表达权利要求1-13所述的蛋白功能开关系统的核酸分子、质粒、细胞系以及包含上述元素装配而成的试剂盒。Or a nucleic acid molecule, a plasmid, a cell line, and a kit comprising the above-described elements for expressing the protein function switch system of claims 1-13.
  15. 权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在调控目标蛋白结构和/或功能中的应用;Use of the protein function switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for regulating the structure and/or function of a protein of interest;
    或,权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白结构和/或功能的产品中的应用。Or the use of the protein functional switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for the preparation of a product that modulates the structure and/or function of a protein of interest.
  16. 权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在调控目标蛋白活性中的应用;Use of the protein function switch system according to any one of claims 1 to 13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for regulating the activity of a protein of interest;
    或,权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白活性的产品中的应用。Or the use of the protein function switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for the preparation of a product that modulates the activity of a protein of interest.
  17. 权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在调控目标蛋白稳定性中的应用;Use of the protein function switch system according to any one of claims 1 to 13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for regulating the stability of a target protein;
    或,权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白稳定性的产品中的应用。Or the use of the protein functional switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for the preparation of a product that modulates the stability of a protein of interest.
  18. 权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在调控目标蛋白的时空位置中的应用;The use of the protein function switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for regulating the spatiotemporal position of a protein of interest;
    或,权利要求1-13任一所述的蛋白功能开关系统或权利要求14所述的核酸分子、质粒、细胞系或试剂盒在制备调控目标蛋白的时空位置的产品中的应用。Or the use of the protein function switch system of any of claims 1-13 or the nucleic acid molecule, plasmid, cell line or kit of claim 14 for the preparation of a product for regulating the spatiotemporal position of a protein of interest.
  19. 一种调控目标蛋白的方法,包括如下步骤:在受体细胞中表达出包含控制子原件、操纵子原件和目标蛋白的融合蛋白,再利用蛋白酶小分子的抑制剂化合物来调控所述控制子原件的活性,进而实现对目标蛋白的调控;A method for regulating a target protein, comprising the steps of: expressing a fusion protein comprising a promoter subgenus, an operon element, and a target protein in a recipient cell, and then using an inhibitor compound of a protease small molecule to regulate the control element original Activity, thereby achieving regulation of the target protein;
    所述控制子原件为单个病毒蛋白酶,或由病毒蛋白酶与蛋白降解子融合而成的蛋白酶-降解子;所述操纵子原件为所述蛋白酶可识别并切割的连接肽。The control element is a single viral protease, or a protease-degradant fused by a viral protease and a protein degrader; the operon element is a linker peptide that the protease recognizes and cleaves.
  20. 根据权利要求19所述的方法,其特征在于,所述操纵子原件的个数为一个或两个或多个。The method according to claim 19, wherein the number of the operon originals is one or two or more.
  21. 根据权利要求19所述的方法,其特征在于,所述病毒蛋白酶为丙肝病毒蛋白酶。The method according to claim 19, wherein the viral protease is a hepatitis C virus protease.
  22. 根据权利要求21所述的方法,其特征在于,所述丙肝病毒蛋白酶为丙肝病毒蛋白酶NS3pro/4A。The method according to claim 21, wherein the hepatitis C virus protease is hepatitis C virus protease NS3pro/4A.
  23. 根据权利要求21所述的方法,其特征在于,所述病毒蛋白酶为与丙肝病毒蛋白酶NS3pro/4A具有70%或70%以上同源性的蛋白酶突变体或异构酶。The method according to claim 21, wherein the viral protease is a protease mutant or isomerase having 70% or more homology with hepatitis C virus protease NS3pro/4A.
  24. 根据权利要求19所述的方法,其特征在于,所述连接肽为下述序列之一:The method according to claim 19, wherein said linker peptide is one of the following sequences:
    TRIF:Pro Ser Ser Thr Pro Cys Ser Ala His Leu;TRIF: Pro Ser Ser Thr Pro Cys Ser Ala His Leu;
    MAVS:Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;MAVS: Glu Arg Glu Val Pro Cys His Arg Pro Ser Pro;
    3-4A:Asp Leu Glu Val Val Thr Ser Thr Trp Val;3-4A: Asp Leu Glu Val Val Thr Ser Thr Trp Val;
    4A4B:Asp Glu Met Glu Glu Cys Ser Gln His Leu;4A4B: Asp Glu Met Glu Glu Cys Ser Gln His Leu;
    4B5A:Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;4B5A: Glu Cys Thr Thr Pro Cys Ser Gly Ser Trp;
    5A5B:Glu Asp Val Val Cys Cys Ser Met Ser Tyr。5A5B: Glu Asp Val Val Cys Cys Ser Met Ser Tyr.
  25. 根据权利要求19所述的方法,其特征在于,所述蛋白酶小分子的抑制剂化合物为下述化合物之一:Telaprevir、Boceprevir、Simeprevir、Faldaprevir、Asunaprevir和Furaprevir。The method according to claim 19, wherein the inhibitor compound of the protease small molecule is one of the following compounds: Telaprevir, Boceprevir, Simeprevir, Faldaprevir, Asunaprevir and Furaprevir.
  26. 根据权利要求19所述的方法,其特征在于,所述降解子为NS3/4A不稳定区域、ddFKBP不稳定区域或ecDHFR不稳定区域。The method according to claim 19, wherein the proton is an NS3/4A unstable region, a ddFKBP unstable region or an ecDHFR unstable region.
  27. 根据权利要求19所述的方法,其特征在于,所述蛋白酶-降解子为N43d蛋白。The method according to claim 19, wherein the protease-degrading agent is an N43d protein.
  28. 根据权利要求19所述的方法,其特征在于,所述目标蛋白可为单个蛋白或融合蛋白。The method according to claim 19, wherein the protein of interest is a single protein or a fusion protein.
  29. 根据权利要求19所述的方法,其特征在于,所述目标蛋白可为荧光蛋白、耐药基因、抗体、细胞因子和抗原识别分子中的任意一种。The method according to claim 19, wherein the target protein is any one of a fluorescent protein, a drug resistance gene, an antibody, a cytokine, and an antigen recognition molecule.
  30. 根据权利要求19所述的方法,其特征在于,所述在受体细胞中表达出包含控制子原件、操纵子原件和目标蛋白的融合蛋白,再利用蛋白酶小分子的抑制剂化合物来调控所述控制子原件的活性的方法为将表达所述控制子原件、所述操纵子原件和所述目标蛋白的载体导入所述受体细胞中,得到重组细胞;在培养体系中培养所述重组细胞,得到所述融合蛋白;通过在所述培养体系中的添加或去除所述抑制剂化合物可实现调控所述控制子原件的活性。The method according to claim 19, wherein said expression of a fusion protein comprising a promoter subgenus, an operon element and a protein of interest is expressed in a recipient cell, and then an inhibitor compound of a protease small molecule is used to regulate said A method of controlling the activity of a subunit is to introduce a vector expressing the control subunit, the operon element, and the target protein into the recipient cell to obtain a recombinant cell; and cultivating the recombinant cell in a culture system, The fusion protein is obtained; the activity of the control subunit is regulated by the addition or removal of the inhibitor compound in the culture system.
  31. 根据权利要求30所述的方法,其特征在于,所述表达所述控制子原件、所述操纵子原件和所述目标蛋白的载体为将含有所述控制子原件的编码基因、所述操纵子原件的编码基因和所述目标蛋白的编码基因的片段插入表达载体的多克隆位点得到的。The method according to claim 30, wherein said vector expressing said control subunit, said operon element and said target protein is a coding gene containing said control element, said operon The coding gene of the original and the fragment of the gene encoding the target protein are inserted into the multiple cloning site of the expression vector.
  32. 根据权利要求31所述的方法,其特征在于,所述含有所述控制子原件的编码基因、所述操纵子原件的编码基因和所述目标蛋白的编码基因的片段为SEQ ID No.1或SEQ ID No.2或SEQ ID No.3或SEQ ID No.4或SEQ ID No.5所示的DNA分子。The method according to claim 31, wherein the coding gene containing the control subunit, the coding gene of the operon original, and the fragment of the coding gene of the target protein are SEQ ID No. 1 or A DNA molecule represented by SEQ ID No. 2 or SEQ ID No. 3 or SEQ ID No. 4 or SEQ ID No. 5.
  33. 根据权利要求31所述的方法,其特征在于:The method of claim 31 wherein:
    所述表达载体为慢病毒载体Pltr载体。The expression vector is a lentiviral vector Pltr vector.
  34. 根据权利要求19或30所述的方法,其特征在于:A method according to claim 19 or 30, wherein:
    所述受体细胞为293T细胞或3T3细胞。The recipient cell is a 293T cell or a 3T3 cell.
PCT/CN2017/118970 2016-12-27 2017-12-27 Protein function switch system controlled by small molecule drug WO2018121602A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201611223345.9A CN108239656A (en) 2016-12-27 2016-12-27 A kind of protein function switching system of small-molecule drug control
CN201611223345.9 2016-12-27

Publications (1)

Publication Number Publication Date
WO2018121602A1 true WO2018121602A1 (en) 2018-07-05

Family

ID=62702472

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/118970 WO2018121602A1 (en) 2016-12-27 2017-12-27 Protein function switch system controlled by small molecule drug

Country Status (2)

Country Link
CN (1) CN108239656A (en)
WO (1) WO2018121602A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020010239A1 (en) * 2018-07-06 2020-01-09 The Board Of Trustees Of The Leland Stanford Junior University Chimeric antigen receptor polypeptides and methods of using same
US20220380801A1 (en) * 2019-07-15 2022-12-01 Medimmune Limited Tripartite systems for protein dimerization and methods of use

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220073565A1 (en) * 2019-01-07 2022-03-10 University Of Washington De novo design of protein switches for tunable control of protein degradation
CN110092838B (en) * 2019-05-09 2023-03-10 拜澳泰克(沈阳)生物医学集团有限公司 Discovery and application of polypeptide capable of enhancing protein degradation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998037180A2 (en) * 1997-02-22 1998-08-27 Abbott Laboratories Hcv fusion protease and polynucleotide encoding same
WO2008054595A2 (en) * 2006-09-28 2008-05-08 The Regents Of The University Of California Drug controlled molecular tags
WO2017004022A2 (en) * 2015-06-29 2017-01-05 The Board Of Trustees Of The Leland Stanford Junior University Degron fusion constructs and methods for controlling protein production

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2601706C (en) * 2005-03-22 2016-09-20 Dana-Farber Cancer Institute, Inc. Treatment of protein degradation disorders
CN104829686A (en) * 2015-05-04 2015-08-12 上海交通大学 Small molecule compound co-immunoprecipitation method for separating target protein

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998037180A2 (en) * 1997-02-22 1998-08-27 Abbott Laboratories Hcv fusion protease and polynucleotide encoding same
WO2008054595A2 (en) * 2006-09-28 2008-05-08 The Regents Of The University Of California Drug controlled molecular tags
WO2017004022A2 (en) * 2015-06-29 2017-01-05 The Board Of Trustees Of The Leland Stanford Junior University Degron fusion constructs and methods for controlling protein production

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BANASZYNSKI, L.A. ET AL.: "A Rapid, Reversible, and Tunable Method to Regulate Protein Function in Living Cells Using Synthetic Small Molecules", CELL, vol. 126, 8 September 2006 (2006-09-08), pages 995 - 1004, XP008119027 *
IWAMOTO, M. ET AL.: "A General Chemical Method to Regulate Protein: Stability in the Mammalian Central Nervous System", CHEMISTRY & BIOLOGY, vol. 17, no. 9, 24 September 2010 (2010-09-24), pages 981 - 988, XP027299452, Retrieved from the Internet <URL:doi:10.1016/j.chembiol.2010.07.009> *
WU, C.Y. ET AL.: "Remote Control of Therapeutic T Cells Through a Small Molecule-gated Chimeric Receptor", SCIENCE, vol. 350, no. 6258, 16 October 2015 (2015-10-16), pages 1 - 21, XP055260068 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020010239A1 (en) * 2018-07-06 2020-01-09 The Board Of Trustees Of The Leland Stanford Junior University Chimeric antigen receptor polypeptides and methods of using same
US20210171602A1 (en) * 2018-07-06 2021-06-10 The Board Of Trustees Of The Leland Stanford Junior University Chimeric Antigen Receptor Polypeptides and Methods of Using Same
US20220380801A1 (en) * 2019-07-15 2022-12-01 Medimmune Limited Tripartite systems for protein dimerization and methods of use

Also Published As

Publication number Publication date
CN108239656A (en) 2018-07-03

Similar Documents

Publication Publication Date Title
WO2018121602A1 (en) Protein function switch system controlled by small molecule drug
US9321847B2 (en) Activatable toxin complexes comprising a cleavable inhibitory peptide
WO2015109931A1 (en) Human tim-3 fusion protein capable of blocking tim-3 signaling pathway
CN104628848B (en) Monoclonal antibody MERS 27 and its encoding gene and application
JP2025032195A (en) Fusion constructs for controlling protein function
US20220380801A1 (en) Tripartite systems for protein dimerization and methods of use
US20240132857A1 (en) Fusion proteins comprising two ring domains
CN117511885B (en) Engineered TIL cells with improved tumor recognition and killing capabilities and their applications
CN114349840A (en) Fusion protein and preparation method and application thereof
CN101225110A (en) Human interleukin-22 mutant and its construction method and application
CN104628849B (en) Monoclonal antibody MERS 4 and its encoding gene and application
Gradauskaite et al. LRP6 is a functional receptor for attenuated canine distemper virus
WO2019129124A1 (en) T cell containing cd40 antibody and muc1 specific chimeric antigen receptor gene and use thereof
Sarratea et al. Zika virus NS4B protein targets TANK-binding kinase 1 and inhibits type I interferon production
Ai et al. Characterization of hepatitis C virus core protein multimerization and membrane envelopment: revelation of a cascade of core-membrane interactions
Houshmand et al. Interaction of hepatitis C virus NS5A with La protein revealed by T7 phage display
WO2019129173A1 (en) Co-expressed cd40 antibody, mesothelin-specific chimeric antigen receptor t cell, and use thereof
CN116120442A (en) A kind of broad-spectrum neutralizing antibody P2-1B1 neutralizing SARS-CoV-2 and its application
Li et al. Production of virus-like particles of FMDV by 3C protease cleaving precursor polyprotein P1 in vitro
EP1261645A1 (en) Anti-viral conjugate comprising a factor allowing the translocation of a protein across a cell membrane and comprising a single-chain antibody fragment directed against a viral protein
AU2759700A (en) Polymeric immunoglobulin receptor (pigr)-binding domains and methods of use therefor
CN114703229B (en) A surface display technology based on human cells and a polypeptide targeting HBV receptor and its application
Xiang et al. Purification and application of bacterially expressed chimeric protein E1E2 of hepatitis C virus
WO2013027217A1 (en) Zymoxins and methods of using the same
CN113501863B (en) Mucin13 antagonistic polypeptide, derivative and application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17886818

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17886818

Country of ref document: EP

Kind code of ref document: A1