WO2024032485A1 - Non-phosphorylated and non-ubiquitinated crept protein and use thereof - Google Patents

Non-phosphorylated and non-ubiquitinated crept protein and use thereof Download PDF

Info

Publication number
WO2024032485A1
WO2024032485A1 PCT/CN2023/111172 CN2023111172W WO2024032485A1 WO 2024032485 A1 WO2024032485 A1 WO 2024032485A1 CN 2023111172 W CN2023111172 W CN 2023111172W WO 2024032485 A1 WO2024032485 A1 WO 2024032485A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
crept
cells
phosphorylation
cell
Prior art date
Application number
PCT/CN2023/111172
Other languages
French (fr)
Chinese (zh)
Inventor
常智杰
任芳丽
王银银
宋云皓
Original Assignee
清华大学
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 清华大学 filed Critical 清华大学
Publication of WO2024032485A1 publication Critical patent/WO2024032485A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/65Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression using markers
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/536Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase
    • G01N33/537Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with separation of immune complex from unbound antigen or antibody
    • G01N33/539Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with separation of immune complex from unbound antigen or antibody involving precipitating reagent, e.g. ammonium sulfate
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2510/00Genetically modified 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/82Translation products from oncogenes

Definitions

  • the present invention relates to the field of molecular biology, and more specifically, to a CREPT protein that can maintain a non-phosphorylated and non-ubiquitinated state and its application.
  • DNA is replicated in the S phase of the cell cycle. Errors in DNA replication will accumulate in stem cells in organisms and are closely related to cell death and individual aging. Therefore, DNA replication that determines cell fate needs to be tightly controlled.
  • the DNA replication mechanism of eukaryotic cells is very complex. The starting point of DNA replication is located in the G1 phase of the cell cycle, and the pre-RC (pre-replication complex) protein is loaded on all potential starting points of the genome. First, the ORC complex (origin recognition complex, ORC1-6) with ATPase activity is recruited to the origin of replication.
  • ORC complex oil recognition complex
  • hexameric complex of CDC6, CDT1 (CDC10-dependent transcript 1) and MCM2-7 (mini-chromosome maintenance) is loaded onto the OCR complex to form the MCM helicase complex.
  • This step is called origin licensing.
  • Activation of replication initiation involves the formation of the pre-IC (pre-initiation complex) complex and the activation of the MCM helicase complex.
  • the assembly of Pre-IC is triggered by DDK (DBF4-dependent kinase) and CDK (Cyclin-dependent kinase) during the G1/S transition period.
  • DDK and CDK phosphorylate several proteins involved in DNA replication, such as MCM10, CDC45, RECQL4 (ATP-dependent DNA helicase Q4), treslin, GINS and TOPBP1 (DNAtopoisomerase 2-binding protein 1).
  • MCM10, CDC45, RECQL4 ATP-dependent DNA helicase Q4
  • treslin GINS
  • TOPBP1 DNAtopoisomerase 2-binding protein 1
  • DDK and CDK can also phosphorylate several bases in the MCM2-7 complex, causing helicase activation and unwinding of DNA.
  • the MCM2-7 double hexamer loaded on DNA separates into two separate hexamers that continue to unwind at both replication forks starting from the replication origin.
  • UPS ubiquitin-proteasome system
  • E1 ubiquitin-activating enzymes
  • E2 ubiquitin-conjugating enzymes
  • E3 ubiquitin ligases
  • the RING-finger subfamily E3 composed of SKP1, CUL1 and F-box protein (FBP) is connected
  • the SCF complex of the enzyme recognizes the substrate to be degraded.
  • FBP primarily determines the target specificity of the SCF complex.
  • the SCF complex prefers to bind to phosphodegrons, which are phosphorylated substrate motifs that create a surface for recognition by E3 ligases.
  • FBP family member SKP2 S-phase kinase-related protein 2 is one of the E3 ligases that regulates G1/S transition.
  • the SKP2-CKS1-p27 complex mediates proteasomal degradation of the CDK inhibitor p27 and releases Cyclin E/CDK2 to promote G1/S transition.
  • CREPT Cell-cycle Related and Expression-elevated
  • the inventors of the present invention have discovered through extensive research that the CREPT protein in human cells promotes cells to enter the S1 phase through degradation during the G1/S transition phase, thus exerting a cell cycle regulatory effect.
  • CREPT was expressed at a very low level in some cells, while maintaining high expression in other cells, and further found that this low expression phenomenon was due to It is caused by the degradation of CREPT during the G1/S transition period, and the degradation of CREPT is crucial for cells to enter the S phase from the G1 phase.
  • the degradation of CREPT is mediated by ubiquitination modification, which depends on the phosphorylation of two sites of CREPT, S134 and S166.
  • the inventor further discovered that if the 134 and 166 positions of the CREPT protein are kept in a non-phosphorylated state during the G1/S transition period and the protein is kept in a non-ubiquitinated state, the CREPT protein cannot be degraded. This kind of protein cannot be degraded. The CREPT protein cannot dissociate from the MCM complex, resulting in cell death.
  • the inventor also found that the homologous protein in non-human eukaryotic cells has the same or similar cell cycle regulatory effect and regulatory mechanism as human CREPT, and when the human CREPT protein is expressed in non-human eukaryotic cells, this Cell cycle regulation remains.
  • the present invention provides a method for phosphorylating SEQ ID No: 4 at positions 134 and 166.
  • the protein obtained by the inactivation modification.
  • the phosphorylation inactivation modification allows the 134 and 166 positions to remain unphosphorylated and remain unphosphorylated when the protein is located in eukaryotic cells at the end of G1 phase or the G1/S transition phase.
  • the protein remains in a non-ubiquitinated state, thereby preventing the protein from being degraded in the eukaryotic cell.
  • the eukaryotic cell may be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell cell.
  • the phosphorylation inactivating modifications on the 134 and 166 positions may be amino acid mutations and/or chemical modifications.
  • the phosphorylation-inactivating modification at the 134 and 166 positions may be a mutation of serine (S) to alanine (A).
  • the present invention also provides a protein that has (i) more than 90% sequence identity and (ii) the same phosphorylation inactivating modifications at positions 134 and 166 with each of the above-mentioned proteins.
  • the present invention provides a protein having a tag sequence or a guide sequence connected to the N-terminal and/or C-terminal of the protein of the first aspect.
  • the invention provides nucleic acids encoding the proteins of the first and second aspects.
  • the present invention provides a vector comprising the nucleic acid of the third aspect.
  • the invention provides cells comprising the vector of the fourth aspect.
  • the present invention provides the use of the proteins, nucleic acids or vectors described in the first to fifth aspects to inhibit eukaryotic cell proliferation, inhibit eukaryotic cell DNA replication, regulate the cell cycle of eukaryotic cells or kill eukaryotic cells.
  • Cellular reagent applications are described in the first to fifth aspects.
  • the present invention provides the use of the protein, nucleic acid or vector described in the first to fifth aspects in the preparation of anti-cancer drugs.
  • the present invention provides a method for treating cancer, the method comprising administering to a subject an effective amount of the protein, nucleic acid or vector described in the first to fifth aspects; or, the method comprising using a method based on The gene editing technology of CRISPR/Cas9 edits the CRPET gene in the genome of the subject's cancer cells so that the cancer cells express the protein of SEQ ID NO:2.
  • the subject can be a mammal, preferably a human.
  • the expression of wild-type CREPT in the cancer cells of the subject can be reduced or eliminated.
  • the present invention provides a method for screening non-phosphorylated non-ubiquitinated modifiers of CREPT protein, wherein the modifier maintains the S134 and S166 sites of CREPT protein in a continuous non-phosphorylated state, thereby allowing The CREPT protein remains in a non-ubiquitinated state in cells and will not be degraded; the CREPT protein
  • the amino acid sequence is SEQ ID No: 4, and the method includes:
  • the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The candidate modifiers are screened as non-phosphorylated, non-ubiquitinated modifiers of the CREPT protein.
  • the present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby causing the CREPT protein to function in cells. It maintains a non-ubiquitinated state and will not be degraded; the amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes:
  • the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The substance is identified as a phosphorylation inhibitor of CREPT protein, which is otherwise not a phosphorylation inhibitor of CREPT protein.
  • mass spectrometry or immunoprecipitation can be used to check the phosphorylation levels of S134 and S166 of the CREPT protein; preferably, the immunoprecipitation method can include: using the method to recognize the S134 and S166 of the CREPT protein. Spot phosphorylated anti-phosphorylated antibodies for immunoprecipitation.
  • the present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby causing the CREPT protein to function in cells.
  • the method includes: i) adding the substance to be identified to eukaryotic cells expressing CREPT protein and culturing the eukaryotic cells, and ii) using an immunoprecipitation method to check the eukaryotic cells
  • the ubiquitination level of CREPT protein compared with the ubiquitination level of CREPT protein in control cells that have not been treated with the substance, if the ubiquitination level of CREPT protein in the treated cells decreases, for example, decreases by more than 10% , more than 20%, more than 30% or more than 40%, then the substance is identified as a phosphorylation inhibitor of CREPT protein, otherwise the substance is not a phosphorylation inhibitor of CREPT protein.
  • step i) may further include: using prediction tools SwissTargetPrediction and SEA to design the substance to be identified for CREPT. Additionally, step ii) may include quantifying the ubiquitination level of the CREPT protein using an anti-CREPT antibody that recognizes the CREPT protein and a ubiquitin antibody that recognizes ubiquitin.
  • the invention provides homologous proteins of human CREPT derived from non-human eukaryotes, which have phosphorylation inactivation at the homology sites corresponding to positions 134 and 166 of SEQ ID No: 4 Modification, the phosphorylation inactivation modification makes the 134 and 166 positions corresponding to SEQ ID No: 4 when the homologous protein is located in the G1 end phase or G1/S transition phase cells of the eukaryotic organism.
  • the homology site remains in a non-phosphorylated state and the homologous protein remains in a non-ubiquitinated state, so that the homologous protein will not be degraded in the cell, leading to cell cycle arrest and apoptosis. Death.
  • the eukaryotic organism may be yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana.
  • the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 may be amino acid mutation and/or chemical modification.
  • the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 may be to mutate serine to alanine.
  • amino acid sequence of the homologous protein may be SEQ ID No: 6.
  • the invention provides a protein selected from:
  • the invention provides nucleic acids encoding proteins of the ninth and tenth aspects.
  • the present invention provides a vector comprising the nucleic acid according to the eleventh aspect.
  • the present invention provides cells comprising the vector of the twelfth aspect.
  • the present invention provides a method for identifying eukaryotic cells in the late G1 phase or the G1/S transition phase, the method comprising:
  • sequence of the human CREPT protein is SEQ ID No: 4.
  • the detectable label may be an isotope label, a fluorescent label or a quantum dot label or a label that can be further combined with an isotope label, a fluorescent label or a quantum dot label, preferably GFP.
  • the eukaryotic cell may be a human, yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell.
  • the present invention provides a method for inhibiting the degradation of CREPT protein in eukaryotic cells, the method comprising:
  • Phosphorylation inactivation modification is performed on the 134th and 166th positions of the CREPT protein, so that when the modified protein is located in eukaryotic cells at the end of G1 phase or the G1/S transition phase, the 134th and 166th positions remain inactive. phosphorylation state and the modified protein remains in a non-ubiquitinated state, thereby preventing the modified protein from being degraded.
  • the eukaryotic cell can be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell .
  • the phosphorylation inactivating modifications at the 134 and 166 positions may be amino acid mutations and/or chemical modifications.
  • the phosphorylation inactivating modifications at the 134 and 166 positions are Serine (S) is mutated into alanine (A).
  • the SKP2 inhibitor can be a double-stranded siRNA against SKP2, the sequence of which is AAUCUAAGCCUGGAAGGCCUGdTdT; the CUL1 inhibitor can be a double-stranded siRNA against CUL1, and the sequence of the neddylation inhibitor can be UAGACAUUGGGUUCGCCGUdTdT; It's MLN4924.
  • the present invention also provides a protein selected from: 1) a protein obtained by mutating serine 166 of SEQ ID No: 4 to alanine; 2) a protein obtained by mutating SEQ ID No: 8 A protein obtained by mutating serine 136 of SEQ ID No: 8 to alanine; 3) A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; and 4) Any one of 1) to 3) A protein with more than 90% sequence identity and the same alanine mutation.
  • the invention also provides nucleic acids encoding the proteins and vectors containing the nucleic acids.
  • Figures 1A-1E show that the expression levels of CREPT oscillate during the cell cycle;
  • Figure 1A Representative fluorescence images of CREPT in tumor cells. White circles indicate cells without CREPT expression. Scale bar, 10 ⁇ m.
  • Figure 1B Time-lapse microscopy live cell image of cells with GFP-CREPT knock-in. Scale bar, 10 ⁇ m.
  • Figure 1C CREPT expression during the cell cycle; DLD1 cells were synchronized to G1/S phase and released with 2mM double thymidine block (DTB). Cell lysates were collected at the indicated time points and analyzed by immunoblotting. Detect Cyclin A/B1/E, SKP2 to confirm cell cycle progression.
  • FIG. 1D Representative fluorescence image of CREPT in DTB-synchronized DLD1 Fucci cells. Scale bar, 10 ⁇ m.
  • FIG. 1E Quantification of CREPT, EdU and CDT1 expression levels in fluorescence images (Fig. 1D). Statistical significance (*P ⁇ 0.05; **P ⁇ 0.01; ***P ⁇ 0.001, ****P ⁇ 0.0001); P>0.05, not significant [n.s.], generated by t test.
  • Figures 2A-2K show that the degradation of CREPT during the G1/S transition phase is dependent on ubiquitin modification;
  • Figure 2A CREPT expression in cycloheximide (CHX)-treated cells. CREPT degradation begins 10 hours after CHX treatment.
  • Figure 2B CREPT expression levels remained stable after MG132 treatment. DLD1 cells were treated with or without 25 ⁇ g/ml MG132 for 4 h before the second thymidine release.
  • Fig. 2C Quantitative immunoblotting results of CREPT in Fig. 2B.
  • Figure 2D CREPT can be ubiquitinated. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids.
  • FIG. 2E K11-polyubiquitination-mediated degradation of CREPT. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated constructs.
  • Figure 2F K11-type ubiquitination accelerates CREPT degradation. HeLa cells were transfected with HA-Ub-K11 plasmid and treated with CHX.
  • Figure 2G CREPT immunoblot quantification results in Figure 2F.
  • Figure 2H K48-type ubiquitination has no effect on CREPT degradation. HeLa cells were treated with CHX and transfected with HA-Ub-K48 plasmid.
  • Figure 2I CREPT immunoblot quantification results in Figure 2H.
  • Figure 3A- Figure 3M show that CRL1 SKP2 directly ubiquitinates CREPT during the G1/S transition phase;
  • Figure 3A SKP2 has the highest binding affinity to CREPT.
  • CREPT pull-down proteins in synchronized cells were analyzed by mass spectrometry. The proteins are ranked based on unique peptide and log10 coverage. The top 10 proteins with known E3 ligase activity are shown. The area of the dots represents the relative coverage value.
  • Figure 3B Interaction between exogenous CREPT and exogenous SKP2.
  • Cell extracts from HEK293T cells expressing HA-CREPT and Flag-SKP2 were immunoprecipitated with anti-HA beads and subjected to immunoblot analysis with the indicated antibodies.
  • FIG. 3C Interaction between endogenous CREPT and endogenous SKP2.
  • Cell extracts from DLD1 cells were immunoprecipitated with anti-CREPT beads and subjected to immunoblot analysis with the indicated antibodies.
  • Figure 3D Purified prokaryotes expressing SKP2 and CREPT were used in in vitro co-IP assays followed by immunoblot analysis.
  • Figure 3E SKP2 mediates ubiquitination of CREPT. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids, with or without treatment of MG132.
  • FIG. 3F and
  • FIG. 3G Effect of overexpression of SKP2 on CREPT degradation.
  • FIG. 3H Effect of knockdown of SKP2 on CREPT degradation.
  • FIG. 3J and (Fig. 3K) SKP2 and CREPT levels after synchronization and release of cells into G1/S phase.
  • FIG. 3L and (Fig. 3M) SKP2 and CREPT levels after synchronizing cells to M phase and releasing them.
  • Figures 4A-4G show that the S134A and S166A mutations of CREPT are unable to interact with Ub;
  • CREPT is ubiquitinated in the CID domain. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids and treated with MG132.
  • Figure 4B CREPT modification was identified by mass spectrometry analysis. Modified mass spectrometry analysis was performed in 293T cells synchronized to G1/S phase, and the phosphorylation level of the serine (S) site on the CREPT protein was ranked according to #PSMs.
  • Figure 4C Amino acid schematic diagram of CREPT and RTT103 in the CREPT 135-170 region.
  • FIG. 4D The CREPT S134A/S166A mutant cannot be ubiquitinated. Immunoassay of ubiquitinated CREPT and CREPT mutants overexpressed in 293T cells.
  • Figure 4E SKP2 recognizes the phosphorylated form of CREPT. 293T cells were transfected with the indicated plasmids and harvested for co-IP assays followed by immunoblot analysis.
  • Figure 4F Interaction between endogenous CREPT and endogenous CDK2/Cyclin E. Cell extracts from DLD1 cells were immunoprecipitated with anti-CREPT beads and subjected to immunoblot analysis with the indicated antibodies.
  • FIG. 4G CREPT is phosphorylated by CDK2/Cyclin E1 at S134/S166.
  • Flag-CDK2 and Flag-Cyclin E1 were purified by Co-IP using anti-Flag antibodies from HEK293T cells transfected with the expression vectors of Flag-CDK2 and Flag-Cyclin E1.
  • GST, GST-CREPT and GST-CREPT(S134A/S166A) were purified from prokaryotic expression systems using GST beads. Phosphorylation of GST or GST-tagged CREPT and its mutants was detected with a universal anti-phosphorylation antibody (top).
  • CREPT(SA) CREPT(S134A/S166A).
  • Figures 5A-5G show that CREPT (S134A/S166A) mutation causes cell apoptosis;
  • Figure 5A CREPT (S134A/S166A) mutant causes cell death.
  • HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 h and then stained with Annexin V and PI for FACS analysis.
  • Figure 5B Statistical analysis of the flow cytometry data in Figure 5A.
  • Figure 5C Cell death is triggered by apoptosis.
  • HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 hours and then subjected to Western blotting.
  • FIG. 5D Cell growth inhibition was caused by CREPT(S134A/S166A) overexpression. Cell viability was determined by CCK-8 assay 48 hours after transfection of HeLa wild-type (Mock) and CREPT knockout (KO) cells with the indicated plasmids.
  • FIG. 5F CREPT(S134A/S166A) mutant is lethal to S. cerevisiae.
  • Figures 6A-6G show that non-degraded mutant CREPT prevents cells from entering S phase;
  • Figure 6A CREPT does not bind to the MCM complex during the G1/S transition phase. ChIP-MS analysis was performed in DLD1 cells to rank proteins according to #PSMs. Cell cycle synchronization with DTB.
  • Figure 6B Interaction between exogenous CREPT and exogenous MCM5. Cell extracts from HEK293T cells expressing Myc-CREPT and Flag-MCM5 were immunoprecipitated with anti-Myc beads and subjected to immunoblot analysis with the indicated antibodies.
  • Figure 6C Interaction between endogenous CREPT and endogenous MCM5.
  • CREPT(WT) represents CREPT wild-type plasmid
  • CREPT(SA) represents CREPT(S134A/S166A) mutant
  • CREPT(SE) represents CREPT(S134E/S166E) mutant.
  • Figures 7A-7D show that non-degraded CREPT halts replication forks;
  • Figure 7A Representative fluorescence images of CREPT in HeLa cells. Cells were transfected with indicated plasmids for 12 h. Immunofluorescence assay RPA2 quantitative results (right picture). Scale bar, 10 ⁇ m.
  • Figure 7B Representative fluorescence image of CREPT in HeLa cells. Quantitative results of p-RPA2 by immunofluorescence assay (right panel). Scale bar, 10 ⁇ m.
  • FIG. 7C DNA replication rates in WT and S134A/S166A mutants treated with HU. Asynchronous HeLa CREPT KO cells were first treated with 4mM HU for 4 hours.
  • CIdU was then added to the culture for 0.5 h, and cells were harvested for DNA fiber analysis to measure the length and distribution of CIdU fibers. Scale bar, 10 ⁇ m.
  • Fig. 7D Percentage of aborted replication forks in WT and S134A/S166A mutants during HU block and release. DNA fiber assay was performed in HeLa CREPT KO cells to measure aborted replication forks. Statistical significance (*P ⁇ 0.05; **P ⁇ 0.01; ****P ⁇ 0.0001) was calculated by t-test.
  • CREPT(WT) represents CREPT wild-type plasmid
  • CREPT(SA) represents CREPT(S134A/S166A) mutant.
  • Figure 8 shows a schematic diagram of the mechanism of action of CREPT in regulating the cell cycle.
  • Figures 9A-9G show that the expression level of CREPT oscillates in the cell cycle, wherein ( Figure 9A) Schematic diagram of GFP-CREPT fusion protein knock-in into Hela cell line. GFP is fused after the ATG sequence in EXON1 of CREPT. (Fig. 9B) Western blotting confirmed that GFP-CREPT expression was knocked into cells. (Fig. 9C) FACS analysis of synchronized DLD1 cells by PI staining at the indicated release time points. (Fig. 9D) CREPT expression during the cell cycle. HeLa cells are synchronized to G1/S phase and released via DTB. ( Figure 9E) Real-time quantitative PCR analysis of CREPT mRNA expression at the indicated release time points after DTB treatment.
  • FIG. 9F Representative fluorescence image of CREPT in DTB-synchronized DLD1 Fucci cells. Scale bar 10 ⁇ m.
  • Figure 9G Quantification results of CREPT and CDT1 expression levels in fluorescence images ( Figure 9F). Statistically significant (****P ⁇ 0.0001); P>0.05, not significant [n.s.]), calculated by t test.
  • Figures 10A-10D show that CREPT is degraded through the ubiquitin pathway during the G1/S transition phase;
  • Figure 10A Quantitative immunoblotting results of CREPT in Figure 2A.
  • Figure 10B FACS analysis of synchronized DLD1 cells with or without MG132 treatment.
  • Figure 10C CREPT degradation is not mediated by autophagy.
  • DLD1 cells were treated with the lysosomal inhibitors chloroquine or leupeptin 6 hours before the second thymidine release.
  • Figure 10D HeLa cells were treated with the lysosomal inhibitors chloroquine or leupeptin 6 hours before the second thymidine release.
  • Figures 11A-11I show that CRL1 SKP2 directly ubiquitinates CREPT during the G1/S transition.
  • Figure 11A GST-CREPT expression was verified. Gels with purified GST-CERPT protein were stained with Coomassie blue.
  • Fig. 11B The expression of His-SKP1 and His-SKP2 was verified. Gels with purified His-SKP1 and His-SKP2 proteins were stained with Coomassie blue.
  • Fig. 11C The expression of His-SKP2 was verified. Immunoassays were performed on purified SKP2 protein at different purification steps.
  • Figure 1 ID FACS analysis of synchronized DLD1 cells by PI staining at the indicated release time points.
  • Figures 12A-12E show that the S134A and S166A mutations of CREPT are unable to interact with Ub;
  • Figure 12A CREPT ubiquitination is independent of single lysine (K) mutations in the CID domain. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids.
  • Figure 12B CREPT ubiquitination does not depend on a single K mutation in the linker region. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids.
  • Figure 12C CREPT ubiquitination is independent of K.
  • CREPT ubiquitination assays were performed in 293T cells transfected with the indicated plasmids and harvested for co-IP assays.
  • Figure 12D CREPT ubiquitination is independent of threonine (T) or cysteine (C).
  • In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids.
  • Figure 12E CREPT is phosphorylated in eukaryotic cells. Purified GST-CREPT in prokaryotic cells (E. coli) and eukaryotic cells (mammalian) was examined by Western blotting using a universal anti-phospho-antibody.
  • p-CREPT(S/T/Y) stands for pan-phosphorylated antibody.
  • Figures 13A-13F show that CREPT (S134A/S166A) mutation leads to apoptosis;
  • Figure 13A Cell growth inhibition and cell death caused by undegraded CREPT mutant protein. HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 hours.
  • Figure 13B Overexpression of CREPT (S134A/S166A) resulted in cell growth inhibition. Cell viability was determined by CCK-8 assay 48 hours after transfection of 293T and NCM460 cells with the indicated plasmids.
  • FIG. 13C and
  • FIG. 13D CREPT(S134A/S166A) mutations result in reduced clonogenicity.
  • Figures 14A-14G show that undegraded CREPT prevents cells from entering S phase;
  • CREPT interacts with MCM2. 293T cells were transfected with the indicated plasmids and harvested for co-IP.
  • Figure 14B CREPT interacts with MCM7. 293T cells were transfected with the indicated plasmids.
  • Figure 14C In the presence of cross-linking, CREPT binds to endogenous MCM5. Endogenous co-IP assay in DLD1 cells. Cells were fixed with 1% paraformaldehyde.
  • CREPT does not bind directly to chromatin.
  • FIG. 14E FACS analysis of CREPT KO HeLa cells transfected with the indicated plasmids for 12 hours.
  • FIG. 14F MCM5 quantification results of immunofluorescence assay in Fig. 6G.
  • Figure 14G Nucleus diameter in Figure 6G.
  • WT CREPT(WT); SA: CREPT(S134A/S166A).
  • Figures 15A-15D show that non-degraded CREPT halts replication forks;
  • Figure 15A Representative fluorescent images of TUNEL signal in HeLa cells. Tunel staining of HeLa wild-type (Mock) and CREPT knockout (KO) cells when overexpressing the indicated plasmids, CREPT (SA): CREPT (S134A/S166A) mutation, CREPT (SE): CREPT (S134E/S166E) mutation . Scale bar, 10 ⁇ m.
  • Figure 15B Representative fluorescence image of ⁇ H2AX in HeLa cells.
  • Figure 16 is the screening results of small molecule inhibitors of CREPT phosphorylation and ubiquitination.
  • #1 to #5 above indicate cells treated with candidate compounds #1 to #5, and the numbers in the middle (0.1 to 1.1) are the relative ubiquitination levels of each sample.
  • Figure 17 shows the effect of candidate compounds #1 to #5 on the proliferation of DLD1 cells (A) and MGC803 cells (B).
  • human CREPT protein or “CREPT protein” used herein refers to human wild-type CREPT protein unless otherwise specified.
  • CREPT protein variant and “modified CREPT protein” used herein refer to protein variants obtained by subjecting the wild-type CREPT protein to amino acid mutations and/or chemical modifications.
  • homologous protein refers to proteins with homologous amino acid sequences that perform the same or similar functions in different organisms.
  • phosphorylation-inactivating modification used in this article means that the modified amino acid residues in the protein can continue to maintain or simulate the non-phosphorylated state in eukaryotic cells. The residues modified by phosphorylation-inactivating cannot be truly modified. Kinase phosphorylation in nuclear cells.
  • a method for identifying eukaryotic cells in the late G1 phase or the G1/S transition phase comprising:
  • sequence of the human CREPT protein is SEQ ID No: 4.
  • the detectable label is an isotope label, a fluorescent label or a quantum dot label or a label that can be further combined with an isotope label, a fluorescent label or a quantum dot label, preferably GFP.
  • the eukaryotic cell is human, yeast, mouse, canine, feline, chicken, toad, zebra Fish, Drosophila, nematode or Arabidopsis thaliana cells, preferably human cells or yeast cells.
  • the inventor further discovered that this degradation of CREPT protein during the G1/S transition phase of cells relies on ubiquitination-mediated proteasomal degradation.
  • the ubiquitination is catalyzed by the E3 ligase CRL1 SKP2 , and the recognition and catalysis of SKP2 Depends on the phosphorylation of CREPT protein at S134 and S166.
  • the inventor simulated the phosphorylation and non-phosphorylation states of CREPT at sites 134 and 166 by mutating these two sites, and found that the CREPT protein that remained unphosphorylated at these two sites remained in a non-ubiquitinated state.
  • the non-phosphorylated and non-ubiquitinated S134A/S166A double mutant protein will not be degraded at the end of G1 or the G1/S transition phase.
  • This non-degraded CREPT protein variant will not be separated from the MCM complex, resulting in cell Blocking in the late G1 phase or the G1/S transition phase with high expression of the MCM complex results in the cell cycle being unable to enter the S phase and stalling the progression of DNA replication forks, ultimately leading to cell death ( Figure 5A- Figure 5G).
  • the S134A mutation alone or the S166A mutant protein can affect ubiquitination to a certain extent (Fig. 4D).
  • the S134E/S166E double mutant protein that simulates the phosphorylation state has no effect on cell ubiquitination and survival ( Figure 4D, Figure 5A- Figure 5B), indicating that it is the same as the wild-type CREPT protein at the G1/S transition. degrade normally.
  • Figure 4D Figure 5A- Figure 5B
  • CREPT protein variants that maintain non-phosphorylation at positions 134 and 166 and maintain non-ubiquitination of the protein cannot be degraded at the end of G1 phase or the G1/S transition phase, leading to cell death.
  • persistent activating mutations include mutating residues to aspartic acid (D) or glutamic acid (E), since these two amino acids are the only negatively charged amino acids;
  • the most common persistent inhibitory mutation that is, a mutation that simulates non-phosphorylation is to mutate serine to alanine (A), because alanine is positively charged and can continuously inhibit the activity of this residue site.
  • activating chemical modifiers may include phosphate donors such as acetyl phosphate, phosphoramide salts, carbamoyl phosphates, and sodium pyrophosphate, as well as beryllium trifluoride.
  • phosphate donors such as acetyl phosphate, phosphoramide salts, carbamoyl phosphates, and sodium pyrophosphate, as well as beryllium trifluoride.
  • some CDK4/6-specific small molecule inhibitors such as Palbociclib, Ribociclib or Abemaciclib can also achieve the effect of keeping the protein non-phosphorylated (Maiani et al., 2021; Simoneschi et al., 2021).
  • the present invention provides a non-phosphorylated and non-ubiquitinated CREPT protein variant.
  • the CREPT protein is located in a eukaryotic cell at the end of G1 or G1/S transition phase, 134 and 166 of the CREPT protein The site is able to remain unphosphorylated and the protein remains unubiquitinated, thereby preventing the protein variant from being degraded at this stage.
  • the inventors also found that the effect of the S134A/S166A mutant form of human CREPT protein on inducing cell apoptosis also exists in other eukaryotes.
  • the inventors exogenously expressed human CREPT S134A/S166A in Saccharomyces cerevisiae.
  • the protein variant impaired the survival of yeast at different temperatures ( Figure 13F).
  • human CREPT S134A/S166A was also exogenously expressed in the Rtt103-deficient yeast strain, and the results also significantly blocked the growth of yeast (Figure 5F).
  • CREPT protein and its homologous proteins are universally applicable in eukaryotes. It also shows that exogenous introduction of CREPT mutants can control the cell cycle of eukaryotic cells, preferably tumor cells, and Induces apoptosis.
  • another aspect of the invention provides a protein obtained by phosphorylation inactivation modification of residues 134 and 166 in the CREPT protein sequence (SEQ ID NO: 4), the phosphorylation inactivation modification So that when the protein is located in eukaryotic cells in the late G1 phase or the G1/S transition phase, the 134 and 166 positions remain in a non-phosphorylated state and the protein remains in a non-ubiquitinated state, so that the protein in It is not degraded in eukaryotic cells.
  • the eukaryotic cell is a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell .
  • the phosphorylation-inactivating modifications at positions 134 and 166 are amino acid mutations and/or chemical modifications.
  • the phosphorylation inactivation modification at positions 134 and 166 is to mutate serine (S) to alanine (A).
  • the amino acid sequence of the protein is SEQ ID No: 2 , the S134A/S166A double mutant form of CREPT.
  • a tag sequence or a guide sequence can be connected to the N-terminus and/or C-terminus of the protein.
  • the linkage is a covalent linkage.
  • the protein with a tag sequence or leader sequence is a fusion protein.
  • the protein with a tag sequence or leader sequence is a conjugated protein.
  • the tag sequence may be, for example, a purification tag, a fluorescent tag, a solubilization tag, an affinity tag, an epitope tag, or the like.
  • the guide sequence may be a polypeptide sequence that guides the protein across the cell membrane into the cell, including, for example, cell-penetrating peptides that are not based on endocytosis, and peptide sequences that are themselves prone to enter cells via endocytosis. or protein sequence.
  • the present invention also provides proteins that have the same phosphorylation inactivation modifications at positions 134 and 166 as the above-mentioned proteins and have a sequence identity of more than 90%, more than 95%, preferably more than 98% or more than 99%.
  • the invention provides nucleic acids encoding the above-mentioned proteins, vectors comprising the nucleic acids, and cells comprising the vectors.
  • the method of introducing the target protein (such as CREPT S134A/S166A of the present invention) into target cells may include introducing a vector expressing the target protein into the target cells through transfection, infection or other means, or chemical modification may also be used mRNA (modRNA) to achieve the expression of target proteins in target cells.
  • the target protein can be directly introduced into cells using, for example, the guide sequence described above.
  • the present invention is not limited to this.
  • precision gene editing technology such as prime editors
  • the present invention provides the use of the above-mentioned protein, nucleic acid or vector in preparing reagents for inhibiting eukaryotic cell proliferation, inhibiting DNA replication of eukaryotic cells, regulating eukaryotic cell cycle or killing eukaryotic cells.
  • the eukaryotic cell is a human, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, yeast or Arabidopsis thaliana cell.
  • the present invention provides the use of the above-mentioned proteins, nucleic acids or vectors in the preparation of anti-cancer drugs.
  • the present invention provides a method of treating cancer, the method comprising administering an effective amount of the above-mentioned protein, nucleic acid or vector to a human subject; alternatively, the method comprising utilizing a CRISPR/Cas9-based gene
  • the editing technology edits the CRPET gene in the genome of the subject's cancer cells so that the cancer cells express the protein of SEQ ID NO: 2.
  • the cancer is liver cancer, kidney cancer, gastric cancer, or colorectal cancer.
  • the method includes introducing the above-described nucleic acid into a tumor cell.
  • expression of wild-type CREPT is reduced or eliminated in cancer cells of the subject before, during, or after administration of an effective amount of the protein, nucleic acid, or vector to the subject.
  • the present invention provides a pharmaceutical composition, which includes: the above-mentioned protein, nucleic acid or carrier, and a pharmaceutically acceptable carrier, excipient or medium.
  • the pharmaceutical composition is used to treat cancer, such as liver, kidney, stomach or colorectal cancer.
  • Another aspect of the present invention provides a method for screening non-phosphorylation and non-ubiquitination of the CREPT protein.
  • a method of modifying the agent or a method of identifying whether the substance is a phosphorylation inhibitor of the CREPT protein wherein the modifying agent or inhibitor maintains the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby making the CREPT protein Maintains a non-ubiquitinated state in cells and will not be degraded; especially when the CREPT protein is located in eukaryotic cells at the end of G1 phase or G1/S transition phase, the S134 and S166 sites can remain continuously non-phosphorylated state and the protein can maintain a non-ubiquitinated state, so that the protein will not be degraded; wherein, the amino acid sequence of the CREPT protein is SEQ ID No: 4.
  • the method can be performed as follows: adding a candidate modifier that simulates a non-phosphorylated state or a substance to be identified into eukaryotic cells expressing CREPT protein that are synchronized to the G1 phase, and then releasing and culturing the eukaryotic cells, and the phosphorylation levels of S134 and S166 of the CREPT protein were examined when the eukaryotic cells were alive.
  • the method can be performed as follows: in vitro, a candidate modifier that simulates a non-phosphorylated state or a substance to be identified is incubated with the CREPT protein, and the results are examined under catalytic conditions of Cyclin E/CDK2 kinase.
  • the phosphorylation levels of S134 and S166 of CREPT protein were described.
  • the phosphorylation levels of S134 and S166 of the CREPT protein treated in step i) or ii) decrease relative to the respective untreated control, for example, by more than 10%, more than 20%, or 30% or more than 40%, preferably more than 50%, more than 60%, more than 70%, more than 80% or more than 90%, then the candidate modifier is screened as a non-phosphorylation non-ubiquitination modifier of the CREPT protein, Or the substance is identified as a phosphorylation inhibitor of the CREPT protein; otherwise the candidate modifier is not a non-phosphorylation non-ubiquitination modifier of the CREPT protein, and the substance is not a phosphorylation inhibitor of the CREPT protein.
  • mass spectrometry or immunoprecipitation can be used to check the phosphorylation status of the S134 and S166 sites of the CREPT protein.
  • the immunoprecipitation method may include: using anti-phosphorylation antibodies that recognize the phosphorylation of S134 and S166 of the CREPT protein. Immunoprecipitation was performed with the body to quantify the phosphorylation level of the S134 site of CREPT protein. Specifically, anti-CREPT antibodies can be used to precipitate CREPT proteins, and the total amount of CREPT proteins can be measured as the background amount. For proteins immunoprecipitated with anti-CREPT antibodies, anti-phosphorylated antibodies can be used to detect phosphorylated proteins and detect them. For quantification, the phosphorylation level can be expressed as the amount of phosphorylated protein relative to the background amount of CREPT protein, and can be normalized to a control.
  • the invention also relates to the following compounds:
  • the cancer is melanoma, liver cancer, kidney cancer, gastric cancer, or colorectal cancer.
  • the present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby making the CREPT protein Maintains a non-ubiquitinated state in cells and will not be degraded;
  • the amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes: i) adding the substance to be identified into eukaryotic cells expressing the CREPT protein and Culturing the eukaryotic cells, and ii) using an immunoprecipitation method to examine the ubiquitination level of the CREPT protein in the eukaryotic cells; compared with the ubiquitination level
  • step ii) may further include: using prediction tools SwissTargetPrediction and SEA to design the substance to be identified for CREPT. Additionally, step ii) may include quantifying the ubiquitination level of the CREPT protein using an anti-CREPT antibody that recognizes the CREPT protein and a ubiquitin antibody that recognizes ubiquitin.
  • the ubiquitination level can be a relative value of the amount of ubiquitinated protein relative to the total amount of CREPT protein and can be normalized to a control.
  • S134 and S166 in CREPT correspond to S136 and S174 in yeast Rtt103 (see Figure 4C), and the inventors found that overexpression of the Rtt103S136A/S174A double mutant protein resulted in a lethal phenotype of Rtt103-deficient yeast ( Figure 5G).
  • the invention provides homologous proteins of human CREPT derived from non-human eukaryotes, which have a loss of phosphorylation at homology sites corresponding to positions 134 and 166 of the human CREPT protein.
  • Inactive modification the phosphorylation inactivating modification enables the homologous site to remain in an unphosphorylated state and the homologous site remains unphosphorylated when the homologous protein is located in cells at the end of G1 phase or G1/S transition phase of the eukaryotic organism.
  • the homologous protein remains in a non-ubiquitinated state, so that the homologous protein will not be degraded in the cell, leading to cell cycle arrest and apoptosis.
  • the eukaryotic organism is yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, or Arabidopsis thaliana.
  • the phosphorylation-inactivating modifications on the homology sites corresponding to positions 134 and 166 of the human CREPT protein are amino acid mutations and/or chemical modifications. In one embodiment, the phosphorylation-inactivating modification at the homology site corresponding to positions 134 and 166 of the human CREPT protein is to mutate serine to alanine. In one embodiment, the homologous protein is Saccharomyces cerevisiae Rtt103 S136A/S174A double mutant protein, whose amino acid sequence is SEQ ID No: 6.
  • the N-terminus and/or C-terminus of the homologous protein can be connected with a tag sequence or a leader sequence to form, for example, a fusion protein or a conjugated protein.
  • the protein has the same phosphorylation inactivation modification at the homology site and has a sequence identity of more than 90%, more than 95%, preferably more than 98% or more than 99%. of protein.
  • the present invention provides nucleic acids encoding the above-mentioned homologous proteins, vectors containing the nucleic acids, and cells containing the vectors.
  • the present invention provides the use of the above homologous proteins, nucleic acids or vectors in preparing reagents for inhibiting eukaryotic cell proliferation, inhibiting DNA replication of eukaryotic cells, regulating eukaryotic cell cycle or killing eukaryotic cells.
  • the eukaryotic cell is a human, yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, or Arabidopsis thaliana cell.
  • the present invention provides a method for inhibiting the degradation of CREPT protein in eukaryotic cells, the method comprising: 1) using an inhibitor selected from the group consisting of SKP2 inhibitors, CUL1 inhibitors, neddylation inhibitors and CDK2 inhibitors
  • the inhibitor enters the eukaryotic cells expressing CREPT; and/or 2) phosphorylation and inactivation modification of the 134th and 166th positions of the CREPT protein, so that when the modified protein is located at the end of G1 phase or the G1/S transition phase,
  • the 134 and 166 sites remain in a non-phosphorylated state and the modified protein remains in a non-ubiquitinated state, thereby preventing the modified protein from being degraded.
  • the eukaryotic cell may be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell cell.
  • the phosphorylation inactivating modification at the 134 and 166 positions may be an amino acid mutation and/or a chemical modification, preferably, the mutation of serine (S) to alanine (A).
  • the SKP2 inhibitor can be a double-stranded siRNA directed against SKP2, and its sequence can be AAUCUAAGCCUGGAAGGCCUGdTdT;
  • the CUL1 inhibitor can be a double-stranded siRNA directed against CUL1, and its sequence can be UAGACAUUGGGUUCGCCGUdTdT;
  • the neddylation inhibitor can be MLN4924.
  • the single mutant proteins S134A or S166A of CREPT can inhibit ubiquitination to a certain extent. Therefore, it can be understood that similar single mutations of these single mutant proteins and homologous proteins in other eukaryotic cells All have the function of inhibiting CREPT degradation.
  • the present invention also provides the following proteins: 1) a protein obtained by mutating serine 166 of SEQ ID No: 4 to alanine; 2) a protein obtained by mutating serine 136 of SEQ ID No: 8 A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; 3) A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; and 4) A protein having 90% affinity with any one of 1) to 3) Proteins with more than % sequence identity and the same alanine mutation.
  • the present invention also provides nucleic acids encoding the proteins and vectors containing the nucleic acids.
  • SEQ ID NO:1 Nucleic acid sequence encoding CREPT S134A/S166A double mutant protein
  • SEQ ID NO:2CREPT Amino acid sequence of S134A/S166A double mutant protein
  • SEQ ID NO:4 Amino acid sequence of wild-type CREPT
  • SEQ ID NO:5 Nucleic acid sequence encoding Rtt103 S136A/S174A double mutant protein
  • SEQ ID NO: 6Rtt103 Amino acid sequence of S136A/S174A double mutant protein
  • SEQ ID NO:8 Amino acid sequence of wild-type Rtt103.
  • Example 1 CREPT degrades in the G1/S transition phase and recovers in the S phase
  • IF staining experiments were performed on DLD1 and HeLa cells to determine the expression pattern of CREPT in tumor cells. It was found that most tumor cells expressed abundant CREPT, but a few tumor cells were CREPT negative ( Figure 1A, see dotted circle). Moreover, the nuclei of CREPT-negative tumor cells were slightly larger and evenly stained with DAPI ( Figure 1A, DAPI staining). Therefore, CREPT-negative tumor cells may be due to the loss of CREPT during specific cell cycle stages. To test this hypothesis, the inventors used CRISPR-Cas9 to generate HeLa cells with GFP-CREPT knocked in (Figure 9A, Figure 9B). Live-cell imaging analysis showed that GFP-CREPT remained in the cells for a period of time, then disappeared for nearly 30 minutes and then recovered ( Figure 1B). This suggests that CREPT protein levels oscillate during the tumor cell cycle.
  • double thymidine block was used to synchronize DLD1 and HeLa cells to the G1/S transition phase and release them to different cell cycle time points.
  • Fluorescence-activated cell sorting (FACS) analysis showed that more than 90% of cells were synchronized in G1 phase after DTB treatment (Fig. 9C).
  • Western blotting showed that CREPT protein was almost undetectable during the G1/S transition, but it increased during S phase (Fig. 1C and Fig. 9D, lane 1, 0 and 1 h).
  • This change in CREPT protein was accompanied by opposite trends for Cyclin E and SKP2 but similar to the expression patterns of Cyclin A and Cyclin B1 ( Figure 1C and Figure 9D).
  • the mRNA level of CREPT remained unchanged from G1/S transition to S phase (Fig. 9E).
  • K63 ubiquitin was also able to moderately induce CREPT ubiquitination (see Figure 2E, lanes 6 and 12). This result was unexpected because K48 ubiquitin has been widely reported to mediate protein degradation.
  • the inventors further overexpressed different types of ubiquitin together with CREPT in the presence of CHX. The results showed that CREPT was reduced in the presence of K11 but not K48 or K63 ubiquitin ( Figure 2F- Figure 2K).
  • the inventors treated DLD1 and HeLa cells with autophagy inhibitors CQ and LEU to synchronize DLD1 and HeLa cells to the G1/S phase.
  • E3 ligases targeting CREPT To identify E3 ligases targeting CREPT, the inventors synchronized cells to G1/S phase under MG132 treatment to precipitate CREPT-interacting proteins. Mass spectrometry analysis revealed the presence of several E3 ligases in the precipitated complex. Among the top 10 E3 ligases that potentially interact with CREPT, SKP2 has the highest probability of binding to CREPT (Fig. 3A). To find out whether SKP2 is the E3 ligase used for CREPT degradation, the inventors verified their interaction under different conditions. IP experiments showed that antibodies against HA precipitated HA-CREPT and Flag-SKP2 ( Figure 3B), indicating that HA-CREPT interacts with Flag-SKP2.
  • SKP2 belongs to the F-box family of SCF complex proteins
  • CTL1-RING E3 ubiquitin ligase (CRL1) complex is involved in CREPT degradation
  • the inventors used the neddylation inhibitor MLN4924, which inhibits all Cullin-RING ligases. Activation of the complex.
  • Results CREPT protein levels increased in control cells (si Ctrl) but not in MLN4924-treated SKP2-depleted cells (si SKP2) (Fig. 11E).
  • Figure 11E shows that Cullin-RING ligase was gradually inactivated by MLN4924 over time (0-6 hours) in control cells.
  • SKP2 Although SKP2 always maintains a significant expression level, the ubiquitination level of CREPT gradually decreases and its degradation is inhibited, and its level gradually increases over time; while in cells depleted of SKP2 with siRNA, the ubiquitination and degradation of CREPT are always inhibited. , so it always remains at a significant level.
  • the above results indicate that the deletion of SKP2 can significantly inhibit the ubiquitination and degradation of CREPT.
  • Myc-CREPT-CID Myc-tagged CID domain
  • Myc CCT domain
  • Western blot analysis showed that Myc-CREPT-CID was ubiquitinated, but Myc-CREPT-CCT was not (Fig. 4A), indicating that ubiquitination occurs within the CID domain.
  • S134 and S166 regulates CREPT ubiquitination are located in the linker region of CREPT and are highly conserved between CREPT and its ortholog Rtt103 in S. cerevisiae (Fig. 4C).
  • the inventors generated different mutants, including S134A, S166A and S134A/S166A double mutations to simulate the non-phosphorylated state of loss of function, and S134E, S166E and S134E/S166E double mutations to simulate continuous phosphorylation.
  • Western blot analysis showed that mutations of S134A, S166A, and S134A/S166A impaired ubiquitination, but other mutations had no effect on ubiquitination (Fig.
  • CREPT amino acid sequence analysis of CREPT showed that both the S134 and S166 sites may be the recognition sites of the Cyclin E-CDK2 complex with sp sequence.
  • CREPT was also observed to interact with Cyclin E1 and CDK2 in IP experiments (Fig. 4F).
  • In vitro kinase assay showed that GST-CREPT was phosphorylated by CDK2 and Cyclin E1, but GST-CREPT (S134A/S166A) was not phosphorylated (Fig. 4G). It was also observed that GST-CREPT was phosphorylated in mammalian cells but not in E. coli (Fig. 12E).
  • the MCM hexamer contains 6 MCMs, including MCM2 to MCM7.
  • Myc-CREPT and Flag-MCM5 were overexpressed in 293T cells. IP experiments showed that Myc-CREPT and Flag-MCM5 interacted strongly in unsynchronized 293T cells ( Figure 6B).
  • Myc-CREPT was also observed to interact with Flag-MCM7 (Fig. 14A) and Myc-CREPT with Flag-MCM2 (Fig. 14B), indicating that CREPT binds to the MCM hexamer.
  • the inventors performed co-IP experiments using antibodies against CREPT under different cross-linking conditions (Figure 14C).
  • the inventor further used undegraded mutant CREPT to conduct IP experiments, and the results showed that the interaction between Myc-CREPT (S134A/S166A) and Flag-MCM5 was greater than the interaction between WT protein and Myc-CREPT (S133E/S166E) and Flag-MCM5. stronger (Fig. 6E). This result indicates that the degradation of CREPT leads to its dissociation from the MCM hexamer.
  • RPA2 replication protein A2
  • ssDNA single-stranded DNA
  • Fig. 7A see ctrl
  • Fig. 7A see ctrl
  • HA-CREPT, Flag-CREPT, Myc-CREPT, Myc-CREPT-CID, Myc-CREPT-CCT, GSTCREPT, GFP-P15RS and Myc-P15RS plasmids were constructed by the inventor's laboratory.
  • Flag-MCM2 plasmid was a gift from Dr. Kong Daochun (School of Life Sciences, Peking University).
  • pRK5-HA-UBI(#17608), pRK5-HA-UBI-K11(#22901), pRK5-HA-UBI-K48(#17605), pRK5-HA-UBI-K63(#17606) and pSpCas9(BB) -2AGFP (PX458, #48138) was purchased from Addgene.
  • Flag-SKP1 ⁇ Flag-SKP2, Flag-CUL1, Flag-MCM5 and Flag-MCM7 were generated from cDNA.
  • CREPT mutated plasmids were generated by site-directed mutagenesis (Muta-direct TM , SBS Genetech).
  • SKP2 and CUL1 siRNA duplexes were transfected by Lipofectamine RNAi MAX (Invitrogen), and the oligonucleotide sequences were AAUCUAAGCCUGGAAGGCCUGdTdT and UAGACAUUGGGUUCGCCGUdTdT, respectively.
  • sgRNA Short guide RNA
  • sgRNA sequence is CTCCTTCTCTGAGTCGGCGC.
  • Annealed sgRNA and BbsI-digested Px458 vector were ligated by solution I (Takara) to construct Cas9 DNA shearing plasmid.
  • the coding sequence of GFP was cloned and ligated into the PCDNA 3.1-HA vector to construct a GFP transcription plasmid.
  • HeLa cells were co-transfected with Cas9 DNA cleavage and GFP transcription plasmids.
  • GFP-positive HeLa cells were sorted by flow cytometry, and individual cells were seeded in 96-well plates to select DNA recombinant clones. One week later, cells were selected for expression of the genomically inserted GFP.
  • Cells were synchronized in G1/S phase by double thymidine block (DTB). Cells were treated with 2mM thymidine for at least 18 hours, released in fresh medium for 8 hours, and then treated with 2mM thymidine for at least another 16 hours. Cells were synchronized in G2/M phase by thymidine-nocodazole block. Cells were treated with 2mM thymidine for at least 24 hours, released for 3 hours, and then treated with 340 nM nocodazole for at least 16 hours. Cells were harvested at the times indicated. Verify the cell cycle stage of harvested cells by flow cytometric analysis. For MG132 treatment, MG132 was added to DLD1 cells 4 hours before harvest.
  • DTB double thymidine block
  • SKP2 protein is unstable in prokaryotic expression system.
  • In vitro protein interaction assay was performed using pET22b-SKP1, pET30A-SKP2 and GST-CREPT proteins. Immunoprecipitation assay verified the interaction between GST-CREPT and pET30A-SKP2.
  • GST-CREPT wild type or GST-CREPT (S134A/S166A) were used for in vitro kinase assay.
  • the protein was dissolved in kinase buffer (10mM HEPES (pH7.5), 50mM NaCl, 2mM MgCl) at 30°C. 2 , 1mM dithiothreitol, 1mM EGTA and 0.1mM ATP) and incubated with Myc-Cyclin E/Myc-CDK2 protein for 30 minutes. Stop the reaction using SDS loading buffer. Phosphorylation of CREPT was detected by Western blotting.
  • DNA fiber experiments were performed as previously described (Genois et al., 2021). Briefly, DLD1 cells were first labeled with 50mM CldU, washed twice with PBS and labeled with 250mM IdU. Harvest the cells and suspend them in cold PBS to a concentration of 1 to 1.5 ⁇ 10 cells/ml, then mix 3ul of cell solution with 7ul of spreading buffer (0.5% SDS, 200mM Tris-HCl pH7.4, 50mM EDTA) , and spread on silanized glass slides. Tilt the slide at a 30-60° angle to spread the fibers and leave at room temperature for 15 minutes. DNA fibers were fixed in methanol:acetic acid (3:1) for 20 minutes.
  • IP samples were centrifuged and eluted using cell lysis buffer 4 times, 10 minutes each time. Add 50 ⁇ l of 2x loading buffer to the eluted IP sample. Boil IP and lysate samples at 100°C for 10 minutes. SDS PAGE gel running test.
  • the results are shown in Figure 16.
  • the CREPT wild-type protein without candidate inhibitor treatment can be ubiquitinated by ubiquitin (Ub) (the third lane of Figure 16).
  • the negative control CREPT mutant SA control group cannot detect ubiquitin. Vegetarianization. In the bands to which candidate compounds #1 to #5 were added, ubiquitination of #4 was significantly attenuated (approximately 20% decrease in ubiquitination level relative to the untreated wild-type protein).
  • compound #4 is a potent inhibitor of CREPT phosphorylation and ubiquitination.
  • the structural formula of compound #4 is as follows:
  • DLD1 human colorectal adenocarcinoma epithelial cells
  • MGC803 human gastric cancer cells
  • compound #4 which is an inhibitor of CREPT phosphorylation and ubiquitination, significantly inhibited cell proliferation, which shows that the test concentration of compound #4 inhibited the degradation of CREPT to a certain extent, leading to apoptosis in some cells. This result is consistent with The results of Examples 2-4 are consistent.

Abstract

The present invention relates to a non-phosphorylated and non-ubiquitinated CREPT protein and use thereof. Specifically, the present invention relates to a protein that is obtained by modifying CREPT or a homologous protein thereof. The modification enables the modified protein to maintain a non-phosphorylation and non-ubiquitination state so as not to be degraded when the protein is located in eukaryotic cells at the end of the G1 phase or during the G1/S transition phase, so that a double-MCM hexamer cannot be separated, thereby stopping the cell cycle, generating a genome stress response, and finally causing cell death. The present invention also relates to a method for screening a non-phosphorylated and non-ubiquitinated modifier of CREPT, a method for identifying whether a substance is a phosphorylation inhibitor of CREPT, a method for identifying eukaryotic cells at the end of the G1 phase or during the G1/S transition phase by using CREPT, and a method for inducing cancer cell apoptosis to treat cancers on the basis of non-phosphorylated and non-ubiquitinated CREPT.

Description

非磷酸化及非泛素化的CREPT蛋白及其应用Non-phosphorylated and non-ubiquitinated CREPT proteins and their applications 技术领域Technical field
本发明涉及分子生物学领域,更具体而言,涉及一种能够保持非磷酸化及非泛素化状态的CREPT蛋白及其应用。The present invention relates to the field of molecular biology, and more specifically, to a CREPT protein that can maintain a non-phosphorylated and non-ubiquitinated state and its application.
背景技术Background technique
DNA在细胞周期的S期进行复制,错误的DNA复制会在生物体内的干细胞中积累,并与细胞死亡和个体衰老密切相关。因此,决定细胞命运的DNA复制需要受到严格的控制。真核细胞的DNA复制机制十分复杂,DNA复制的起始点位于细胞周期的G1期,pre-RC(pre-replication complex)蛋白加载在基因组所有潜在起始点上。首先,具有ATPase活性的ORC复合体(origin recognition complex,ORC1-6)被募集到复制起点。进一步的,CDC6、CDT1(CDC10-dependent transcript 1)和MCM2-7(mini-chromosome maintenance)的六聚体复合体加载到OCR复合体上形成MCM解旋酶复合体。这一步被称为复制起始许可(origin licensing)。复制起始的激活涉及pre-IC(pre-initiation complex)复合体的形成和MCM解旋酶复合体的激活。Pre-IC的组装由DDK(DBF4-dependent kinase)和CDK(Cyclin-dependent kinase)在G1/S转换时期触发。DDK和CDK使几种参与DNA复制的蛋白例如MCM10、CDC45、RECQL4(ATP-dependent DNA helicase Q4)、treslin、GINS和TOPBP1(DNAtopoisomerase 2-binding protein 1)磷酸化。此外,DDK和CDK也可以磷酸化MCM2-7复合体中的几个碱基,导致解旋酶激活并解旋DNA。在解旋酶激活期间,加载在DNA上的MCM2-7双六聚体分成两个单独的六聚体,在从复制起始点开始的两个复制叉上持续解旋。DNA is replicated in the S phase of the cell cycle. Errors in DNA replication will accumulate in stem cells in organisms and are closely related to cell death and individual aging. Therefore, DNA replication that determines cell fate needs to be tightly controlled. The DNA replication mechanism of eukaryotic cells is very complex. The starting point of DNA replication is located in the G1 phase of the cell cycle, and the pre-RC (pre-replication complex) protein is loaded on all potential starting points of the genome. First, the ORC complex (origin recognition complex, ORC1-6) with ATPase activity is recruited to the origin of replication. Further, the hexameric complex of CDC6, CDT1 (CDC10-dependent transcript 1) and MCM2-7 (mini-chromosome maintenance) is loaded onto the OCR complex to form the MCM helicase complex. This step is called origin licensing. Activation of replication initiation involves the formation of the pre-IC (pre-initiation complex) complex and the activation of the MCM helicase complex. The assembly of Pre-IC is triggered by DDK (DBF4-dependent kinase) and CDK (Cyclin-dependent kinase) during the G1/S transition period. DDK and CDK phosphorylate several proteins involved in DNA replication, such as MCM10, CDC45, RECQL4 (ATP-dependent DNA helicase Q4), treslin, GINS and TOPBP1 (DNAtopoisomerase 2-binding protein 1). In addition, DDK and CDK can also phosphorylate several bases in the MCM2-7 complex, causing helicase activation and unwinding of DNA. During helicase activation, the MCM2-7 double hexamer loaded on DNA separates into two separate hexamers that continue to unwind at both replication forks starting from the replication origin.
细胞周期受到泛素-蛋白酶体系统(UPS)介导的Cyclin相关蛋白和CDK抑制剂降解的精确调控。UPS通过将泛素添加到底物蛋白的赖氨酸(K)残基供蛋白酶体识别来实现高效的蛋白降解。有三种主要类型的酶控UPS:泛素激活酶(E1)、泛素结合酶酶(E2)和泛素连接酶(E3)。连接酶E3是最多样化的酶,其可提供针对靶标的特异性。在G1/S期,由SKP1、CUL1和F-box蛋白(FBP)组成的RING-finger亚家族E3连接 酶的SCF复合物识别待降解的底物。FBP主要决定SCF复合物的靶标特异性。SCF复合物更倾向于与磷酸化降解决定子(phosphodegron)结合,磷酸化降解决定子是产生供E3连接酶识别的表面的磷酸化底物基序。FBP家族成员SKP2(S期激酶相关蛋白2)是调节G1/S转换的E3连接酶之一。SKP2-CKS1-p27复合物介导CDK抑制剂p27的蛋白酶体降解并释放Cyclin E/CDK2以促进G1/S转换。The cell cycle is precisely regulated by the degradation of Cyclin-related proteins and CDK inhibitors mediated by the ubiquitin-proteasome system (UPS). UPS achieves efficient protein degradation by adding ubiquitin to lysine (K) residues of substrate proteins for recognition by the proteasome. There are three main types of enzyme-controlled UPS: ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3). Ligase E3 is the most diverse enzyme that provides specificity for targets. In the G1/S phase, the RING-finger subfamily E3 composed of SKP1, CUL1 and F-box protein (FBP) is connected The SCF complex of the enzyme recognizes the substrate to be degraded. FBP primarily determines the target specificity of the SCF complex. The SCF complex prefers to bind to phosphodegrons, which are phosphorylated substrate motifs that create a surface for recognition by E3 ligases. FBP family member SKP2 (S-phase kinase-related protein 2) is one of the E3 ligases that regulates G1/S transition. The SKP2-CKS1-p27 complex mediates proteasomal degradation of the CDK inhibitor p27 and releases Cyclin E/CDK2 to promote G1/S transition.
细胞周期的调控与肿瘤的发生发展有着紧密的关系,本实验室在前期研究中在人细胞中发现并克隆了一个与细胞周期调控和肿瘤形成相关的基因CREPT(Cell-cycle Related and Expression-elevated Protein in Tumor,中国专利号200510135513.4,其相应的蛋白序列见SEQ ID NO:4)。进而发现该基因在多种真核生物中存在十分保守的同源基因,例如在酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫和拟南芥的细胞中。已知CREPT蛋白通过调控Cyclin D1和B1的表达来促进细胞增殖。The regulation of cell cycle is closely related to the occurrence and development of tumors. In previous studies, our laboratory discovered and cloned a gene related to cell cycle regulation and tumor formation, CREPT (Cell-cycle Related and Expression-elevated), in human cells. Protein in Tumor, Chinese patent number 200510135513.4, its corresponding protein sequence is shown in SEQ ID NO: 4). It was further found that this gene has very conserved homologous genes in a variety of eukaryotes, such as in cells of yeast, mouse, dog, cat, chicken, toad, zebrafish, Drosophila, nematode and Arabidopsis. CREPT protein is known to promote cell proliferation by regulating the expression of Cyclin D1 and B1.
然而,CREPT具体如何调控细胞周期的机理尚不清楚。However, the specific mechanism of how CREPT regulates the cell cycle is unclear.
发明内容Contents of the invention
本发明人经大量研究发现,人细胞中的CREPT蛋白通过在G1/S转换期发生降解来推动细胞进入S1期,由此发挥细胞周期调控作用。The inventors of the present invention have discovered through extensive research that the CREPT protein in human cells promotes cells to enter the S1 phase through degradation during the G1/S transition phase, thus exerting a cell cycle regulatory effect.
具体而言,本发明人发现,在同一培养物中的多个细胞中,CREPT在部分细胞中表达量很低,而在其他细胞中保持较高表达,进而发现这种低表达现象是由于在G1/S转换期CREPT的降解所致,而CREPT的降解对于细胞由G1期进入S期至关重要。CREPT的降解由泛素化修饰来介导,而该泛素化修饰依赖于CREPT的两个位点S134和S166的磷酸化。发明人进一步发现,如果使CREPT蛋白的134和166位点在G1/S转换期保持非磷酸化状态且使该蛋白保持非泛素化状态,则CREPT蛋白不能被降解,这种不能被降解的CREPT蛋白不能与MCM复合体分离,由此会导致细胞死亡。Specifically, the inventors found that among multiple cells in the same culture, CREPT was expressed at a very low level in some cells, while maintaining high expression in other cells, and further found that this low expression phenomenon was due to It is caused by the degradation of CREPT during the G1/S transition period, and the degradation of CREPT is crucial for cells to enter the S phase from the G1 phase. The degradation of CREPT is mediated by ubiquitination modification, which depends on the phosphorylation of two sites of CREPT, S134 and S166. The inventor further discovered that if the 134 and 166 positions of the CREPT protein are kept in a non-phosphorylated state during the G1/S transition period and the protein is kept in a non-ubiquitinated state, the CREPT protein cannot be degraded. This kind of protein cannot be degraded. The CREPT protein cannot dissociate from the MCM complex, resulting in cell death.
此外,发明人还发现,非人真核细胞中的同源蛋白具有与人CREPT相同或相似的细胞周期调控作用和调控机理,而且人CREPT蛋白在表达于非人真核细胞中时,这种细胞周期调控作用依然存在。In addition, the inventor also found that the homologous protein in non-human eukaryotic cells has the same or similar cell cycle regulatory effect and regulatory mechanism as human CREPT, and when the human CREPT protein is expressed in non-human eukaryotic cells, this Cell cycle regulation remains.
基于上述发现,发明人完成了本发明。Based on the above findings, the inventor completed the present invention.
在第一方面,本发明提供一种通过对SEQ ID No:4的134和166位点进行磷酸化 失活修饰而得到的蛋白,所述磷酸化失活修饰使得当所述蛋白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述蛋白保持非泛素化状态,从而使所述蛋白在所述真核细胞中不被降解。In a first aspect, the present invention provides a method for phosphorylating SEQ ID No: 4 at positions 134 and 166. The protein obtained by the inactivation modification. The phosphorylation inactivation modification allows the 134 and 166 positions to remain unphosphorylated and remain unphosphorylated when the protein is located in eukaryotic cells at the end of G1 phase or the G1/S transition phase. The protein remains in a non-ubiquitinated state, thereby preventing the protein from being degraded in the eukaryotic cell.
在第一方面中,所述真核细胞可以是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。In a first aspect, the eukaryotic cell may be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell cell.
在第一方面中,所述134和166位点上的所述磷酸化失活修饰可以是氨基酸突变和/或化学修饰。In the first aspect, the phosphorylation inactivating modifications on the 134 and 166 positions may be amino acid mutations and/or chemical modifications.
在第一方面中,所述134和166位点上的所述磷酸化失活修饰可以是将丝氨酸(S)突变为丙氨酸(A)。In a first aspect, the phosphorylation-inactivating modification at the 134 and 166 positions may be a mutation of serine (S) to alanine (A).
在第一方面中,本发明还提供一种蛋白,其与上述每种蛋白具有(i)90%以上的序列同一性和(ii)相同的134和166位点上的磷酸化失活修饰。在第二方面,本发明提供一种蛋白,其在第一方面的蛋白的N端和/或C端连接有标签序列或引导序列。In a first aspect, the present invention also provides a protein that has (i) more than 90% sequence identity and (ii) the same phosphorylation inactivating modifications at positions 134 and 166 with each of the above-mentioned proteins. In a second aspect, the present invention provides a protein having a tag sequence or a guide sequence connected to the N-terminal and/or C-terminal of the protein of the first aspect.
在第三方面,本发明提供编码第一和第二方面所述的蛋白的核酸。In a third aspect, the invention provides nucleic acids encoding the proteins of the first and second aspects.
在第四方面,本发明提供包含第三方面所述的核酸的载体。In a fourth aspect, the present invention provides a vector comprising the nucleic acid of the third aspect.
在第五方面,本发明提供包含第四方面所述的载体的细胞。In a fifth aspect, the invention provides cells comprising the vector of the fourth aspect.
在第六方面,本发明提供第一至第五方面所述的蛋白、核酸或载体在制备抑制真核细胞增殖、抑制真核细胞的DNA复制、调控真核细胞的细胞周期或杀灭真核细胞的试剂中的应用。In the sixth aspect, the present invention provides the use of the proteins, nucleic acids or vectors described in the first to fifth aspects to inhibit eukaryotic cell proliferation, inhibit eukaryotic cell DNA replication, regulate the cell cycle of eukaryotic cells or kill eukaryotic cells. Cellular reagent applications.
在第七方面,本发明提供第一至第五方面所述的蛋白、核酸或载体在制备抗癌药中的应用。In a seventh aspect, the present invention provides the use of the protein, nucleic acid or vector described in the first to fifth aspects in the preparation of anti-cancer drugs.
在第八方面,本发明提供一种治疗癌症的方法,所述方法包括向受试者施用有效量的第一至第五方面所述的蛋白、核酸或载体;或者,所述方法包括利用基于CRISPR/Cas9的基因编辑技术编辑受试者癌细胞基因组中的CRPET基因,以使所述癌细胞表达SEQ ID NO:2的蛋白。其中,所述受试者可以是哺乳动物,优选是人。其中,在向受试者施用有效量的所述蛋白、核酸或载体之前、过程中或之后,可以降低或消除受试者的癌细胞中的野生型CREPT的表达。In an eighth aspect, the present invention provides a method for treating cancer, the method comprising administering to a subject an effective amount of the protein, nucleic acid or vector described in the first to fifth aspects; or, the method comprising using a method based on The gene editing technology of CRISPR/Cas9 edits the CRPET gene in the genome of the subject's cancer cells so that the cancer cells express the protein of SEQ ID NO:2. Wherein, the subject can be a mammal, preferably a human. Wherein, before, during or after administering an effective amount of the protein, nucleic acid or vector to the subject, the expression of wild-type CREPT in the cancer cells of the subject can be reduced or eliminated.
在第八方面,本发明提供一种筛选CREPT蛋白的非磷酸化非泛素化修饰剂的方法,其中,所述修饰剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白 的氨基酸序列为SEQ ID No:4,所述方法包括:In an eighth aspect, the present invention provides a method for screening non-phosphorylated non-ubiquitinated modifiers of CREPT protein, wherein the modifier maintains the S134 and S166 sites of CREPT protein in a continuous non-phosphorylated state, thereby allowing The CREPT protein remains in a non-ubiquitinated state in cells and will not be degraded; the CREPT protein The amino acid sequence is SEQ ID No: 4, and the method includes:
i)将模拟非磷酸化状态的候选修饰剂加入表达CREPT蛋白的被同步至G1期的真核细胞中,然后释放并培养所述真核细胞,并在所述真核细胞存活时检查所述CREPT蛋白的S134和S166位点的磷酸化水平;i) Add candidate modifiers that mimic the non-phosphorylated state to eukaryotic cells expressing CREPT protein that are synchronized to G1 phase, then release and culture the eukaryotic cells, and examine the eukaryotic cells while they are alive Phosphorylation levels of S134 and S166 of CREPT protein;
或者or
ii)在体外将模拟非磷酸化状态的候选修饰剂与CREPT蛋白温育,并在Cyclin E/CDK2激酶的催化条件下检查所述CREPT蛋白的S134和S166位点的磷酸化水平,ii) incubate the candidate modifier that simulates the non-phosphorylated state with the CREPT protein in vitro, and check the phosphorylation levels of the S134 and S166 sites of the CREPT protein under the catalytic conditions of Cyclin E/CDK2 kinase,
如果经步骤i)或ii)处理的CREPT蛋白的S134和S166位点的磷酸化水平相对于未经处理的对照下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述候选修饰剂筛选为CREPT蛋白的非磷酸化非泛素化修饰剂。If the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The candidate modifiers are screened as non-phosphorylated, non-ubiquitinated modifiers of the CREPT protein.
本发明还提供一种鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列为SEQ ID No:4,所述方法包括:The present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby causing the CREPT protein to function in cells. It maintains a non-ubiquitinated state and will not be degraded; the amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes:
i)将待鉴定的物质加入表达CREPT蛋白的被同步至G1期的真核细胞中,然后释放并培养所述真核细胞,并在所述真核细胞存活时检查所述CREPT蛋白的S134和S166位点的磷酸化水平;或者i) Add the substance to be identified to eukaryotic cells expressing CREPT protein that are synchronized to the G1 phase, then release and culture the eukaryotic cells, and examine the S134 and S134 of the CREPT protein while the eukaryotic cells are alive. Phosphorylation level at S166; or
ii)在体外将待鉴定的物质与CREPT蛋白温育,并在Cyclin E/CDK2激酶的催化条件下检查所述CREPT蛋白的S134和S166位点的磷酸化水平;ii) Incubate the substance to be identified with the CREPT protein in vitro, and check the phosphorylation levels of the S134 and S166 sites of the CREPT protein under the catalytic conditions of Cyclin E/CDK2 kinase;
如果经步骤i)或ii)处理的CREPT蛋白的S134和S166位点的磷酸化水平相对于未经处理的对照下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述物质鉴定为CREPT蛋白的磷酸化抑制剂,否则所述物质不是CREPT蛋白的磷酸化抑制剂。If the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The substance is identified as a phosphorylation inhibitor of CREPT protein, which is otherwise not a phosphorylation inhibitor of CREPT protein.
在上述方法中,可以采用质谱法或免疫沉淀法来检查所述CREPT蛋白的S134和S166位点的磷酸化水平;优选地,所述免疫沉淀法可以包括:使用识别CREPT蛋白的S134和S166位点的磷酸化的抗磷酸化抗体来进行免疫沉淀。In the above method, mass spectrometry or immunoprecipitation can be used to check the phosphorylation levels of S134 and S166 of the CREPT protein; preferably, the immunoprecipitation method can include: using the method to recognize the S134 and S166 of the CREPT protein. Spot phosphorylated anti-phosphorylated antibodies for immunoprecipitation.
本发明还提供一种鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列 为SEQ ID No:4,所述方法包括:i)将待鉴定的物质加入表达CREPT蛋白的真核细胞中并培养所述真核细胞,和ii)使用免疫沉淀法检查所述真核细胞的CREPT蛋白的泛素化水平;与未经所述物质处理的对照细胞中CREPT蛋白的泛素化水平相比,如果经处理的细胞中的CREPT蛋白的泛素化水平下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述物质鉴定为CREPT蛋白的磷酸化抑制剂,否则所述物质不是CREPT蛋白的磷酸化抑制剂。在步骤i)之前,所述方法还可以包括:使用预测工具SwissTargetPrediction和SEA针对CREPT来设计所述待鉴定的物质。此外,步骤ii)可以包括用识别CREPT蛋白的抗CREPT抗体和识别泛素的泛素抗体对CREPT蛋白的泛素化水平进行定量。The present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby causing the CREPT protein to function in cells. Maintains a non-ubiquitinated state and will not be degraded; the amino acid sequence of the CREPT protein Is SEQ ID No: 4, the method includes: i) adding the substance to be identified to eukaryotic cells expressing CREPT protein and culturing the eukaryotic cells, and ii) using an immunoprecipitation method to check the eukaryotic cells The ubiquitination level of CREPT protein; compared with the ubiquitination level of CREPT protein in control cells that have not been treated with the substance, if the ubiquitination level of CREPT protein in the treated cells decreases, for example, decreases by more than 10% , more than 20%, more than 30% or more than 40%, then the substance is identified as a phosphorylation inhibitor of CREPT protein, otherwise the substance is not a phosphorylation inhibitor of CREPT protein. Before step i), the method may further include: using prediction tools SwissTargetPrediction and SEA to design the substance to be identified for CREPT. Additionally, step ii) may include quantifying the ubiquitination level of the CREPT protein using an anti-CREPT antibody that recognizes the CREPT protein and a ubiquitin antibody that recognizes ubiquitin.
在第九方面,本发明提供源自非人真核生物的人CREPT的同源蛋白,其在对应于SEQ ID No:4的134和166位点的同源性位点处具有磷酸化失活修饰,所述磷酸化失活修饰使得当所述同源蛋白位于所述真核生物的G1末期或G1/S转换期细胞中时,所述对应于SEQ ID No:4的134和166位点的同源性位点保持非磷酸化状态且所述同源蛋白保持非泛素化状态,从而使所述同源蛋白在所述细胞中不会被降解,进而导致细胞周期阻滞和细胞凋亡。In a ninth aspect, the invention provides homologous proteins of human CREPT derived from non-human eukaryotes, which have phosphorylation inactivation at the homology sites corresponding to positions 134 and 166 of SEQ ID No: 4 Modification, the phosphorylation inactivation modification makes the 134 and 166 positions corresponding to SEQ ID No: 4 when the homologous protein is located in the G1 end phase or G1/S transition phase cells of the eukaryotic organism. The homology site remains in a non-phosphorylated state and the homologous protein remains in a non-ubiquitinated state, so that the homologous protein will not be degraded in the cell, leading to cell cycle arrest and apoptosis. Death.
在第九方面中,所述真核生物可以是酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥。In a ninth aspect, the eukaryotic organism may be yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana.
在第九方面中,所述对应于SEQ ID No:4的134和166位点的同源性位点上的所述磷酸化失活修饰可以是氨基酸突变和/或化学修饰。In the ninth aspect, the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 may be amino acid mutation and/or chemical modification.
在第九方面中,所述对应于SEQ ID No:4的134和166位点的同源性位点上的所述磷酸化失活修饰可以是将丝氨酸突变为丙氨酸。In the ninth aspect, the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 may be to mutate serine to alanine.
在第九方面中,所述同源蛋白的氨基酸序列可以为SEQ ID No:6。In the ninth aspect, the amino acid sequence of the homologous protein may be SEQ ID No: 6.
在第十方面,本发明提供一种蛋白,其选自:In a tenth aspect, the invention provides a protein selected from:
i)在第九方面所述的蛋白的N端和/或C端连接有标签序列或引导序列的蛋白;或i) A protein with a tag sequence or guide sequence connected to the N-terminus and/or C-terminus of the protein described in the ninth aspect; or
ii)与第九方面所述的蛋白具有90%以上的序列同一性且在所述同源性位点上具有相同的修饰的蛋白。ii) A protein that has more than 90% sequence identity with the protein described in the ninth aspect and has the same modification at the homology site.
在第十一方面,本发明提供编码第九和第十方面所述的蛋白的核酸。In an eleventh aspect, the invention provides nucleic acids encoding proteins of the ninth and tenth aspects.
在第十二方面,本发明提供包含第十一方面所述的核酸的载体。 In a twelfth aspect, the present invention provides a vector comprising the nucleic acid according to the eleventh aspect.
在第十三方面,本发明提供包含第十二方面所述的载体的细胞。In a thirteenth aspect, the present invention provides cells comprising the vector of the twelfth aspect.
在第十四方面,本发明提供一种鉴别处于G1末期或G1/S转换期的真核细胞的方法,所述方法包括:In a fourteenth aspect, the present invention provides a method for identifying eukaryotic cells in the late G1 phase or the G1/S transition phase, the method comprising:
1)制备能够内源性表达带可检测标记的人CREPT蛋白或其在非人真核生物中的同源蛋白的真核细胞,并在允许细胞周期进行的条件下培养该真核细胞;1) Prepare eukaryotic cells capable of endogenously expressing a detectably labeled human CREPT protein or its homologous protein in non-human eukaryotes, and culture the eukaryotic cells under conditions that allow the cell cycle to proceed;
2)利用所述可检测标记观察或测量所述真核细胞中的所述人CREPT蛋白或其同源蛋白的表达水平;2) using the detectable marker to observe or measure the expression level of the human CREPT protein or its homologous protein in the eukaryotic cell;
3)将所述人CREPT蛋白或其同源蛋白的表达水平最低的细胞鉴别为处于G1末期或G1/S转换期的细胞;3) Identify cells with the lowest expression level of the human CREPT protein or its homologous protein as cells in the late G1 phase or G1/S transition phase;
其中,所述人CREPT蛋白的序列为SEQ ID No:4。Wherein, the sequence of the human CREPT protein is SEQ ID No: 4.
在第十四方面中,所述可检测标记可以是同位素标记、荧光标记或量子点标记或者是能够进一步与同位素标记、荧光标记或量子点标记结合的标记,优选为GFP。In the fourteenth aspect, the detectable label may be an isotope label, a fluorescent label or a quantum dot label or a label that can be further combined with an isotope label, a fluorescent label or a quantum dot label, preferably GFP.
在第十四方面中,所述真核细胞可以是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞。In a fourteenth aspect, the eukaryotic cell may be a human, yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell.
在第十五方面,本发明提供一种抑制CREPT蛋白在真核细胞中的降解的方法,所述方法包括:In a fifteenth aspect, the present invention provides a method for inhibiting the degradation of CREPT protein in eukaryotic cells, the method comprising:
1)使选自SKP2抑制剂、CUL1抑制剂、neddylation抑制剂和CDK2抑制剂的抑制剂进入表达CREPT的真核细胞中;和/或1) Allowing an inhibitor selected from the group consisting of SKP2 inhibitors, CUL1 inhibitors, neddylation inhibitors and CDK2 inhibitors to enter eukaryotic cells expressing CREPT; and/or
2)对CREPT蛋白的第134和166位点进行磷酸化失活修饰,使得当经修饰的蛋白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述经修饰的蛋白保持非泛素化状态,从而使所述经修饰的蛋白不被降解。2) Phosphorylation inactivation modification is performed on the 134th and 166th positions of the CREPT protein, so that when the modified protein is located in eukaryotic cells at the end of G1 phase or the G1/S transition phase, the 134th and 166th positions remain inactive. phosphorylation state and the modified protein remains in a non-ubiquitinated state, thereby preventing the modified protein from being degraded.
在该方法中,所述真核细胞可以是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。In this method, the eukaryotic cell can be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell .
在该方法中,所述134和166位点上的所述磷酸化失活修饰可以是氨基酸突变和/或化学修饰,优选地,所述134和166位点上的所述磷酸化失活修饰是将丝氨酸(S)突变为丙氨酸(A)。In this method, the phosphorylation inactivating modifications at the 134 and 166 positions may be amino acid mutations and/or chemical modifications. Preferably, the phosphorylation inactivating modifications at the 134 and 166 positions are Serine (S) is mutated into alanine (A).
在该方法中,所述SKP2抑制剂可以是针对SKP2的双链siRNA,其序列为AAUCUAAGCCUGGAAGGCCUGdTdT;所述CUL1抑制剂可以是针对CUL1的双链siRNA,其序列为UAGACAUUGGGUUCGCCGUdTdT;所述neddylation抑制剂可以 是MLN4924。In this method, the SKP2 inhibitor can be a double-stranded siRNA against SKP2, the sequence of which is AAUCUAAGCCUGGAAGGCCUGdTdT; the CUL1 inhibitor can be a double-stranded siRNA against CUL1, and the sequence of the neddylation inhibitor can be UAGACAUUGGGUUCGCCGUdTdT; It's MLN4924.
在第十六方面,本发明还提供一种蛋白,其选自:1)通过将SEQ ID No:4的166位丝氨酸突变为丙氨酸而得到的蛋白;2)通过将SEQ ID No:8的136位丝氨酸突变为丙氨酸而得到的蛋白;3)通过将SEQ ID No:8的174位丝氨酸突变为丙氨酸而得到的蛋白;和4)与1)至3)中的任一种蛋白具有90%以上的序列同一性且具有相同的丙氨酸突变的蛋白。本发明还提供编码所述蛋白的核酸和包含所述核酸的载体。In a sixteenth aspect, the present invention also provides a protein selected from: 1) a protein obtained by mutating serine 166 of SEQ ID No: 4 to alanine; 2) a protein obtained by mutating SEQ ID No: 8 A protein obtained by mutating serine 136 of SEQ ID No: 8 to alanine; 3) A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; and 4) Any one of 1) to 3) A protein with more than 90% sequence identity and the same alanine mutation. The invention also provides nucleic acids encoding the proteins and vectors containing the nucleic acids.
附图说明Description of drawings
现将结合附图来描述本发明的具体实施方式,但附图和下文的具体实施方式都不应解读为对本发明的范围的限制,在附图中:The specific embodiments of the present invention will now be described with reference to the accompanying drawings, but neither the accompanying drawings nor the following detailed description should be construed as limiting the scope of the present invention. In the accompanying drawings:
图1A-图1E显示了CREPT的表达水平在细胞周期中振荡;(图1A)肿瘤细胞中CREPT的代表性荧光图像。白色圆圈表示没有CREPT表达的细胞。比例尺,10μm。(图1B)敲入了GFP-CREPT的细胞的延时显微镜活细胞图像。比例尺,10μm。(图1C)细胞周期中的CREPT表达;用2mM双胸苷阻滞(DTB)将DLD1细胞同步至G1/S期并释放。在所示的时间点收集并通过免疫印迹分析细胞裂解物。检测Cyclin A/B1/E、SKP2以确认细胞周期进展。CREPT蛋白水平在G1/S转换期达到最低。(图1D)DTB同步的DLD1Fucci细胞中CREPT的代表性荧光图像。比例尺,10μm。(图1E)荧光图像(图1D)中的CREPT、EdU和CDT1表达水平的量化结果。统计学显著性(*P<0.05;**P<0.01;***P<0.001,****P<0.0001);P>0.05,不显著[n.s.],由t检验产生。Figures 1A-1E show that the expression levels of CREPT oscillate during the cell cycle; (Figure 1A) Representative fluorescence images of CREPT in tumor cells. White circles indicate cells without CREPT expression. Scale bar, 10 μm. (Figure 1B) Time-lapse microscopy live cell image of cells with GFP-CREPT knock-in. Scale bar, 10 μm. (Figure 1C) CREPT expression during the cell cycle; DLD1 cells were synchronized to G1/S phase and released with 2mM double thymidine block (DTB). Cell lysates were collected at the indicated time points and analyzed by immunoblotting. Detect Cyclin A/B1/E, SKP2 to confirm cell cycle progression. CREPT protein levels reach their lowest levels during the G1/S transition period. (Figure 1D) Representative fluorescence image of CREPT in DTB-synchronized DLD1 Fucci cells. Scale bar, 10 μm. (Fig. 1E) Quantification of CREPT, EdU and CDT1 expression levels in fluorescence images (Fig. 1D). Statistical significance (*P<0.05; **P<0.01; ***P<0.001, ****P<0.0001); P>0.05, not significant [n.s.], generated by t test.
图2A-图2K显示出CREPT在G1/S转换期的降解依赖于泛素修饰;(图2A)放线菌酮(CHX)处理的细胞中的CREPT表达。CREPT降解从CHX处理后10小时开始。(图2B)MG132处理后CREPT表达水平保持稳定。在第二次胸苷释放之前,用或不用25μg/ml MG132处理DLD1细胞4小时。(图2C)图2B中CREPT的定量免疫印迹结果。(图2D)CREPT可以被泛素化。在转染有指定质粒的293T细胞中进行体内泛素化测定。(图2E)K11-聚泛素化介导的CREPT降解。在转染有指定构建体的293T细胞中进行体内泛素化测定。(图2F)K11型泛素化加速了CREPT降解。HeLa细胞用HA-Ub-K11质粒转染并用CHX处理。(图2G)图2F中的CREPT免疫印迹定量结果。(图2H)K48型泛素化对CREPT降解没有影响。HeLa细胞用CHX处理并转染了HA-Ub-K48质粒。(图2I)图2H中的CREPT免疫印迹定量结果。(图2J)K63型泛素 化对CREPT降解没有影响。HeLa细胞用CHX处理并转染了HA-Ub-K63质粒。(图2K)图2J中的CREPT免疫印迹定量结果。通过t检验计算统计学显著性(*P<0.05;**P<0.01;****P<0.0001)。Figures 2A-2K show that the degradation of CREPT during the G1/S transition phase is dependent on ubiquitin modification; (Figure 2A) CREPT expression in cycloheximide (CHX)-treated cells. CREPT degradation begins 10 hours after CHX treatment. (Figure 2B) CREPT expression levels remained stable after MG132 treatment. DLD1 cells were treated with or without 25 μg/ml MG132 for 4 h before the second thymidine release. (Fig. 2C) Quantitative immunoblotting results of CREPT in Fig. 2B. (Figure 2D) CREPT can be ubiquitinated. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids. (Figure 2E) K11-polyubiquitination-mediated degradation of CREPT. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated constructs. (Figure 2F) K11-type ubiquitination accelerates CREPT degradation. HeLa cells were transfected with HA-Ub-K11 plasmid and treated with CHX. (Figure 2G) CREPT immunoblot quantification results in Figure 2F. (Figure 2H) K48-type ubiquitination has no effect on CREPT degradation. HeLa cells were treated with CHX and transfected with HA-Ub-K48 plasmid. (Figure 2I) CREPT immunoblot quantification results in Figure 2H. (Figure 2J) K63 type ubiquitin Chemicalization has no effect on CREPT degradation. HeLa cells were treated with CHX and transfected with HA-Ub-K63 plasmid. (Figure 2K) CREPT immunoblot quantification results in Figure 2J. Statistical significance was calculated by t test (*P<0.05;**P<0.01;****P<0.0001).
图3A-图3M显示出CRL1SKP2在G1/S转换期直接将CREPT泛素化;(图3A)SKP2与CREPT的结合亲和力最高。通过质谱分析了同步细胞中的CREPT下拉蛋白。这些蛋白根据独特的肽和log10覆盖率来排名。显示了具有已知E3连接酶活性的前10位蛋白。圆点的面积代表相对覆盖率值。(图3B)外源CREPT和外源SKP2之间的相互作用。用抗HA珠子免疫沉淀来自表达HA-CREPT和Flag-SKP2的HEK293T细胞的细胞提取物,并用所示抗体进行免疫印迹分析。(图3C)内源CREPT和内源SKP2之间的相互作用。用抗CREPT珠子免疫沉淀DLD1细胞的细胞提取物,并用所示抗体进行免疫印迹分析。(图3D)纯化的原核生物表达SKP2和CREPT用于体外co-IP测定,然后进行免疫印迹分析。(图3E)SKP2介导CREPT的泛素化。在转染了所示质粒的293T细胞中进行体内泛素化测定,细胞用或不用MG132处理。(图3F)和(图3G)SKP2的过表达对CREPT降解的影响。(图3H)和(图3I)SKP2的敲低对CREPT降解的影响。(图3J)和(图3K)将细胞同步至G1/S期并释放后SKP2和CREPT的水平。(图3L)和(图3M)将细胞同步至M期并释放后SKP2和CREPT的水平。Figure 3A-Figure 3M show that CRL1 SKP2 directly ubiquitinates CREPT during the G1/S transition phase; (Figure 3A) SKP2 has the highest binding affinity to CREPT. CREPT pull-down proteins in synchronized cells were analyzed by mass spectrometry. The proteins are ranked based on unique peptide and log10 coverage. The top 10 proteins with known E3 ligase activity are shown. The area of the dots represents the relative coverage value. (Figure 3B) Interaction between exogenous CREPT and exogenous SKP2. Cell extracts from HEK293T cells expressing HA-CREPT and Flag-SKP2 were immunoprecipitated with anti-HA beads and subjected to immunoblot analysis with the indicated antibodies. (Figure 3C) Interaction between endogenous CREPT and endogenous SKP2. Cell extracts from DLD1 cells were immunoprecipitated with anti-CREPT beads and subjected to immunoblot analysis with the indicated antibodies. (Figure 3D) Purified prokaryotes expressing SKP2 and CREPT were used in in vitro co-IP assays followed by immunoblot analysis. (Figure 3E) SKP2 mediates ubiquitination of CREPT. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids, with or without treatment of MG132. (Fig. 3F) and (Fig. 3G) Effect of overexpression of SKP2 on CREPT degradation. (Figure 3H) and (Figure 3I) Effect of knockdown of SKP2 on CREPT degradation. (Fig. 3J) and (Fig. 3K) SKP2 and CREPT levels after synchronization and release of cells into G1/S phase. (Fig. 3L) and (Fig. 3M) SKP2 and CREPT levels after synchronizing cells to M phase and releasing them.
图4A-图4G显示出CREPT的S134A和S166A突变不能与Ub相互作用;(图4A)CREPT在CID结构域被泛素化。在转染了指定质粒并用MG132处理的293T细胞中进行体内泛素化测定。(图4B)通过质谱分析鉴定了CREPT修饰。在同步至G1/S期的293T细胞中进行改良质谱分析,CREPT蛋白上丝氨酸(S)位点的磷酸化水平根据#PSMs排序。(图4C)CREPT 135-170区域中CREPT和RTT103的氨基酸示意图。(图4D)CREPT S134A/S166A突变体不能被泛素化。对293T细胞中过表达的泛素化CREPT和CREPT突变体的免疫测定。(图4E)SKP2识别CREPT的磷酸化形式。用指定的质粒转染293T细胞并收获用于co-IP测定,然后进行免疫印迹分析。(图4F)内源CREPT和内源CDK2/Cyclin E之间的相互作用。用抗CREPT珠子免疫沉淀DLD1细胞的细胞提取物,并用所示抗体进行免疫印迹分析。(图4G)CREPT在S134/S166被CDK2/Cyclin E1磷酸化。从转染了Flag-CDK2和Flag-Cyclin E1的表达载体的HEK293T细胞中用抗Flag抗体通过Co-IP纯化出Flag-CDK2和Flag-Cyclin E1。GST、GST-CREPT和GST-CREPT(S134A/S166A)用GST珠从原核生物表达系统中纯化。 GST或GST标记的CREPT及其突变体的磷酸化通过通用抗磷酸化抗体来检测(顶部)。CREPT(SA):CREPT(S134A/S166A)。Figures 4A-4G show that the S134A and S166A mutations of CREPT are unable to interact with Ub; (Figure 4A) CREPT is ubiquitinated in the CID domain. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids and treated with MG132. (Figure 4B) CREPT modification was identified by mass spectrometry analysis. Modified mass spectrometry analysis was performed in 293T cells synchronized to G1/S phase, and the phosphorylation level of the serine (S) site on the CREPT protein was ranked according to #PSMs. (Figure 4C) Amino acid schematic diagram of CREPT and RTT103 in the CREPT 135-170 region. (Fig. 4D) The CREPT S134A/S166A mutant cannot be ubiquitinated. Immunoassay of ubiquitinated CREPT and CREPT mutants overexpressed in 293T cells. (Figure 4E) SKP2 recognizes the phosphorylated form of CREPT. 293T cells were transfected with the indicated plasmids and harvested for co-IP assays followed by immunoblot analysis. (Figure 4F) Interaction between endogenous CREPT and endogenous CDK2/Cyclin E. Cell extracts from DLD1 cells were immunoprecipitated with anti-CREPT beads and subjected to immunoblot analysis with the indicated antibodies. (Figure 4G) CREPT is phosphorylated by CDK2/Cyclin E1 at S134/S166. Flag-CDK2 and Flag-Cyclin E1 were purified by Co-IP using anti-Flag antibodies from HEK293T cells transfected with the expression vectors of Flag-CDK2 and Flag-Cyclin E1. GST, GST-CREPT and GST-CREPT(S134A/S166A) were purified from prokaryotic expression systems using GST beads. Phosphorylation of GST or GST-tagged CREPT and its mutants was detected with a universal anti-phosphorylation antibody (top). CREPT(SA): CREPT(S134A/S166A).
图5A-图5G显示出CREPT(S134A/S166A)突变导致细胞凋亡;(图5A)CREPT(S134A/S166A)突变体导致细胞死亡。HeLa野生型(Mock)和CREPT敲除(KO)细胞用所示的质粒转染48小时,然后用Annexin V和PI染色以进行FACS分析。(图5B)图5A中的流式细胞数据的统计分析。(图5C)细胞死亡由细胞凋亡触发。HeLa野生型(Mock)和CREPT敲除(KO)细胞用所示的质粒转染48小时,然后进行蛋白质印迹。(图5D)细胞生长抑制是由CREPT(S134A/S166A)过表达引起的。在用所示的质粒转染HeLa野生型(Mock)和CREPT敲除(KO)细胞48小时后,通过CCK-8测定来确定细胞活力。(图5E)CREPT(S134A/S166A)抑制肿瘤生长。将1×106个过表达所示质粒的B16细胞注射到C57BL/6小鼠中(n=3)。在第10天处死小鼠并测量肿瘤大小。(图5F)CREPT(S134A/S166A)突变体对酿酒酵母是致命的。在By4741 RTT103-KO细胞中比较WT、RTT103KO(Rtt103Δ)和人CREPT WT或突变质粒对温度的生长敏感性。(图5G)Rtt103(S136A/S174A)突变体对酿酒酵母是致命的。在By4741RTT103-KO细胞中比较WT、Rtt103Δ和酵母RTT103WT或突变质粒对温度的生长敏感性。CREPT(SA)代表CREPT(S134A/S166A)突变体,CREPT(SE)代表CREPT(S134E/S166E)突变体。Figures 5A-5G show that CREPT (S134A/S166A) mutation causes cell apoptosis; (Figure 5A) CREPT (S134A/S166A) mutant causes cell death. HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 h and then stained with Annexin V and PI for FACS analysis. (Figure 5B) Statistical analysis of the flow cytometry data in Figure 5A. (Figure 5C) Cell death is triggered by apoptosis. HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 hours and then subjected to Western blotting. (Figure 5D) Cell growth inhibition was caused by CREPT(S134A/S166A) overexpression. Cell viability was determined by CCK-8 assay 48 hours after transfection of HeLa wild-type (Mock) and CREPT knockout (KO) cells with the indicated plasmids. (Figure 5E) CREPT(S134A/S166A) inhibits tumor growth. 1×10 6 B16 cells overexpressing the indicated plasmids were injected into C57BL/6 mice (n=3). Mice were sacrificed on day 10 and tumor size was measured. (Fig. 5F) CREPT(S134A/S166A) mutant is lethal to S. cerevisiae. The growth sensitivity of WT, RTT103KO (Rtt103Δ) and human CREPT WT or mutant plasmids to temperature was compared in By4741 RTT103-KO cells. (Fig. 5G) Rtt103(S136A/S174A) mutant is lethal to S. cerevisiae. The growth sensitivity of WT, Rtt103Δ and yeast RTT103WT or mutant plasmids to temperature was compared in By4741RTT103-KO cells. CREPT(SA) represents CREPT(S134A/S166A) mutant, and CREPT(SE) represents CREPT(S134E/S166E) mutant.
图6A-图6G显示出未降解的突变CREPT阻止细胞进入S期;(图6A)CREPT在G1/S转换期未与MCM复合物结合。在DLD1细胞中进行了ChIP-MS分析,根据#PSMs对蛋白进行排序。细胞周期用DTB同步。(图6B)外源CREPT和外源MCM5之间的相互作用。用抗Myc珠子免疫沉淀来自表达Myc-CREPT和Flag-MCM5的HEK293T细胞的细胞提取物,并用指定的抗体进行免疫印迹分析。(图6C)内源CREPT和内源MCM5之间的相互作用。DLD1细胞的细胞提取物与1%多聚甲醛交联10分钟。然后,用抗CREPT珠子进行免疫沉淀,并用指定的抗体进行免疫印迹分析。(图6D)细胞周期中CREPT和MCM5之间的相互作用。DLD1细胞同步至G2/M期。在所示时间收集细胞裂解液用于内源性co-IP测定。(图6E)CREPT(S134A/S166A)突变体下拉了更多的MCM蛋白。用指定的质粒转染293T细胞并收获用于co-IP。(图6F)-(图6G)未降解的CREPT突变体阻止细胞进入S期。HeLa细胞中MCM5和EdU的代表性荧光图像,细胞过表达CREPT(S134A/S166A)突变体16小时,然后用预提 取方法染色。比例尺,10μm。CREPT(WT)代表CREPT野生型质粒,CREPT(SA)代表CREPT(S134A/S166A)突变体,CREPT(SE)代表CREPT(S134E/S166E)突变体。Figures 6A-6G show that non-degraded mutant CREPT prevents cells from entering S phase; (Figure 6A) CREPT does not bind to the MCM complex during the G1/S transition phase. ChIP-MS analysis was performed in DLD1 cells to rank proteins according to #PSMs. Cell cycle synchronization with DTB. (Figure 6B) Interaction between exogenous CREPT and exogenous MCM5. Cell extracts from HEK293T cells expressing Myc-CREPT and Flag-MCM5 were immunoprecipitated with anti-Myc beads and subjected to immunoblot analysis with the indicated antibodies. (Figure 6C) Interaction between endogenous CREPT and endogenous MCM5. Cell extracts of DLD1 cells were cross-linked with 1% paraformaldehyde for 10 min. Then, immunoprecipitation was performed with anti-CREPT beads and immunoblot analysis was performed with the indicated antibodies. (Figure 6D) Interaction between CREPT and MCM5 during the cell cycle. DLD1 cells are synchronized to G2/M phase. Cell lysates were collected at the indicated times for endogenous co-IP assay. (Figure 6E) The CREPT(S134A/S166A) mutant pulled down more MCM protein. 293T cells were transfected with the indicated plasmids and harvested for co-IP. (Figure 6F) - (Figure 6G) Undegraded CREPT mutants prevent cells from entering S phase. Representative fluorescence images of MCM5 and EdU in HeLa cells overexpressing CREPT(S134A/S166A) mutants for 16 hours and then treated with pre- Use method to dye. Scale bar, 10 μm. CREPT(WT) represents CREPT wild-type plasmid, CREPT(SA) represents CREPT(S134A/S166A) mutant, and CREPT(SE) represents CREPT(S134E/S166E) mutant.
图7A-图7D显示出未降解的CREPT使复制叉中止;(图7A)HeLa细胞中CREPT的代表性荧光图像。用指定的质粒转染细胞12小时。免疫荧光测定RPA2定量结果(右图)。比例尺,10μm。(图7B)HeLa细胞中CREPT的代表性荧光图像。免疫荧光测定的p-RPA2定量结果(右图)。比例尺,10μm。(图7C)用HU处理的WT和S134A/S166A突变体中的DNA复制速率。首先用4mM HU处理不同步的HeLa CREPT KO细胞4小时。然后将CIdU添加到培养物中0.5小时,然后收获细胞用于DNA纤维分析,以测量CIdU纤维的长度和分布。比例尺,10μm。(图7D)在HU阻断和释放期间,WT和S134A/S166A突变体中中止的复制叉的百分比。在HeLa CREPT KO细胞中进行了DNA纤维测定以测量中止的复制叉。统计学显著性(*P<0.05;**P<0.01;****P<0.0001)通过t检验计算。CREPT(WT)代表CREPT野生型质粒,CREPT(SA)代表CREPT(S134A/S166A)突变体。Figures 7A-7D show that non-degraded CREPT halts replication forks; (Figure 7A) Representative fluorescence images of CREPT in HeLa cells. Cells were transfected with indicated plasmids for 12 h. Immunofluorescence assay RPA2 quantitative results (right picture). Scale bar, 10 μm. (Figure 7B) Representative fluorescence image of CREPT in HeLa cells. Quantitative results of p-RPA2 by immunofluorescence assay (right panel). Scale bar, 10 μm. (Fig. 7C) DNA replication rates in WT and S134A/S166A mutants treated with HU. Asynchronous HeLa CREPT KO cells were first treated with 4mM HU for 4 hours. CIdU was then added to the culture for 0.5 h, and cells were harvested for DNA fiber analysis to measure the length and distribution of CIdU fibers. Scale bar, 10 μm. (Fig. 7D) Percentage of aborted replication forks in WT and S134A/S166A mutants during HU block and release. DNA fiber assay was performed in HeLa CREPT KO cells to measure aborted replication forks. Statistical significance (*P<0.05; **P<0.01; ****P<0.0001) was calculated by t-test. CREPT(WT) represents CREPT wild-type plasmid, and CREPT(SA) represents CREPT(S134A/S166A) mutant.
图8显示了CREPT调控细胞周期的作用机制的示意图。Figure 8 shows a schematic diagram of the mechanism of action of CREPT in regulating the cell cycle.
图9A-图9G显示了CREPT的表达水平在细胞周期中振荡,其中,(图9A)GFP-CREPT融合蛋白敲入Hela细胞系的示意图。GFP融合在CREPT的EXON1中的ATG序列之后。(图9B)通过蛋白质印迹验证了GFP-CREPT表达敲入了细胞中。(图9C)在所示的释放时间点通过PI染色对同步的DLD1细胞进行FACS分析。(图9D)细胞周期中的CREPT表达。HeLa细胞通过DTB同步至G1/S期并释放。(图9E)在DTB处理后的所示释放时间点对CREPT mRNA表达进行实时定量PCR分析。(图9F)DTB同步的DLD1Fucci细胞中CREPT的代表性荧光图像。比例尺10μm。(图9G)荧光图像(图9F)中CREPT和CDT1表达水平的量化结果。统计学显著(****P<0.0001);P>0.05,不显著[n.s.]),通过t检验计算。Figures 9A-9G show that the expression level of CREPT oscillates in the cell cycle, wherein (Figure 9A) Schematic diagram of GFP-CREPT fusion protein knock-in into Hela cell line. GFP is fused after the ATG sequence in EXON1 of CREPT. (Fig. 9B) Western blotting confirmed that GFP-CREPT expression was knocked into cells. (Figure 9C) FACS analysis of synchronized DLD1 cells by PI staining at the indicated release time points. (Fig. 9D) CREPT expression during the cell cycle. HeLa cells are synchronized to G1/S phase and released via DTB. (Figure 9E) Real-time quantitative PCR analysis of CREPT mRNA expression at the indicated release time points after DTB treatment. (Figure 9F) Representative fluorescence image of CREPT in DTB-synchronized DLD1 Fucci cells. Scale bar 10 μm. (Figure 9G) Quantification results of CREPT and CDT1 expression levels in fluorescence images (Figure 9F). Statistically significant (****P<0.0001); P>0.05, not significant [n.s.]), calculated by t test.
图10A-图10D显示出CREPT在G1/S转换期通过泛素途径降解;(图10A)图2A中CREPT的定量免疫印迹结果。(图10B)有或没有MG132处理时同步化的DLD1细胞的FACS分析。(图10C)CREPT降解不是由自噬介导的。在第二次胸苷释放前6小时用溶酶体抑制剂氯喹或亮肽素处理DLD1细胞。(图10D)在第二次胸苷释放前6小时,用溶酶体抑制剂氯喹或亮肽素处理HeLa细胞。Figures 10A-10D show that CREPT is degraded through the ubiquitin pathway during the G1/S transition phase; (Figure 10A) Quantitative immunoblotting results of CREPT in Figure 2A. (Figure 10B) FACS analysis of synchronized DLD1 cells with or without MG132 treatment. (Figure 10C) CREPT degradation is not mediated by autophagy. DLD1 cells were treated with the lysosomal inhibitors chloroquine or leupeptin 6 hours before the second thymidine release. (Figure 10D) HeLa cells were treated with the lysosomal inhibitors chloroquine or leupeptin 6 hours before the second thymidine release.
图11A-图11I显示出CRL1SKP2在G1/S转换期直接将CREPT泛素化。(图11A) 验证了GST-CREPT表达。对带有纯化的GST-CERPT蛋白的凝胶进行考马斯蓝染色。(图11B)验证了His-SKP1和His-SKP2的表达。对带有纯化的His-SKP1和His-SKP2蛋白的凝胶进行考马斯蓝染色。(图11C)验证了His-SKP2的表达。在不同纯化步骤中对纯化的SKP2蛋白进行免疫测定。(图11D)在所示的释放时间点通过PI染色对同步的DLD1细胞进行FACS分析。(图11E)cullin连接酶的激活对于CREPT积累至关重要。用1μM neddylation抑制剂MLN4924处理了0-8小时的DLD1细胞中的CREPT的免疫测定。其中si Ctrl为对照细胞,si SKP2为用针对SKP2的siRNA调低了SKP2的细胞(图11F)CUL1与CREPT相互作用。用所示的质粒转染293T细胞并收获用于免疫共沉淀(co-IP)测定,然后进行免疫印迹分析。(图11G)CUL1介导CREPT的泛素化。在用所示质粒转染的293T细胞中进行体内泛素化测定,细胞经或未经MG132处理。(图11H)过表达的CUL1加速CREPT降解。在所示的时间向Hela细胞添加CHX,用或不用MG132处理。(图11I)敲低的CUL1延长了CREPT半衰期。Hela细胞用CHX处理,通过siRNA敲低CUL1或N/C。Figures 11A-11I show that CRL1 SKP2 directly ubiquitinates CREPT during the G1/S transition. (Figure 11A) GST-CREPT expression was verified. Gels with purified GST-CERPT protein were stained with Coomassie blue. (Fig. 11B) The expression of His-SKP1 and His-SKP2 was verified. Gels with purified His-SKP1 and His-SKP2 proteins were stained with Coomassie blue. (Fig. 11C) The expression of His-SKP2 was verified. Immunoassays were performed on purified SKP2 protein at different purification steps. (Figure 1 ID) FACS analysis of synchronized DLD1 cells by PI staining at the indicated release time points. (Figure 11E) Activation of cullin ligase is critical for CREPT accumulation. Immunoassay of CREPT in DLD1 cells treated with 1 μM neddylation inhibitor MLN4924 for 0-8 h. Among them, si Ctrl is the control cell, and si SKP2 is the cell in which SKP2 was downregulated with siRNA against SKP2 (Figure 11F). CUL1 interacts with CREPT. 293T cells were transfected with the indicated plasmids and harvested for co-immunoprecipitation (co-IP) assays followed by immunoblot analysis. (Figure 11G) CUL1 mediates ubiquitination of CREPT. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids, with or without MG132 treatment. (Figure 11H) Overexpressed CUL1 accelerates CREPT degradation. CHX was added to HeLa cells with or without MG132 treatment at the times indicated. (Figure 11I) Knockdown of CUL1 prolonged CREPT half-life. HeLa cells were treated with CHX to knock down CUL1 or N/C via siRNA.
图12A-图12E显示出CREPT的S134A和S166A突变不能与Ub相互作用;(图12A)CREPT泛素化不依赖于CID结构域的单个赖氨酸(K)突变。在用所示质粒转染的293T细胞中进行体内泛素化测定。(图12B)CREPT泛素化不依赖于连接区的单个K突变。在用所示质粒转染的293T细胞中进行体内泛素化测定。(图12C)CREPT泛素化不依赖于K。在用所示质粒转染并收获用于co-IP测定的293T细胞中进行体内泛素化测定。(图12D)CREPT泛素化不依赖于苏氨酸(T)或半胱氨酸(C)。在用所示质粒转染的293T细胞中进行体内泛素化测定。(图12E)CREPT在真核细胞中被磷酸化。通过蛋白质印迹使用通用抗磷酸化抗体检查原核细胞(大肠杆菌)和真核细胞(哺乳动物)中的纯化GST-CREPT。p-CREPT(S/T/Y)代表泛磷酸化抗体。Figures 12A-12E show that the S134A and S166A mutations of CREPT are unable to interact with Ub; (Figure 12A) CREPT ubiquitination is independent of single lysine (K) mutations in the CID domain. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids. (Figure 12B) CREPT ubiquitination does not depend on a single K mutation in the linker region. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids. (Figure 12C) CREPT ubiquitination is independent of K. In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids and harvested for co-IP assays. (Figure 12D) CREPT ubiquitination is independent of threonine (T) or cysteine (C). In vivo ubiquitination assays were performed in 293T cells transfected with the indicated plasmids. (Figure 12E) CREPT is phosphorylated in eukaryotic cells. Purified GST-CREPT in prokaryotic cells (E. coli) and eukaryotic cells (mammalian) was examined by Western blotting using a universal anti-phospho-antibody. p-CREPT(S/T/Y) stands for pan-phosphorylated antibody.
图13A-图13F显示出CREPT(S134A/S166A)突变导致细胞凋亡;(图13A)由未降解的CREPT突变蛋白引起的细胞生长抑制和细胞死亡。HeLa野生型(Mock)和CREPT敲除(KO)细胞用所示质粒转染48小时。(图13B)CREPT(S134A/S166A)的过表达导致细胞生长抑制。在用所示质粒转染293T和NCM460细胞48小时后,通过CCK-8测定来确定细胞活力。(图13C)和(图13D)CREPT(S134A/S166A)突变导致克隆形成性降低。用所示质粒转染HeLa野生型和CREPT敲除细胞6小时,然后计数1000个细胞用于克隆形成测定。(图13E)CREPT(S134A/S166A)抑制肿瘤生长。将1×106个过表 达所示质粒的B16细胞注射到C57BL/6小鼠中(n=3)。在第10天处死小鼠并测量肿瘤大小。(图13F)比较By4741RTT103WT细胞中WT、RTT103KO(Rtt103Δ)和酵母RTT103WT或突变质粒对温度的生长敏感性。CREPT(SA):CREPT(S134A/S166A)突变;CREPT(SE):CREPT(S134E/S166E)突变。Figures 13A-13F show that CREPT (S134A/S166A) mutation leads to apoptosis; (Figure 13A) Cell growth inhibition and cell death caused by undegraded CREPT mutant protein. HeLa wild-type (Mock) and CREPT knockout (KO) cells were transfected with the indicated plasmids for 48 hours. (Figure 13B) Overexpression of CREPT (S134A/S166A) resulted in cell growth inhibition. Cell viability was determined by CCK-8 assay 48 hours after transfection of 293T and NCM460 cells with the indicated plasmids. (Fig. 13C) and (Fig. 13D) CREPT(S134A/S166A) mutations result in reduced clonogenicity. HeLa wild-type and CREPT knockout cells were transfected with the indicated plasmids for 6 h, and then 1000 cells were counted for clonogenic assay. (Figure 13E) CREPT(S134A/S166A) inhibits tumor growth. Pass 1×10 6 pieces across the table B16 cells harboring the indicated plasmids were injected into C57BL/6 mice (n=3). Mice were sacrificed on day 10 and tumor size was measured. (Figure 13F) Comparison of the growth sensitivity of WT, RTT103KO (Rtt103Δ) and yeast RTT103WT or mutant plasmids to temperature in By4741RTT103WT cells. CREPT(SA): CREPT(S134A/S166A) mutation; CREPT(SE): CREPT(S134E/S166E) mutation.
图14A-图14G显示出未降解的CREPT阻止细胞进入S期;(图14A)CREPT与MCM2相互作用。293T细胞用所示的质粒转染并收获用于co-IP。(图14B)CREPT与MCM7相互作用。293T细胞用所示的质粒转染。(图14C)在交联的情况下,CREPT与内源性MCM5结合。在DLD1细胞中的内源性co-IP测定,细胞用1%多聚甲醛固定。(图14D)CREPT不直接与染色质结合。HeLa细胞中MCM5和EdU的代表性荧光图像,细胞用直接固定和预提取方法染色。比例尺,10μm。(图14E)用所示质粒转染12小时的CREPT KO HeLa细胞的FACS分析。(图14F)图6G中免疫荧光测定的MCM5定量结果。(图14G)图6G中的细胞核直径。WT:CREPT(WT);SA:CREPT(S134A/S166A)。Figures 14A-14G show that undegraded CREPT prevents cells from entering S phase; (Figure 14A) CREPT interacts with MCM2. 293T cells were transfected with the indicated plasmids and harvested for co-IP. (Figure 14B) CREPT interacts with MCM7. 293T cells were transfected with the indicated plasmids. (Figure 14C) In the presence of cross-linking, CREPT binds to endogenous MCM5. Endogenous co-IP assay in DLD1 cells. Cells were fixed with 1% paraformaldehyde. (Figure 14D) CREPT does not bind directly to chromatin. Representative fluorescence images of MCM5 and EdU in HeLa cells stained with direct fixation and pre-extraction methods. Scale bar, 10 μm. (Figure 14E) FACS analysis of CREPT KO HeLa cells transfected with the indicated plasmids for 12 hours. (Fig. 14F) MCM5 quantification results of immunofluorescence assay in Fig. 6G. (Figure 14G) Nucleus diameter in Figure 6G. WT: CREPT(WT); SA: CREPT(S134A/S166A).
图15A-图15D显示出未降解的CREPT使复制叉中止;(图15A)HeLa细胞中TUNEL信号的代表性荧光图像。过表达所示质粒时对HeLa野生型(Mock)和CREPT敲除(KO)细胞进行Tunel染色,CREPT(SA):CREPT(S134A/S166A)突变,CREPT(SE):CREPT(S134E/S166E)突变。比例尺,10μm。(图15B)HeLa细胞中γH2AX的代表性荧光图像。过表达所示质粒12小时时的HeLa CREPT敲除细胞,CREPT(SA):CREPT(S134A/S166A)突变,CREPT(SE):CREPT(S134E/S166E)突变。比例尺,10μm。(图15C)免疫荧光染色的细胞的γH2AX intden定量值。(图15D)免疫荧光染色的细胞的γH2AX焦点数。统计学显著性(****P<0.0001)通过t检验计算。Figures 15A-15D show that non-degraded CREPT halts replication forks; (Figure 15A) Representative fluorescent images of TUNEL signal in HeLa cells. Tunel staining of HeLa wild-type (Mock) and CREPT knockout (KO) cells when overexpressing the indicated plasmids, CREPT (SA): CREPT (S134A/S166A) mutation, CREPT (SE): CREPT (S134E/S166E) mutation . Scale bar, 10 μm. (Figure 15B) Representative fluorescence image of γH2AX in HeLa cells. HeLa CREPT knockout cells at 12 hours overexpression of the indicated plasmids, CREPT(SA): CREPT(S134A/S166A) mutation, CREPT(SE): CREPT(S134E/S166E) mutation. Scale bar, 10 μm. (Figure 15C) Quantitative values of γH2AX in cells stained by immunofluorescence. (Fig. 15D) Number of γH2AX foci of immunofluorescence stained cells. Statistical significance (****P<0.0001) was calculated by t-test.
图16是CREPT磷酸化泛素化的小分子抑制剂的筛选结果。上方的#1至#5表示细胞经候选化合物#1至#5处理,中间的数字(0.1至1.1)是各样品的相对泛素化水平。SA:CREPT(S134A/S166A)。Figure 16 is the screening results of small molecule inhibitors of CREPT phosphorylation and ubiquitination. #1 to #5 above indicate cells treated with candidate compounds #1 to #5, and the numbers in the middle (0.1 to 1.1) are the relative ubiquitination levels of each sample. SA: CREPT(S134A/S166A).
图17显示了候选化合物#1至#5对DLD1细胞(A)和MGC803细胞(B)的增殖的影响。Figure 17 shows the effect of candidate compounds #1 to #5 on the proliferation of DLD1 cells (A) and MGC803 cells (B).
具体实施方式Detailed ways
定义 definition
本文中使用的术语“人CREPT蛋白”或“CREPT蛋白”在没有特别说明的情况下指人野生型CREPT蛋白。The term "human CREPT protein" or "CREPT protein" used herein refers to human wild-type CREPT protein unless otherwise specified.
本文中使用的术语“CREPT蛋白变体”、“经修饰的CREPT蛋白”是指在野生型CREPT蛋白的基础上进行了氨基酸突变和/或化学修饰而得到的蛋白变体。The terms "CREPT protein variant" and "modified CREPT protein" used herein refer to protein variants obtained by subjecting the wild-type CREPT protein to amino acid mutations and/or chemical modifications.
本文中使用的术语“同源蛋白”是指氨基酸序列具有同源性、在不同生物体中行使相同或相似功能的蛋白。The term "homologous protein" used herein refers to proteins with homologous amino acid sequences that perform the same or similar functions in different organisms.
本文中使用的术语“磷酸化失活修饰”是指蛋白中的被修饰的氨基酸残基在真核细胞中能够持续保持或模拟非磷酸化状态,经磷酸化失活修饰的残基不能被真核细胞中的激酶磷酸化。The term "phosphorylation-inactivating modification" used in this article means that the modified amino acid residues in the protein can continue to maintain or simulate the non-phosphorylated state in eukaryotic cells. The residues modified by phosphorylation-inactivating cannot be truly modified. Kinase phosphorylation in nuclear cells.
下文将详细说明及本发明的具体实施方式。The following will describe in detail the specific embodiments of the present invention.
发明人经过大量研究工作,发现了CREPT蛋白在调控细胞周期方面的作用机理,并由此完成了本发明。After extensive research work, the inventor discovered the mechanism of action of CREPT protein in regulating cell cycle, and thus completed the present invention.
首先,发明人发现,在细胞周期中CREPT蛋白的表达水平呈振荡状态(图1A),这是由于CREPT蛋白在G1/S转换期的降解所致,这种降解导致其与MCM复合体分离,由此驱动细胞进入S期并开始进行DNA复制。因此,CREPT蛋白在细胞的G1/S转换期处在最低水平,而随着细胞进入S期,CREPT蛋白的表达水平逐渐恢复(图1B和图1D)。First, the inventors found that the expression level of CREPT protein was in an oscillating state during the cell cycle (Figure 1A). This was due to the degradation of CREPT protein during the G1/S transition phase, which caused its separation from the MCM complex. This drives the cell into S phase and begins DNA replication. Therefore, CREPT protein is at its lowest level during the G1/S transition phase of cells, and as cells enter S phase, the expression level of CREPT protein gradually recovers (Figure 1B and Figure 1D).
因此,在本发明的一个方面,提供了一种鉴别处于G1末期或G1/S转换期的真核细胞的方法,所述方法包括:Therefore, in one aspect of the present invention, a method for identifying eukaryotic cells in the late G1 phase or the G1/S transition phase is provided, the method comprising:
1)制备能够内源性表达带可检测标记的人CREPT蛋白或其在非人真核生物中的同源蛋白的真核细胞,并在允许细胞周期进行的条件下培养该真核细胞;1) Prepare eukaryotic cells capable of endogenously expressing a detectably labeled human CREPT protein or its homologous protein in non-human eukaryotes, and culture the eukaryotic cells under conditions that allow the cell cycle to proceed;
2)利用所述可检测标记观察或测量所述真核细胞中的所述人CREPT蛋白或其同源蛋白的表达水平;2) using the detectable marker to observe or measure the expression level of the human CREPT protein or its homologous protein in the eukaryotic cell;
3)将所述人CREPT蛋白或其同源蛋白的表达水平最低的细胞鉴别为处于G1末期或G1/S转换期的细胞;3) Identify cells with the lowest expression level of the human CREPT protein or its homologous protein as cells in the late G1 phase or G1/S transition phase;
其中,所述人CREPT蛋白的序列为SEQ ID No:4。Wherein, the sequence of the human CREPT protein is SEQ ID No: 4.
在一个实施方式中,所述可检测标记是同位素标记、荧光标记或量子点标记或者是能够进一步与同位素标记、荧光标记或量子点标记结合的标记,优选为GFP。In one embodiment, the detectable label is an isotope label, a fluorescent label or a quantum dot label or a label that can be further combined with an isotope label, a fluorescent label or a quantum dot label, preferably GFP.
在一个实施方式中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马 鱼、果蝇、线虫或拟南芥的细胞,优选人细胞或酵母细胞。In one embodiment, the eukaryotic cell is human, yeast, mouse, canine, feline, chicken, toad, zebra Fish, Drosophila, nematode or Arabidopsis thaliana cells, preferably human cells or yeast cells.
发明人进一步发现,CREPT蛋白在细胞的G1/S转换期的这种降解依赖于泛素化介导的蛋白酶体降解,该泛素化由E3连接酶CRL1SKP2催化,而SKP2的识别和催化作用依赖于CREPT蛋白在S134和S166这两个位点的磷酸化。发明人通过对这两个位点进行突变模拟了CREPT的134和166位点的磷酸化和非磷酸化状态,结果发现,这两个位点保持非磷酸化的CREPT蛋白保持非泛素化状态,非磷酸化及非泛素化状态的S134A/S166A双突变蛋白在G1末期或G1/S转换期不会降解,这种不降解的CREPT蛋白变体不会与MCM复合体分离,从而导致细胞阻滞于MCM复合体表达量较高的G1末期或G1/S转换期,导致细胞周期无法进入S期,并停滞DNA复制叉的行进,最终导致细胞死亡(图5A-图5G)。单独的S134A突变或S166A突变蛋白能够在一定程度上影响泛素化(图4D)。另一方面,模拟磷酸化状态的S134E/S166E双突变蛋白对细胞的泛素化和生存均没有影响(图4D,图5A-图5B),说明它和野生型CREPT蛋白一样在G1/S转换期正常降解。以上证明,保持134和166位点非磷酸化且保持蛋白非泛素化的CREPT蛋白变体在G1末期或G1/S转换期不能发生降解,进而导致细胞死亡。The inventor further discovered that this degradation of CREPT protein during the G1/S transition phase of cells relies on ubiquitination-mediated proteasomal degradation. The ubiquitination is catalyzed by the E3 ligase CRL1 SKP2 , and the recognition and catalysis of SKP2 Depends on the phosphorylation of CREPT protein at S134 and S166. The inventor simulated the phosphorylation and non-phosphorylation states of CREPT at sites 134 and 166 by mutating these two sites, and found that the CREPT protein that remained unphosphorylated at these two sites remained in a non-ubiquitinated state. , the non-phosphorylated and non-ubiquitinated S134A/S166A double mutant protein will not be degraded at the end of G1 or the G1/S transition phase. This non-degraded CREPT protein variant will not be separated from the MCM complex, resulting in cell Blocking in the late G1 phase or the G1/S transition phase with high expression of the MCM complex results in the cell cycle being unable to enter the S phase and stalling the progression of DNA replication forks, ultimately leading to cell death (Figure 5A-Figure 5G). The S134A mutation alone or the S166A mutant protein can affect ubiquitination to a certain extent (Fig. 4D). On the other hand, the S134E/S166E double mutant protein that simulates the phosphorylation state has no effect on cell ubiquitination and survival (Figure 4D, Figure 5A-Figure 5B), indicating that it is the same as the wild-type CREPT protein at the G1/S transition. degrade normally. The above proves that CREPT protein variants that maintain non-phosphorylation at positions 134 and 166 and maintain non-ubiquitination of the protein cannot be degraded at the end of G1 phase or the G1/S transition phase, leading to cell death.
本领域已知负责蛋白泛素化的连接酶对底物的识别依赖于底物上特定位点(通常为丝氨酸、苏氨酸、酪氨酸)的磷酸化,而磷酸化的实质在于蛋白质的电荷发生了改变。因此,通过模拟相应位点的磷酸化状态(即,带负电荷),往往能够达到使底物蛋白模拟磷酸化的状态。在本案中,磷酸化的蛋白可以被泛素化连接酶识别,进而促使了蛋白的降解。磷酸化/非磷酸化的模拟通常采用突变或化学修饰来实现。例如,持续激活性突变(即模拟磷酸化状态的突变)包括将残基突变成天冬氨酸(D)或谷氨酸(E),因为这两个氨基酸是唯二的带负电荷的氨基酸;而持续抑制性突变(即模拟非磷酸化的突变)最常见的是将丝氨酸突变成丙氨酸(A),因为丙氨酸带正电荷,能够持续抑制该残基位点的活性另一方面,激活性化学修饰剂(即模拟磷酸化状态的化学修饰剂)可以包括乙酰磷酸酯、磷酰胺盐、氨基甲酰磷酸酯和焦磷酸钠等磷酸供体以及三氟化铍。此外,一些CDK4/6特异性小分子抑制剂例如Palbociclib、Ribociclib或Abemaciclib也可以实现使蛋白保持非磷酸化的效果(Maiani et al.,2021;Simoneschi et al.,2021)。It is known in the art that the recognition of substrates by ligases responsible for protein ubiquitination relies on the phosphorylation of specific sites on the substrate (usually serine, threonine, tyrosine), and the essence of phosphorylation lies in the protein's The charge changes. Therefore, by simulating the phosphorylation state of the corresponding site (ie, negatively charged), it is often possible to achieve a simulated phosphorylation state of the substrate protein. In this case, the phosphorylated protein can be recognized by ubiquitin ligase, thereby promoting protein degradation. Simulation of phosphorylation/non-phosphorylation is usually achieved using mutations or chemical modifications. For example, persistent activating mutations (i.e., mutations that mimic the phosphorylation state) include mutating residues to aspartic acid (D) or glutamic acid (E), since these two amino acids are the only negatively charged amino acids; The most common persistent inhibitory mutation (that is, a mutation that simulates non-phosphorylation) is to mutate serine to alanine (A), because alanine is positively charged and can continuously inhibit the activity of this residue site. In one aspect, activating chemical modifiers (i.e., chemical modifiers that mimic the phosphorylation state) may include phosphate donors such as acetyl phosphate, phosphoramide salts, carbamoyl phosphates, and sodium pyrophosphate, as well as beryllium trifluoride. In addition, some CDK4/6-specific small molecule inhibitors such as Palbociclib, Ribociclib or Abemaciclib can also achieve the effect of keeping the protein non-phosphorylated (Maiani et al., 2021; Simoneschi et al., 2021).
基于本发明人的上述发现,本领域技术人员能够合理推断,只要CREPT的134 和166位残基在G1末期或G1/S转换期能够保持非磷酸状态,则该蛋白就不会被泛素化,从而不能发生降解,进而导致细胞死亡。Based on the above findings of the inventor, those skilled in the art can reasonably infer that as long as CREPT's 134 If the residue at position 166 can remain non-phosphorylated at the end of G1 or the G1/S transition, the protein will not be ubiquitinated and thus cannot be degraded, leading to cell death.
因此,本发明提供一种非磷酸化及非泛素化的CREPT蛋白变体,当所述CREPT蛋白位于G1末期或G1/S转换期的真核细胞中时,所述CREPT蛋白的134和166位点能够保持非磷酸化状态且所述蛋白保持非泛素化状态,由此使该蛋白变体在该阶段不会降解。Therefore, the present invention provides a non-phosphorylated and non-ubiquitinated CREPT protein variant. When the CREPT protein is located in a eukaryotic cell at the end of G1 or G1/S transition phase, 134 and 166 of the CREPT protein The site is able to remain unphosphorylated and the protein remains unubiquitinated, thereby preventing the protein variant from being degraded at this stage.
另外已知,SKP2连接酶或CDK2激酶的缺失能抑制细胞生长,但不能使细胞死亡(Lin et al.,2010;Zhu,2010;Berthet et al.,2003;Tadesse et al.,2019),这说明CREPT在调控细胞周期中发挥重要的核心作用。CREPT的134和166位的S残基位于CREPT的连接区域,构成被SKP2连接酶识别的磷酸化降解决定子,并且在同源蛋白之间高度保守(图4C)。以上说明134和166这两个残基的磷酸化状态在调控细胞周期中具有至关重要的作用。It is also known that deletion of SKP2 ligase or CDK2 kinase can inhibit cell growth but not cell death (Lin et al., 2010; Zhu, 2010; Berthet et al., 2003; Tadesse et al., 2019). This shows that CREPT plays an important core role in regulating the cell cycle. S residues 134 and 166 of CREPT are located in the connection region of CREPT, constitute a phosphorylated degron recognized by SKP2 ligase, and are highly conserved among homologous proteins (Figure 4C). The above shows that the phosphorylation status of residues 134 and 166 plays a crucial role in regulating the cell cycle.
发明人还发现,人CREPT蛋白的S134A/S166A突变形式诱导细胞凋亡的效果在其他真核生物中也存在。发明人在酿酒酵母中外源表达了人CREPT S134A/S166A,结果该蛋白变体在不同温度下会损害酵母的存活(图13F)。为了排除酵母中内源性Rtt103(CREPT的同源蛋白)的影响,在Rtt103缺失型酵母菌株中也外源表达了人CREPT S134A/S166A,结果其也显著阻断酵母的生长(图5F)。这说明CREPT蛋白及其同源蛋白的细胞周期调控作用和调控机理在真核生物中是普遍适用的,也说明了外源导入CREPT突变体能够控制真核细胞、优选肿瘤细胞的细胞周期,并诱导细胞凋亡。The inventors also found that the effect of the S134A/S166A mutant form of human CREPT protein on inducing cell apoptosis also exists in other eukaryotes. The inventors exogenously expressed human CREPT S134A/S166A in Saccharomyces cerevisiae. As a result, the protein variant impaired the survival of yeast at different temperatures (Figure 13F). In order to exclude the influence of endogenous Rtt103 (homologous protein of CREPT) in yeast, human CREPT S134A/S166A was also exogenously expressed in the Rtt103-deficient yeast strain, and the results also significantly blocked the growth of yeast (Figure 5F). This shows that the cell cycle regulatory role and regulatory mechanism of CREPT protein and its homologous proteins are universally applicable in eukaryotes. It also shows that exogenous introduction of CREPT mutants can control the cell cycle of eukaryotic cells, preferably tumor cells, and Induces apoptosis.
因此,本发明的另一方面提供了一种通过对CREPT蛋白序列(SEQ ID NO:4)中的134和166位残基进行磷酸化失活修饰而得到的蛋白,所述磷酸化失活修饰使得当所述蛋白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述蛋白保持非泛素化状态,从而使所述蛋白在所述真核细胞中不被降解。Therefore, another aspect of the invention provides a protein obtained by phosphorylation inactivation modification of residues 134 and 166 in the CREPT protein sequence (SEQ ID NO: 4), the phosphorylation inactivation modification So that when the protein is located in eukaryotic cells in the late G1 phase or the G1/S transition phase, the 134 and 166 positions remain in a non-phosphorylated state and the protein remains in a non-ubiquitinated state, so that the protein in It is not degraded in eukaryotic cells.
在一个实施方式中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。In one embodiment, the eukaryotic cell is a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell .
在一个实施方式中,134和166位点上的所述磷酸化失活修饰是氨基酸突变和/或化学修饰。 In one embodiment, the phosphorylation-inactivating modifications at positions 134 and 166 are amino acid mutations and/or chemical modifications.
在一个实施方式中,134和166位点上的所述磷酸化失活修饰是将丝氨酸(S)均突变为丙氨酸(A),此时所述蛋白的氨基酸序列为SEQ ID No:2,即CREPT的S134A/S166A双突变形式。In one embodiment, the phosphorylation inactivation modification at positions 134 and 166 is to mutate serine (S) to alanine (A). At this time, the amino acid sequence of the protein is SEQ ID No: 2 , the S134A/S166A double mutant form of CREPT.
在一个实施方式中,所述蛋白的N端和/或C端可以连接有标签序列或引导序列。在一个实施方式中,所述连接是共价连接。在一个实施方式中,带有标签序列或引导序列的所述蛋白是融合蛋白。在一个实施方式中,带有标签序列或引导序列的所述蛋白是缀合蛋白。在一个实施方式中,标签序列可以是例如纯化标签、荧光标签、增溶标签、亲和标签或抗原表位标签等。在一个实施方式中,引导序列可以是将所述蛋白跨过细胞膜引导至细胞内的多肽序列,包括例如不基于胞吞作用的细胞穿膜肽,和本身易于通过胞吞作用进入细胞的肽序列或蛋白序列。In one embodiment, a tag sequence or a guide sequence can be connected to the N-terminus and/or C-terminus of the protein. In one embodiment, the linkage is a covalent linkage. In one embodiment, the protein with a tag sequence or leader sequence is a fusion protein. In one embodiment, the protein with a tag sequence or leader sequence is a conjugated protein. In one embodiment, the tag sequence may be, for example, a purification tag, a fluorescent tag, a solubilization tag, an affinity tag, an epitope tag, or the like. In one embodiment, the guide sequence may be a polypeptide sequence that guides the protein across the cell membrane into the cell, including, for example, cell-penetrating peptides that are not based on endocytosis, and peptide sequences that are themselves prone to enter cells via endocytosis. or protein sequence.
本发明还提供与上述蛋白具有相同的134和166位点上的磷酸化失活修饰且具有90%以上、95%以上、优选98%以上或99%以上的序列同一性的蛋白。The present invention also provides proteins that have the same phosphorylation inactivation modifications at positions 134 and 166 as the above-mentioned proteins and have a sequence identity of more than 90%, more than 95%, preferably more than 98% or more than 99%.
在另一方面,本发明提供编码上述蛋白的核酸,包含所述核酸的载体,以及包含所述载体的细胞。In another aspect, the invention provides nucleic acids encoding the above-mentioned proteins, vectors comprising the nucleic acids, and cells comprising the vectors.
将目标蛋白(例如本发明的CREPT S134A/S166A)导入靶细胞(例如癌细胞)的方法可以包括将表达目标蛋白的载体通过转染、感染或其他方式导入靶细胞中,或者也可以采用化学修饰的mRNA(modRNA)来实现目标蛋白在靶细胞中的表达。此外,可以采用例如上述引导序列将目标蛋白直接导入细胞。但本发明不限于此。例如,可以利用精准基因编辑技术(例如prime editors)直接对肿瘤基因组中的目标位点进行突变(Anzalone,et al.,2019),例如将基因组CREPT的相应碱基突变以使细胞表达CREPT S134A/S166A。The method of introducing the target protein (such as CREPT S134A/S166A of the present invention) into target cells (such as cancer cells) may include introducing a vector expressing the target protein into the target cells through transfection, infection or other means, or chemical modification may also be used mRNA (modRNA) to achieve the expression of target proteins in target cells. In addition, the target protein can be directly introduced into cells using, for example, the guide sequence described above. However, the present invention is not limited to this. For example, precision gene editing technology (such as prime editors) can be used to directly mutate target sites in the tumor genome (Anzalone, et al., 2019), such as mutating the corresponding bases of CREPT in the genome to make cells express CREPT S134A/ S166A.
在又一方面,本发明提供上述蛋白、核酸或载体在制备抑制真核细胞增殖、抑制真核细胞的DNA复制、调控真核细胞周期或杀灭真核细胞的试剂中的应用。In another aspect, the present invention provides the use of the above-mentioned protein, nucleic acid or vector in preparing reagents for inhibiting eukaryotic cell proliferation, inhibiting DNA replication of eukaryotic cells, regulating eukaryotic cell cycle or killing eukaryotic cells.
在一个实施方式中,所述真核细胞是人、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫、酵母或拟南芥的细胞。In one embodiment, the eukaryotic cell is a human, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, yeast or Arabidopsis thaliana cell.
由于CREPT在多数癌中高表达(Li et al.,2021;Lu et al.,2012),因此,在另一方面,本发明提供上述蛋白、核酸或载体在制备抗癌药中的应用。Since CREPT is highly expressed in most cancers (Li et al., 2021; Lu et al., 2012), on the other hand, the present invention provides the use of the above-mentioned proteins, nucleic acids or vectors in the preparation of anti-cancer drugs.
在又一方面,本发明提供一种治疗癌症的方法,所述方法包括向人类受试者施用有效量的上述蛋白、核酸或载体;或者,所述方法包括利用基于CRISPR/Cas9的基 因编辑技术编辑受试者癌细胞基因组中的CRPET基因,以使所述癌细胞表达SEQ ID NO:2的蛋白。在一个实施方式中,所述癌症是肝癌、肾癌、胃癌或结直肠癌。在一个实施方式中,所述方法包括将上述核酸导入肿瘤细胞。在一个实施方式中,在向受试者施用有效量的所述蛋白、核酸或载体之前、过程中或之后,降低或消除受试者的癌细胞中的野生型CREPT的表达。在另一方面,本发明提供了一种药物组合物,其包含:上述蛋白、核酸或载体,和药学上可接受的载体、赋形剂或介质。在一个实施方式中,所述药物组合物用于治疗癌症,例如肝癌、肾癌、胃癌或结直肠癌。In yet another aspect, the present invention provides a method of treating cancer, the method comprising administering an effective amount of the above-mentioned protein, nucleic acid or vector to a human subject; alternatively, the method comprising utilizing a CRISPR/Cas9-based gene The editing technology edits the CRPET gene in the genome of the subject's cancer cells so that the cancer cells express the protein of SEQ ID NO: 2. In one embodiment, the cancer is liver cancer, kidney cancer, gastric cancer, or colorectal cancer. In one embodiment, the method includes introducing the above-described nucleic acid into a tumor cell. In one embodiment, expression of wild-type CREPT is reduced or eliminated in cancer cells of the subject before, during, or after administration of an effective amount of the protein, nucleic acid, or vector to the subject. In another aspect, the present invention provides a pharmaceutical composition, which includes: the above-mentioned protein, nucleic acid or carrier, and a pharmaceutically acceptable carrier, excipient or medium. In one embodiment, the pharmaceutical composition is used to treat cancer, such as liver, kidney, stomach or colorectal cancer.
由于CREPT蛋白的134和166这两个残基的磷酸化状态在调控细胞周期中具有至关重要的作用,因此,本发明的另一方面提供一种筛选CREPT蛋白的非磷酸化非泛素化修饰剂的方法或鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述修饰剂或抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;特别是当所述CREPT蛋白位于G1末期或G1/S转换期的真核细胞中时,所述S134和S166位点能够保持持续非磷酸化状态且所述蛋白能够保持非泛素化状态,从而使所述蛋白不会被降解;其中,所述CREPT蛋白的氨基酸序列为SEQ ID No:4。Since the phosphorylation status of residues 134 and 166 of the CREPT protein plays a crucial role in regulating the cell cycle, another aspect of the present invention provides a method for screening non-phosphorylation and non-ubiquitination of the CREPT protein. A method of modifying the agent or a method of identifying whether the substance is a phosphorylation inhibitor of the CREPT protein, wherein the modifying agent or inhibitor maintains the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby making the CREPT protein Maintains a non-ubiquitinated state in cells and will not be degraded; especially when the CREPT protein is located in eukaryotic cells at the end of G1 phase or G1/S transition phase, the S134 and S166 sites can remain continuously non-phosphorylated state and the protein can maintain a non-ubiquitinated state, so that the protein will not be degraded; wherein, the amino acid sequence of the CREPT protein is SEQ ID No: 4.
在一个实施方式中,所述方法可以如下进行:将模拟非磷酸化状态的候选修饰剂或待鉴定的物质加入表达CREPT蛋白的被同步至G1期的真核细胞中,然后释放并培养所述真核细胞,并在所述真核细胞存活时检查所述CREPT蛋白的S134和S166位点的磷酸化水平。在另一个实施方式中,所述方法可以如下进行:在体外将模拟非磷酸化状态的候选修饰剂或待鉴定的物质与CREPT蛋白温育,并在Cyclin E/CDK2激酶的催化条件下检查所述CREPT蛋白的S134和S166位点的磷酸化水平。在上述法中,如果经步骤i)或ii)处理的CREPT蛋白的S134和S166位点的磷酸化水平相对于各自的未经处理的对照下降,例如下降10%以上、20%以上、30%以上或40%以上,优选下降50%以上、60%以上、70%以上、80%以上或90%以上,则将所述候选修饰剂筛选为CREPT蛋白的非磷酸化非泛素化修饰剂,或将所述物质鉴定为CREPT蛋白的磷酸化抑制剂;否则所述候选修饰剂不是CREPT蛋白的非磷酸化非泛素化修饰剂,且所述物质不是CREPT蛋白的磷酸化抑制剂。在上述方法中,可以采用质谱法或免疫沉淀法来检查所述CREPT蛋白的S134和S166位点的磷酸化状态。所述免疫沉淀法可以包括:使用识别CREPT蛋白的S134和S166位点的磷酸化的抗磷酸化抗 体来进行免疫沉淀,由此对CREPT蛋白的S134位点的磷酸化水平进行定量。具体而言,可以用抗CREPT抗体使CREPT蛋白沉淀,并测定CREPT蛋白总量作为本底量,对于用抗CREPT抗体免疫沉淀下来的蛋白,利用抗磷酸化抗体来检测磷酸化的蛋白并对其进行定量,此时,磷酸化水平可以是磷酸化蛋白的量相对于CREPT蛋白本底量的相对值,并可以针对对照进行归一化。In one embodiment, the method can be performed as follows: adding a candidate modifier that simulates a non-phosphorylated state or a substance to be identified into eukaryotic cells expressing CREPT protein that are synchronized to the G1 phase, and then releasing and culturing the eukaryotic cells, and the phosphorylation levels of S134 and S166 of the CREPT protein were examined when the eukaryotic cells were alive. In another embodiment, the method can be performed as follows: in vitro, a candidate modifier that simulates a non-phosphorylated state or a substance to be identified is incubated with the CREPT protein, and the results are examined under catalytic conditions of Cyclin E/CDK2 kinase. The phosphorylation levels of S134 and S166 of CREPT protein were described. In the above method, if the phosphorylation levels of S134 and S166 of the CREPT protein treated in step i) or ii) decrease relative to the respective untreated control, for example, by more than 10%, more than 20%, or 30% or more than 40%, preferably more than 50%, more than 60%, more than 70%, more than 80% or more than 90%, then the candidate modifier is screened as a non-phosphorylation non-ubiquitination modifier of the CREPT protein, Or the substance is identified as a phosphorylation inhibitor of the CREPT protein; otherwise the candidate modifier is not a non-phosphorylation non-ubiquitination modifier of the CREPT protein, and the substance is not a phosphorylation inhibitor of the CREPT protein. In the above method, mass spectrometry or immunoprecipitation can be used to check the phosphorylation status of the S134 and S166 sites of the CREPT protein. The immunoprecipitation method may include: using anti-phosphorylation antibodies that recognize the phosphorylation of S134 and S166 of the CREPT protein. Immunoprecipitation was performed with the body to quantify the phosphorylation level of the S134 site of CREPT protein. Specifically, anti-CREPT antibodies can be used to precipitate CREPT proteins, and the total amount of CREPT proteins can be measured as the background amount. For proteins immunoprecipitated with anti-CREPT antibodies, anti-phosphorylated antibodies can be used to detect phosphorylated proteins and detect them. For quantification, the phosphorylation level can be expressed as the amount of phosphorylated protein relative to the background amount of CREPT protein, and can be normalized to a control.
本发明还涉及以下化合物:
The invention also relates to the following compounds:
以及该化合物在制备CREPT蛋白的S134和S166位点的磷酸化抑制剂或CREPT蛋白的泛素化抑制剂中的用途、在制备治疗癌症的药物中的用途。在一个实施方式中,所述癌症是黑色素瘤、肝癌、肾癌、胃癌或结直肠癌。As well as the use of the compound in preparing phosphorylation inhibitors of S134 and S166 sites of CREPT protein or ubiquitination inhibitors of CREPT protein, and its use in preparing drugs for treating cancer. In one embodiment, the cancer is melanoma, liver cancer, kidney cancer, gastric cancer, or colorectal cancer.
由于CREPT蛋白的S134和S166位点的磷酸化是该蛋白泛素化的必要条件,因此,可以利用泛素化水平来鉴定CREPT蛋白的磷酸化泛素化抑制剂。因此,本发明还提供一种鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列为SEQ ID No:4,所述方法包括:i)将待鉴定的物质加入表达CREPT蛋白的真核细胞中并培养所述真核细胞,和ii)使用免疫沉淀法检查所述真核细胞的CREPT蛋白的泛素化水平;与未经所述物质处理的对照细胞中CREPT蛋白的泛素化水平相比,如果经处理的细胞中的CREPT蛋白的泛素化水平下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述物质鉴定为CREPT蛋白的磷酸化抑制剂,否则所述物质不是CREPT蛋白的磷酸化抑制剂。在步骤i)之前,所述方法还可以包括:使用预测工具SwissTargetPrediction和SEA针对CREPT来设计所述待鉴定的物质。此外,步骤ii)可以包括用识别CREPT蛋白的抗CREPT抗体和识别泛素的泛素抗体对CREPT蛋白的泛素化水平进行定量。泛素化水平可以是泛素化的蛋白量相对于CREPT蛋白总量的相对值,并可以针对对照进行归一化。 Since the phosphorylation of S134 and S166 of CREPT protein is a necessary condition for ubiquitination of the protein, the ubiquitination level can be used to identify phosphorylation ubiquitination inhibitors of CREPT protein. Therefore, the present invention also provides a method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby making the CREPT protein Maintains a non-ubiquitinated state in cells and will not be degraded; the amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes: i) adding the substance to be identified into eukaryotic cells expressing the CREPT protein and Culturing the eukaryotic cells, and ii) using an immunoprecipitation method to examine the ubiquitination level of the CREPT protein in the eukaryotic cells; compared with the ubiquitination level of the CREPT protein in control cells not treated with the substance, If the ubiquitination level of CREPT protein in the treated cells decreases, for example, by more than 10%, more than 20%, more than 30%, or more than 40%, the substance is identified as a phosphorylation inhibitor of CREPT protein, otherwise Said substance is not a phosphorylation inhibitor of CREPT protein. Before step i), the method may further include: using prediction tools SwissTargetPrediction and SEA to design the substance to be identified for CREPT. Additionally, step ii) may include quantifying the ubiquitination level of the CREPT protein using an anti-CREPT antibody that recognizes the CREPT protein and a ubiquitin antibody that recognizes ubiquitin. The ubiquitination level can be a relative value of the amount of ubiquitinated protein relative to the total amount of CREPT protein and can be normalized to a control.
此外,CREPT中的S134和S166对应于酵母Rtt103中的S136和S174(见图4C),发明人发现Rtt103S136A/S174A双突变蛋白的过表达导致了Rtt103缺失型酵母的致命表型(图5G)。这些结果表明,人CREPT蛋白在其他真核细胞生物中的同源蛋白具有相同或相似的调控细胞周期的作用机理。In addition, S134 and S166 in CREPT correspond to S136 and S174 in yeast Rtt103 (see Figure 4C), and the inventors found that overexpression of the Rtt103S136A/S174A double mutant protein resulted in a lethal phenotype of Rtt103-deficient yeast (Figure 5G). These results indicate that homologous proteins of human CREPT protein in other eukaryotic cell organisms have the same or similar mechanism of action in regulating the cell cycle.
因此,在又一方面,本发明提供了源自非人真核生物的人CREPT的同源蛋白,其在对应于人CREPT蛋白的134和166位点的同源性位点处具有磷酸化失活修饰,所述磷酸化失活修饰使得当所述同源蛋白位于所述真核生物的G1末期或G1/S转换期细胞中时,所述同源性位点保持非磷酸化状态且所述同源蛋白保持非泛素化状态,从而使所述同源蛋白在所述细胞中不会被降解,进而导致细胞周期阻滞和细胞凋亡。Therefore, in yet another aspect, the invention provides homologous proteins of human CREPT derived from non-human eukaryotes, which have a loss of phosphorylation at homology sites corresponding to positions 134 and 166 of the human CREPT protein. Inactive modification, the phosphorylation inactivating modification enables the homologous site to remain in an unphosphorylated state and the homologous site remains unphosphorylated when the homologous protein is located in cells at the end of G1 phase or G1/S transition phase of the eukaryotic organism. The homologous protein remains in a non-ubiquitinated state, so that the homologous protein will not be degraded in the cell, leading to cell cycle arrest and apoptosis.
在一个实施方式中,所述真核生物是酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥。In one embodiment, the eukaryotic organism is yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, or Arabidopsis thaliana.
在一个实施方式中,所述对应于人CREPT蛋白的134和166位点的同源性位点上的所述磷酸化失活修饰是氨基酸突变和/或化学修饰。在一个实施方式中,所述对应于人CREPT蛋白的134和166位点的同源性位点上的所述磷酸化失活修饰是将丝氨酸突变为丙氨酸。在一个实施方式中,所述同源蛋白是酿酒酵母Rtt103 S136A/S174A双突变蛋白,其氨基酸序列为SEQ ID No:6。In one embodiment, the phosphorylation-inactivating modifications on the homology sites corresponding to positions 134 and 166 of the human CREPT protein are amino acid mutations and/or chemical modifications. In one embodiment, the phosphorylation-inactivating modification at the homology site corresponding to positions 134 and 166 of the human CREPT protein is to mutate serine to alanine. In one embodiment, the homologous protein is Saccharomyces cerevisiae Rtt103 S136A/S174A double mutant protein, whose amino acid sequence is SEQ ID No: 6.
在一个实施方式中,所述同源蛋白的N端和/或C端可以连接有标签序列或引导序列,以形成例如融合蛋白或缀合蛋白。In one embodiment, the N-terminus and/or C-terminus of the homologous protein can be connected with a tag sequence or a leader sequence to form, for example, a fusion protein or a conjugated protein.
在一个实施方式中,还提供与上述蛋白在所述同源性位点上具有相同的磷酸化失活修饰且具有90%以上、95%以上、优选98%以上或99%以上的序列同一性的蛋白。In one embodiment, it is also provided that the protein has the same phosphorylation inactivation modification at the homology site and has a sequence identity of more than 90%, more than 95%, preferably more than 98% or more than 99%. of protein.
在一个实施方式中,本发明提供编码上述同源蛋白的核酸,包含所述核酸的载体,以及包含所述载体的细胞。In one embodiment, the present invention provides nucleic acids encoding the above-mentioned homologous proteins, vectors containing the nucleic acids, and cells containing the vectors.
在一个实施方式中,本发明提供上述同源蛋白、核酸或载体在制备抑制真核细胞增殖、抑制真核细胞的DNA复制、调控真核细胞周期或杀灭真核细胞的试剂中的应用。In one embodiment, the present invention provides the use of the above homologous proteins, nucleic acids or vectors in preparing reagents for inhibiting eukaryotic cell proliferation, inhibiting DNA replication of eukaryotic cells, regulating eukaryotic cell cycle or killing eukaryotic cells.
在一个实施方式中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞。In one embodiment, the eukaryotic cell is a human, yeast, mouse, canine, feline, chicken, toad, zebrafish, Drosophila, nematode, or Arabidopsis thaliana cell.
除了对CREPT蛋白的134和166位点进行磷酸化失活修饰外,发明人还发现,即使不修饰CREPT,使用针对SKP2或CUL1的siRNA将它们敲低或耗尽,也能够 在细胞中显著抑制野生型CREPT蛋白的降解(见图3H和图3I,图11E和图11I)。In addition to phosphorylation inactivation modifications at positions 134 and 166 of the CREPT protein, the inventors also found that even without modifying CREPT, knocking down or depleting them using siRNA targeting SKP2 or CUL1 can also Significantly inhibited the degradation of wild-type CREPT protein in cells (see Figure 3H and Figure 3I, Figure 11E and Figure 11I).
因此,在另一方面,本发明提供一种抑制CREPT蛋白在真核细胞中的降解的方法,所述方法包括:1)使选自SKP2抑制剂、CUL1抑制剂、neddylation抑制剂和CDK2抑制剂的抑制剂进入表达CREPT的真核细胞中;和/或2)对CREPT蛋白的第134和166位点进行磷酸化失活修饰,使得当经修饰的蛋白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述经修饰的蛋白保持非泛素化状态,从而使所述经修饰的蛋白不被降解。在一个实施方式中,所述真核细胞可以是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。在一个实施方式中,所述134和166位点上的所述磷酸化失活修饰可以是氨基酸突变和/或化学修饰,优选地,是将丝氨酸(S)突变为丙氨酸(A)。在一个实施方式中,SKP2抑制剂可以是针对SKP2的双链siRNA,其序列可以为AAUCUAAGCCUGGAAGGCCUGdTdT;CUL1抑制剂可以是针对CUL1的双链siRNA,其序列可以为UAGACAUUGGGUUCGCCGUdTdT;neddylation抑制剂可以是MLN4924。Therefore, on the other hand, the present invention provides a method for inhibiting the degradation of CREPT protein in eukaryotic cells, the method comprising: 1) using an inhibitor selected from the group consisting of SKP2 inhibitors, CUL1 inhibitors, neddylation inhibitors and CDK2 inhibitors The inhibitor enters the eukaryotic cells expressing CREPT; and/or 2) phosphorylation and inactivation modification of the 134th and 166th positions of the CREPT protein, so that when the modified protein is located at the end of G1 phase or the G1/S transition phase, In eukaryotic cells, the 134 and 166 sites remain in a non-phosphorylated state and the modified protein remains in a non-ubiquitinated state, thereby preventing the modified protein from being degraded. In one embodiment, the eukaryotic cell may be a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell, preferably a human cell, more preferably a human cancer cell cell. In one embodiment, the phosphorylation inactivating modification at the 134 and 166 positions may be an amino acid mutation and/or a chemical modification, preferably, the mutation of serine (S) to alanine (A). In one embodiment, the SKP2 inhibitor can be a double-stranded siRNA directed against SKP2, and its sequence can be AAUCUAAGCCUGGAAGGCCUGdTdT; the CUL1 inhibitor can be a double-stranded siRNA directed against CUL1, and its sequence can be UAGACAUUGGGUUCGCCGUdTdT; the neddylation inhibitor can be MLN4924.
此外,如图4D所示,CREPT的单突变蛋白S134A或S166A都能够在一定程度上抑制泛素化,因此能够理解,这些单突变蛋白及在其他真核细胞中的同源蛋白的类似单突变体都具有抑制CREPT降解的功能。In addition, as shown in Figure 4D, the single mutant proteins S134A or S166A of CREPT can inhibit ubiquitination to a certain extent. Therefore, it can be understood that similar single mutations of these single mutant proteins and homologous proteins in other eukaryotic cells All have the function of inhibiting CREPT degradation.
因此,在又一方面,本发明还提供以下蛋白:1)通过将SEQ ID No:4的166位丝氨酸突变为丙氨酸而得到的蛋白;2)通过将SEQ ID No:8的136位丝氨酸突变为丙氨酸而得到的蛋白;3)通过将SEQ ID No:8的174位丝氨酸突变为丙氨酸而得到的蛋白;和4)与1)至3)中的任一种蛋白具有90%以上的序列同一性且具有相同的丙氨酸突变的蛋白。此外,本发明还提供编码所述蛋白的核酸和包含所述核酸的载体。Therefore, in yet another aspect, the present invention also provides the following proteins: 1) a protein obtained by mutating serine 166 of SEQ ID No: 4 to alanine; 2) a protein obtained by mutating serine 136 of SEQ ID No: 8 A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; 3) A protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine; and 4) A protein having 90% affinity with any one of 1) to 3) Proteins with more than % sequence identity and the same alanine mutation. In addition, the present invention also provides nucleic acids encoding the proteins and vectors containing the nucleic acids.
序列说明:Sequence description:
SEQ ID NO:1编码CREPT S134A/S166A双突变蛋白的核酸序列;SEQ ID NO:1 Nucleic acid sequence encoding CREPT S134A/S166A double mutant protein;
SEQ ID NO:2CREPT S134A/S166A双突变蛋白的氨基酸序列;SEQ ID NO:2CREPT Amino acid sequence of S134A/S166A double mutant protein;
SEQ ID NO:3野生型CREPT的编码核酸序列;SEQ ID NO:3 Coding nucleic acid sequence of wild-type CREPT;
SEQ ID NO:4野生型CREPT的氨基酸序列;SEQ ID NO:4 Amino acid sequence of wild-type CREPT;
SEQ ID NO:5编码Rtt103 S136A/S174A双突变蛋白的核酸序列;SEQ ID NO:5 Nucleic acid sequence encoding Rtt103 S136A/S174A double mutant protein;
SEQ ID NO:6Rtt103 S136A/S174A双突变蛋白的氨基酸序列; SEQ ID NO: 6Rtt103 Amino acid sequence of S136A/S174A double mutant protein;
SEQ ID NO:7野生型Rtt103的编码核酸序列;SEQ ID NO:7 Coding nucleic acid sequence of wild-type Rtt103;
SEQ ID NO:8野生型Rtt103的氨基酸序列。SEQ ID NO:8 Amino acid sequence of wild-type Rtt103.
实施例Example
实施例1.CREPT在G1/S转换期降解、在S期恢复Example 1. CREPT degrades in the G1/S transition phase and recovers in the S phase
对DLD1和Hela细胞进行了免疫荧光(IF)染色实验以确定CREPT在肿瘤细胞中的表达模式。结果发现,大多数肿瘤细胞表达丰富的CREPT,但有少数肿瘤细胞为CREPT阴性(图1A,见虚线圆圈)。而且CREPT阴性的肿瘤细胞的核稍大,DAPI均匀染色(图1A,DAPI染色)。因此,CREPT阴性的肿瘤细胞可能是由于在特定细胞周期阶段内CREPT消失。为了验证这个假设,发明人用CRISPR-Cas9生成了敲入了GFP-CREPT的HeLa细胞(图9A,图9B)。活细胞成像分析显示,GFP-CREPT在细胞中保持了一段时间,然后消失了近30分钟,而后又恢复(图1B)。这表明CREPT蛋白的水平在肿瘤细胞周期中振荡。Immunofluorescence (IF) staining experiments were performed on DLD1 and HeLa cells to determine the expression pattern of CREPT in tumor cells. It was found that most tumor cells expressed abundant CREPT, but a few tumor cells were CREPT negative (Figure 1A, see dotted circle). Moreover, the nuclei of CREPT-negative tumor cells were slightly larger and evenly stained with DAPI (Figure 1A, DAPI staining). Therefore, CREPT-negative tumor cells may be due to the loss of CREPT during specific cell cycle stages. To test this hypothesis, the inventors used CRISPR-Cas9 to generate HeLa cells with GFP-CREPT knocked in (Figure 9A, Figure 9B). Live-cell imaging analysis showed that GFP-CREPT remained in the cells for a period of time, then disappeared for nearly 30 minutes and then recovered (Figure 1B). This suggests that CREPT protein levels oscillate during the tumor cell cycle.
为了进一步确认CREPT蛋白在细胞周期中的振荡,使用双胸苷阻滞(DTB)将DLD1和HeLa细胞同步至G1/S转换期,并释放到不同的细胞周期时间点。荧光激活细胞分选(FACS)分析显示,在DTB处理后,超过90%的细胞同步在G1期(图9C)。蛋白质印迹显示G1/S转换期几乎检测不到CREPT蛋白,但其在S期增加(图1C和图9D,泳道1,0小时和1小时)。CREPT蛋白的这种变化伴随着Cyclin E和SKP2的相反趋势,但与Cyclin A和Cyclin B1的表达模式相似(图1C和图9D)。同时,CREPT的mRNA水平从G1/S转换到S期保持不变(图9E)。这些结果表明,CREPT蛋白在肿瘤细胞周期的G1/S转换期被降解。To further confirm the oscillation of CREPT protein in the cell cycle, double thymidine block (DTB) was used to synchronize DLD1 and HeLa cells to the G1/S transition phase and release them to different cell cycle time points. Fluorescence-activated cell sorting (FACS) analysis showed that more than 90% of cells were synchronized in G1 phase after DTB treatment (Fig. 9C). Western blotting showed that CREPT protein was almost undetectable during the G1/S transition, but it increased during S phase (Fig. 1C and Fig. 9D, lane 1, 0 and 1 h). This change in CREPT protein was accompanied by opposite trends for Cyclin E and SKP2 but similar to the expression patterns of Cyclin A and Cyclin B1 (Figure 1C and Figure 9D). At the same time, the mRNA level of CREPT remained unchanged from G1/S transition to S phase (Fig. 9E). These results indicate that CREPT protein is degraded during the G1/S transition phase of the tumor cell cycle.
为了直接可视化CREPT消失的特定时间点,在DLD1细胞中建立了荧光泛素化的细胞周期指示剂(Fucci)细胞系。在嵌入有5-乙炔基-2'-脱氧尿苷(EdU)的Fucci细胞中进行内源性IF染色(图1D、图1E和图9F、图9G)。结果显示,CREPT在同步后消失,伴随着阴性的EdU染色(S期的标志),但呈现阳性的GEMININ和CDT1染色(G1/S期的标志)(图1D,0小时)。随着肿瘤细胞周期进入S期,CREPT蛋白逐渐增加(图1D,1小时后)。定量分析表明,CREPT和EdU在G1/S转换期处于最低水平,但在肿瘤细胞释放到S期后恢复(图1E)。这些结果表明CREPT蛋白在肿瘤细胞的G1/S转换期处在最低水平。 To directly visualize specific time points of CREPT disappearance, a fluorescent ubiquitinated cell cycle indicator (Fucci) cell line was established in DLD1 cells. Endogenous IF staining was performed in Fucci cells embedded with 5-ethynyl-2'-deoxyuridine (EdU) (Figure 1D, Figure 1E and Figure 9F, Figure 9G). The results showed that CREPT disappeared after synchronization, accompanied by negative EdU staining (a marker of S phase), but positive GEMININ and CDT1 staining (markers of G1/S phase) (Figure 1D, 0 h). As the tumor cell cycle enters the S phase, CREPT protein gradually increases (Figure 1D, 1 hour later). Quantitative analysis showed that CREPT and EdU were at their lowest levels during the G1/S transition phase but recovered after tumor cells were released into the S phase (Figure 1E). These results indicate that CREPT protein is at its lowest level during the G1/S transition phase of tumor cells.
实施例2.GREPT在G1/S转换期通过SKP2的泛素化而降解Example 2. GREPT is degraded by ubiquitination of SKP2 during the G1/S transition phase
为了阐明CREPT在G1/S转换期的降解机制,发明人首先用放线菌酮(CHX)阻断蛋白合成来检查CREPT的蛋白稳定性。结果表明,CHX处理后CREPT蛋白减少(图2A,图10A,10-12h),表明CREPT蛋白降解。此外,当在细胞周期同步化过程中添加MG132(蛋白酶体诱导降解的抑制剂)时,CREPT蛋白在G1/S转换期保持在相对较高的水平(图2B,泳道7vs 1,图2C,图10B)。这些结果表明CREPT表达的降低是由于蛋白酶体诱导其降解。In order to elucidate the degradation mechanism of CREPT during the G1/S transition period, the inventors first used cycloheximide (CHX) to block protein synthesis to check the protein stability of CREPT. The results showed that CREPT protein decreased after CHX treatment (Figure 2A, Figure 10A, 10-12h), indicating that CREPT protein was degraded. Furthermore, when MG132, an inhibitor of proteasome-induced degradation, was added during cell cycle synchronization, CREPT protein remained at relatively high levels during the G1/S transition phase (Figure 2B, lanes 7 vs 1, Figure 2C, Figure 10B). These results indicate that the decrease in CREPT expression is due to the induction of its degradation by the proteasome.
为了确认CREPT的降解是否依赖于泛素,发明人进行了免疫共沉淀(Co-IP)实验来检测CREPT与泛素的相互作用。结果表明,针对Myc的抗体沉淀了Myc-CREPT和HA-泛素,而MG132增加了沉淀的HA-泛素水平(图2D),表明CREPT可被泛素修饰。进一步的泛素连接类型表征实验发现CREPT主要以K11连接被泛素化,而非K48连接(图2E)。值得注意的是,K63泛素也能够适度诱导CREPT泛素化(见图2E,泳道6和12)。这一结果是出乎意料的,因为已广泛报道K48泛素可介导蛋白降解。发明人进一步在CHX存在下与CREPT一起过度表达了不同类型的泛素。结果表明,在存在K11而不是K48或K63泛素的情况下,CREPT在减少(图2F-图2K)。此外,发明人用自噬抑制剂CQ和LEU处理将DLD1和HeLa细胞同步到G1/S期,结果表明,在CQ或LEU处理下,CREPT蛋白在DTR后0小时仍保持降解(图10C-图10D)。上述这些结果表明CREPT在G1/S转换期的降解依赖于K11连接的泛素化。In order to confirm whether the degradation of CREPT depends on ubiquitin, the inventors performed a co-immunoprecipitation (Co-IP) experiment to detect the interaction between CREPT and ubiquitin. The results showed that antibodies against Myc precipitated Myc-CREPT and HA-ubiquitin, and MG132 increased the level of precipitated HA-ubiquitin (Fig. 2D), indicating that CREPT can be modified by ubiquitin. Further experiments to characterize the type of ubiquitin linkage found that CREPT was mainly ubiquitinated via K11 linkage rather than K48 linkage (Figure 2E). Notably, K63 ubiquitin was also able to moderately induce CREPT ubiquitination (see Figure 2E, lanes 6 and 12). This result was unexpected because K48 ubiquitin has been widely reported to mediate protein degradation. The inventors further overexpressed different types of ubiquitin together with CREPT in the presence of CHX. The results showed that CREPT was reduced in the presence of K11 but not K48 or K63 ubiquitin (Figure 2F-Figure 2K). In addition, the inventors treated DLD1 and HeLa cells with autophagy inhibitors CQ and LEU to synchronize DLD1 and HeLa cells to the G1/S phase. The results showed that under CQ or LEU treatment, CREPT protein remained degraded 0 hours after DTR (Figure 10C-Figure 10D). These results indicate that the degradation of CREPT during the G1/S transition depends on K11-linked ubiquitination.
为了鉴定出针对CREPT的E3连接酶,发明人在MG132处理下将细胞同步至G1/S期,以沉淀CREPT相互作用蛋白。质谱分析表明在沉淀复合物中存在若干种E3连接酶。在与CREPT潜在相互作用的前10种E3连接酶中,SKP2与CREPT结合的可能性最高(图3A)。为了查明SKP2是否是用于CREPT降解的E3连接酶,发明人验证了它们在不同条件下的相互作用。IP实验表明,针对HA的抗体沉淀了HA-CREPT和Flag-SKP2(图3B),表明HA-CREPT与Flag-SKP2相互作用。重要的是,观察到针对CREPT的抗体在DLD1细胞中沉淀出内源性SKP2(图3C),表明CREPT和SKP2在完整细胞中相互作用。为了验证物理相互作用,发明人根据先前报道的策略(Chan等人,2013;Schulman等人,2000)在大肠杆菌中纯化了GST-CREPT和His-SKP2。GST沉降实验表明,针对GST的抗体同时沉降了GST-CREPT和His-SKP2,表明SKP2直接与CREPT结合(图3D)。这些结果表明CREPT和SKP2 在体内和体外都直接相互作用。To identify E3 ligases targeting CREPT, the inventors synchronized cells to G1/S phase under MG132 treatment to precipitate CREPT-interacting proteins. Mass spectrometry analysis revealed the presence of several E3 ligases in the precipitated complex. Among the top 10 E3 ligases that potentially interact with CREPT, SKP2 has the highest probability of binding to CREPT (Fig. 3A). To find out whether SKP2 is the E3 ligase used for CREPT degradation, the inventors verified their interaction under different conditions. IP experiments showed that antibodies against HA precipitated HA-CREPT and Flag-SKP2 (Figure 3B), indicating that HA-CREPT interacts with Flag-SKP2. Importantly, antibodies against CREPT were observed to precipitate endogenous SKP2 in DLD1 cells (Fig. 3C), indicating that CREPT and SKP2 interact in intact cells. To verify the physical interaction, the inventors purified GST-CREPT and His-SKP2 in E. coli according to previously reported strategies (Chan et al., 2013; Schulman et al., 2000). The GST sedimentation experiment showed that antibodies against GST simultaneously sedimented GST-CREPT and His-SKP2, indicating that SKP2 directly binds to CREPT (Figure 3D). These results indicate that CREPT and SKP2 Direct interactions both in vivo and in vitro.
为了证明SKP2在CREPT泛素化中的作用,发明人检查了泛素化的CREPT水平。体内泛素化测定表明,SKP2的过表达增强了Myc-CREPT的多泛素化(图3E)。为了查明SKP2是否介导CREPT降解,发明人在CHX处理下在Hela细胞中过表达了SKP2。蛋白质印迹分析显示,在CHX处理后8小时,SKP2过表达细胞的CREPT蛋白水平下降,而对照细胞中时在10小时下降,表明SKP2的过表达导致CREPT的降解加快了2小时(图3F-图3G,10-12小时)。值得注意的是,添加MG132会削弱CREPT的降解(图3F,MG132的第12小时),表明SKP2诱导的CREPT降解依赖于蛋白酶体。为了确认SKP2在体内CREPT降解中的作用,在Hela细胞中利用siRNA耗尽了内源性SKP2。结果表明,在SKP2耗尽的细胞中,在CHX处理后的不同时间点CREPT蛋白水平保持稳定(图3H-图3I)。这些表明CREPT泛素化是由F-box家族E3连接酶SKP2介导的。To demonstrate the role of SKP2 in CREPT ubiquitination, the inventors examined ubiquitinated CREPT levels. In vivo ubiquitination assay showed that overexpression of SKP2 enhanced polyubiquitination of Myc-CREPT (Fig. 3E). To find out whether SKP2 mediates CREPT degradation, the inventors overexpressed SKP2 in HeLa cells under CHX treatment. Western blot analysis showed that CREPT protein levels decreased in SKP2-overexpressing cells at 8 h after CHX treatment, compared with 10 h in control cells, indicating that overexpression of SKP2 resulted in accelerated degradation of CREPT by 2 h (Figure 3F—figure supplement 3G, 10-12 hours). Notably, addition of MG132 impaired CREPT degradation (Fig. 3F, 12 h of MG132), indicating that SKP2-induced CREPT degradation is proteasome dependent. To confirm the role of SKP2 in CREPT degradation in vivo, endogenous SKP2 was depleted using siRNA in HeLa cells. The results showed that in SKP2-depleted cells, CREPT protein levels remained stable at different time points after CHX treatment (Figure 3H-Figure 3I). These indicate that CREPT ubiquitination is mediated by the F-box family E3 ligase SKP2.
为了查明SKP2在肿瘤细胞周期对CREPT降解的作用,发明人将细胞同步至G1/S期,并在不同的释放时间点沉淀出CREPT复合物。结果表明,在细胞周期进程中,SKP2从G1/S转换期到S期表达,而CREPT在G1/S期表达最低,然后逐渐恢复(图3J,裂解物)。值得注意的是,针对CREPT的抗体在在释放后0到4小时使SKP2强烈沉淀,但10小时时没有沉淀,表明SKP2与CREPT的相互作用特定地在G1/S期而不是G2/M期(图3J-图3K,0-8小时)。In order to find out the role of SKP2 in CREPT degradation during the tumor cell cycle, the inventors synchronized the cells to the G1/S phase and precipitated the CREPT complex at different release time points. The results showed that during the cell cycle progression, SKP2 was expressed from the G1/S transition phase to the S phase, while the expression of CREPT was lowest in the G1/S phase and then gradually recovered (Fig. 3J, lysate). Notably, antibodies against CREPT strongly precipitated SKP2 from 0 to 4 hours after release, but not at 10 hours, indicating that SKP2 interacts with CREPT specifically in the G1/S phase rather than the G2/M phase ( Figure 3J-Figure 3K, 0-8 hours).
为了进一步确定肿瘤细胞周期中CREPT/SKP2相互作用的时间点,将细胞同步至G2/M期。co-IP实验表明在释放后10小时SKP2出现,但CREPT表达处于最低水平(图3J)。还观察到CREPT和SKP2的相互作用出现在细胞释放后第10小时,并一直持续到第18小时(图3L-图3M,10-18h)。流式细胞仪分析表明,在释放后10小时细胞仍处于G1/S期(图11D)。这些结果表明SKP2与CREPT在G1/S转换期相互作用并诱导其降解。To further determine the time point of CREPT/SKP2 interaction in the tumor cell cycle, cells were synchronized to G2/M phase. Co-IP experiments showed that SKP2 was present at 10 h after release, but CREPT expression was at its lowest level (Fig. 3J). It was also observed that the interaction of CREPT and SKP2 appeared at 10 hours after cell release and continued until 18 hours (Figure 3L-Figure 3M, 10-18h). Flow cytometry analysis showed that cells were still in the G1/S phase 10 hours after release (Fig. 11D). These results indicate that SKP2 interacts with CREPT during the G1/S transition phase and induces its degradation.
由于SKP2属于SCF复合蛋白的F-box家族,为了查明Cul1-RING E3泛素连接酶(CRL1)复合物是否参与CREPT降解,发明人使用了neddylation抑制剂MLN4924,它抑制所有Cullin-RING连接酶复合物的激活。结果CREPT蛋白水平在对照细胞(si Ctrl)中增加,但在MLN4924处理的SKP2耗尽细胞(si SKP2)中没有增加(图11E)。图11E表明,在对照细胞中Cullin-RING连接酶随时间(0-6小时)逐渐被MLN4924失活, 尽管SKP2始终保持显著的表达水平,但CREPT的泛素化水平逐渐下降且降解受到抑制,其水平随时间逐渐增加;而在用siRNA耗尽SKP2的细胞中CREPT的泛素化和降解始终受到抑制,因此始终保持在显著水平。以上结果说明,SKP2的缺失能够显著抑制CREPT的泛素化和降解。Since SKP2 belongs to the F-box family of SCF complex proteins, to find out whether the Cul1-RING E3 ubiquitin ligase (CRL1) complex is involved in CREPT degradation, the inventors used the neddylation inhibitor MLN4924, which inhibits all Cullin-RING ligases. Activation of the complex. Results CREPT protein levels increased in control cells (si Ctrl) but not in MLN4924-treated SKP2-depleted cells (si SKP2) (Fig. 11E). Figure 11E shows that Cullin-RING ligase was gradually inactivated by MLN4924 over time (0-6 hours) in control cells. Although SKP2 always maintains a significant expression level, the ubiquitination level of CREPT gradually decreases and its degradation is inhibited, and its level gradually increases over time; while in cells depleted of SKP2 with siRNA, the ubiquitination and degradation of CREPT are always inhibited. , so it always remains at a significant level. The above results indicate that the deletion of SKP2 can significantly inhibit the ubiquitination and degradation of CREPT.
相应地,免疫沉淀测定结果发现Flag-Cul1与HA-CREPT相互作用(图11F),并且在有或没有MG132处理的情况下Cul1的过表达增加了的CREPT的泛素化水平(图11G,见HA条带)。与此一致地,Cul1的过表将CREPT的降解加快了2小时(图11H),但用siRNA导致的Cul1的缺失保护了CREPT免受降解(图11I)。这些结果表明CRL1SKP2是识别待降解的CREPT的E3连接酶,且该连接酶的失活能够显著抑制CREPT的泛素化和降解。Correspondingly, immunoprecipitation assay results found that Flag-Cul1 interacted with HA-CREPT (Fig. 11F), and overexpression of Cul1 increased the ubiquitination level of CREPT with or without MG132 treatment (Fig. 11G, see HA band). Consistently, overexpression of Cul1 accelerated CREPT degradation by 2 hours (Fig. 11H), but deletion of Cul1 with siRNA protected CREPT from degradation (Fig. 11I). These results indicate that CRL1 SKP2 is an E3 ligase that recognizes CREPT to be degraded, and the inactivation of this ligase can significantly inhibit the ubiquitination and degradation of CREPT.
实施例3.CREPT的泛素化依赖于S134和S166的磷酸化Example 3. Ubiquitination of CREPT depends on phosphorylation of S134 and S166
为了鉴定CREPT蛋白上的泛素缀合位点,发明人在293T细胞中在MG132的存在下用HA-Ub表达了带Myc标签的CID结构域(Myc-CREPT-CID)和CCT结构域(Myc-CREPT-CCT)。蛋白质印迹分析显示Myc-CREPT-CID被泛素化,而Myc-CREPT-CCT没有(图4A),表明泛素化发生在CID结构域域。而后发明人将CID结构域中的所有单个赖氨酸(K)残基突变为精氨酸(R)以绘制泛素化位点,但是蛋白质印迹分析表明所有突变都未能损害泛素化水平(图12A和12B)。而K56R突变增加了泛素化水平(图12A,泳道9),表明K56是CREPT泛素化的抑制性残基。在突变所有K残基时,Myc-CREPT-CID仍处于高泛素化水平(图12C,最后一个泳道),然而,当突变所有K残基但不突变K56时,泛素化处于基线水平(图12C,泳道4)。这些结果表明泛素化可能发生在其他残基上,而K56是抑制性残基。To identify the ubiquitin conjugation site on the CREPT protein, the inventors expressed Myc-tagged CID domain (Myc-CREPT-CID) and CCT domain (Myc) using HA-Ub in 293T cells in the presence of MG132. -CREPT-CCT). Western blot analysis showed that Myc-CREPT-CID was ubiquitinated, but Myc-CREPT-CCT was not (Fig. 4A), indicating that ubiquitination occurs within the CID domain. The inventors then mutated all single lysine (K) residues in the CID domain to arginine (R) to map ubiquitination sites, but Western blot analysis showed that all mutations failed to impair ubiquitination levels. (Figures 12A and 12B). The K56R mutation increased ubiquitination levels (Fig. 12A, lane 9), indicating that K56 is an inhibitory residue for CREPT ubiquitination. When all K residues were mutated, Myc-CREPT-CID remained at high ubiquitination levels (Fig. 12C, last lane), however, when all K residues but not K56 were mutated, ubiquitination was at baseline levels ( Figure 12C, lane 4). These results suggest that ubiquitination may occur on other residues, with K56 being the inhibitory residue.
为了寻找其他可能的泛素连接残基,发明人还在CID结构域中的苏氨酸(T)和半胱氨酸(C)残基上产生了突变,但CREPT泛素化水平在这些突变中仍然保持不变(图12D)。这些负面结果促使发明人验证CREPT是否是具有磷酸化降解决定子的底物,因为SCF复合物更倾向于与磷酸化降解决定子结合以进行泛素化。为此,发明人通过质谱分析分析了CREPT的磷酸化修饰,发现丝氨酸134(S134)和丝氨酸166(S166)位点被高度磷酸化(图4B)。值得注意的是,这两个S残基位于CREPT的连接区域,并且在CREPT和其在酿酒酵母中的直系同源物Rtt103之间高度保守(图4C)。为了验证S134和S166的磷酸化是否调控CREPT泛素化,发明人生成了不同的突变体,包 模拟功能丧失的非磷酸化状态的S134A、S166A和S134A/S166A双突变,以及用于模拟连续磷酸化的S134E、S166E和S134E/S166E双突变。蛋白质印迹分析显示,S134A、S166A和S134A/S166A的突变损害了泛素化,但其他突变对泛素化没有影响(图4D)。该结果表明S134和S166的磷酸化对于CREPT的泛素化是必不可少的。为了确认这两个S残基是否对SKP2识别至关重要,发明人进行了IP实验,结果表明,Myc抗体沉淀了Myc-CREPT(S134E/S166E)/Flag-SKP2复合物,但没有沉淀Myc-CREPT(S134A/S166A)/Flag-SKP2复合物(图4E),表明CREPT(S134A/S166A)未能与SKP2相互作用。这些结果表明S134和S166参与形成CREPT的磷酸化降解决定子。To search for other possible ubiquitin-linking residues, the inventors also created mutations on threonine (T) and cysteine (C) residues in the CID domain, but CREPT ubiquitination levels were significantly lower with these mutations. remains unchanged (Figure 12D). These negative results prompted the inventors to verify whether CREPT is a substrate with phosphorylated degron, because the SCF complex prefers to bind to phosphorylated degron for ubiquitination. To this end, the inventors analyzed the phosphorylation modification of CREPT through mass spectrometry analysis and found that serine 134 (S134) and serine 166 (S166) sites are highly phosphorylated (Figure 4B). Notably, these two S residues are located in the linker region of CREPT and are highly conserved between CREPT and its ortholog Rtt103 in S. cerevisiae (Fig. 4C). To verify whether phosphorylation of S134 and S166 regulates CREPT ubiquitination, the inventors generated different mutants, including S134A, S166A and S134A/S166A double mutations to simulate the non-phosphorylated state of loss of function, and S134E, S166E and S134E/S166E double mutations to simulate continuous phosphorylation. Western blot analysis showed that mutations of S134A, S166A, and S134A/S166A impaired ubiquitination, but other mutations had no effect on ubiquitination (Fig. 4D). This result indicates that phosphorylation of S134 and S166 is essential for ubiquitination of CREPT. In order to confirm whether these two S residues are critical for SKP2 recognition, the inventors performed IP experiments. The results showed that the Myc antibody precipitated the Myc-CREPT(S134E/S166E)/Flag-SKP2 complex, but did not precipitate Myc- CREPT(S134A/S166A)/Flag-SKP2 complex (Fig. 4E), indicating that CREPT(S134A/S166A) failed to interact with SKP2. These results indicate that S134 and S166 are involved in forming the phosphorylated degron of CREPT.
此外,CREPT的氨基酸序列分析显示S134和S166位点都可能是具有sp序列的Cyclin E-CDK2复合物识别位点。还观察到CREPT在IP实验中与Cyclin E1和CDK2相互作用(图4F)。体外激酶测定显示GST-CREPT被CDK2和Cyclin E1磷酸化,但GST-CREPT(S134A/S166A)没有被磷酸化(图4G)。还观察到GST-CREPT在哺乳动物细胞中被磷酸化,但在大肠杆菌中没有被磷酸化(图12E)。这些结果表明,CREPT在G1/S期的降解取决于S134和S166位点处的被CRLSKP2识别的磷酸化降解决定子。In addition, the amino acid sequence analysis of CREPT showed that both the S134 and S166 sites may be the recognition sites of the Cyclin E-CDK2 complex with sp sequence. CREPT was also observed to interact with Cyclin E1 and CDK2 in IP experiments (Fig. 4F). In vitro kinase assay showed that GST-CREPT was phosphorylated by CDK2 and Cyclin E1, but GST-CREPT (S134A/S166A) was not phosphorylated (Fig. 4G). It was also observed that GST-CREPT was phosphorylated in mammalian cells but not in E. coli (Fig. 12E). These results indicate that the degradation of CREPT in G1/S phase depends on the phosphorylated degron recognized by CRL SKP2 at S134 and S166.
实施例4.未降解的CREPT蛋白变体引起细胞凋亡Example 4. Undegraded CREPT protein variants cause apoptosis
发明人发现突变蛋白CREPT(S134A/S166A)总是诱导细胞死亡(图13A,上图)。具体而言,发明人在CREPT缺失型细胞中过表达了野生型CREPT和突变CREPT。结果显示野生型(WT)CREPT和CREPT(S134E/S166E)的过表达挽救了细胞增殖,然而CREPT(S134A/S166A)的过表达导致细胞死亡显著增加(图13A,下图)。FACS分析显示CREPT(S134A/S166A)的表达导致野生型细胞和CREPT缺失型细胞的死亡显著增加,而野生型CREPT和CREPT(S134E/S166E)则没有影响(图5A-图5B)。而且CREPT(S134A/S166A)所诱导的CREPT缺失型细胞的死亡率远高于其所诱导的野生型细胞的死亡率(图5A-图5B)。相应地,当CREPT(S134A/S166A)在野生型细胞或CREPT缺失型细胞中表达时,细胞生长速率(图5D)和集落形成能力(图5D和图13C)显著受损。蛋白质印迹分析表明,CREPT(S134A/S166A)的过表达诱导了切割形式的半胱天冬酶7的表达,这是一种典型的凋亡执行器(图5C)。上述结果表明,未降解的CREPT变体诱导细胞凋亡。The inventors found that the mutant protein CREPT (S134A/S166A) always induced cell death (Figure 13A, top panel). Specifically, the inventors overexpressed wild-type CREPT and mutant CREPT in CREPT-deficient cells. The results showed that overexpression of wild-type (WT) CREPT and CREPT(S134E/S166E) rescued cell proliferation, whereas overexpression of CREPT(S134A/S166A) resulted in a significant increase in cell death (Fig. 13A, lower panel). FACS analysis showed that expression of CREPT (S134A/S166A) resulted in a significant increase in the death of wild-type cells and CREPT-deficient cells, while wild-type CREPT and CREPT (S134E/S166E) had no effect (Figure 5A-Figure 5B). Moreover, the death rate of CREPT-deficient cells induced by CREPT (S134A/S166A) was much higher than that of wild-type cells induced by CREPT (Figure 5A-Figure 5B). Correspondingly, when CREPT (S134A/S166A) was expressed in wild-type cells or CREPT-deficient cells, cell growth rate (Fig. 5D) and colony formation ability (Fig. 5D and Fig. 13C) were significantly impaired. Western blot analysis showed that overexpression of CREPT (S134A/S166A) induced the expression of the cleaved form of caspase 7, a typical apoptotic executor (Fig. 5C). The above results indicate that undegraded CREPT variants induce apoptosis.
为了确认S134A/S166A突变诱导细胞凋亡的效果是否在其他物种中也存在,发 明人在酿酒酵母(Saccharomyces cerevisiae)中外源表达了CREPT突变蛋白。结果观察到CREPT(S134A/S166A)在不同温度下会显著阻断酵母的生长(图13F)。人p15RS的外源表达对酵母存活没有影响(图13F,底部)。为了排除酵母中内源性Rtt103(CREPT的同源蛋白)的影响,在Rtt103缺失型酵母菌株中外源表达了人CREPT及其突变蛋白。结果表明,CREPT(S134A/S166A)显著阻断了酵母的生长(图5F),这表明CREPT(S134A/S166A)在酵母中也是致命的。此外,p15RS似乎也抑制了Rtt103缺失型酵母的生长,这与p15RS在哺乳动物细胞中的抑制作用相呼应。上述结果表明CREPT中的S134A和S166A的双突变能够在哺乳动物和酵母细胞中诱导细胞死亡。In order to confirm whether the apoptosis-inducing effect of S134A/S166A mutation also exists in other species, we found Akito exogenously expressed CREPT mutant protein in Saccharomyces cerevisiae. As a result, it was observed that CREPT(S134A/S166A) significantly blocked the growth of yeast at different temperatures (Figure 13F). Exogenous expression of human p15RS had no effect on yeast survival (Fig. 13F, bottom). In order to eliminate the influence of endogenous Rtt103 (homologous protein of CREPT) in yeast, human CREPT and its mutant proteins were exogenously expressed in Rtt103-deficient yeast strains. The results showed that CREPT(S134A/S166A) significantly blocked yeast growth (Figure 5F), indicating that CREPT(S134A/S166A) is also lethal in yeast. In addition, p15RS also appears to inhibit the growth of Rtt103-deficient yeast, which echoes the inhibitory effect of p15RS in mammalian cells. The above results indicate that double mutations of S134A and S166A in CREPT can induce cell death in mammalian and yeast cells.
由于CREPT中的S134和S166对应于酵母Rtt103中的S136和S174(见图4C),发明人生成了相应的酵母突变蛋白,并且观察到Rtt103(S136A/S174A)的过表达导致Rtt103缺失型酵母的致命表型(图5G)。这些结果表明,CREPT和RTT103中磷酸化降解决定子的破坏导致细胞死亡。Since S134 and S166 in CREPT correspond to S136 and S174 in yeast Rtt103 (see Figure 4C), the inventors generated the corresponding yeast mutant proteins and observed that overexpression of Rtt103 (S136A/S174A) resulted in Rtt103-deficient yeast. lethal phenotype (Figure 5G). These results indicate that disruption of the phosphorylated degron in CREPT and RTT103 leads to cell death.
实施例5.未降解的CREPT变体阻止细胞进入S期Example 5. Undegraded CREPT variants prevent cells from entering S phase
之前的结果表明CREPT的磷酸化降解决定子对G1/S转变至关重要。为了调查CREPT磷酸化对G1/S转变的作用机制,发明人鉴定了在G1/S期不能与CREPT相互作用的蛋白,具体而言,利用双胸苷阻滞将DLD1细胞系同步至G1/S期,并通过染色质免疫沉淀质谱(ChIP-MS)实验分析了与CREPT相互作用的蛋白,比较非同步和同步DLD1细胞之间的CREPT沉淀的蛋白的差异发现,MCM六聚体蛋白MCM5和MCM7在G1/S期看上去没有与CREPT相互作用,但在其他阶段仍然保持与CREPT相互作用(图6A)。该结果表明CREPT在G1/S转换期可能与MCM六聚体分离,这可能是由于CREPT的降解。Previous results indicate that the phosphorylated degron of CREPT is critical for the G1/S transition. In order to investigate the mechanism of CREPT phosphorylation on the G1/S transition, the inventors identified proteins that cannot interact with CREPT in the G1/S phase. Specifically, double thymidine blockade was used to synchronize the DLD1 cell line to G1/S. phase, and analyzed the proteins that interact with CREPT through chromatin immunoprecipitation mass spectrometry (ChIP-MS) experiments. Comparing the differences in CREPT-precipitated proteins between asynchronous and synchronous DLD1 cells, it was found that MCM hexameric proteins MCM5 and MCM7 It does not appear to interact with CREPT in the G1/S phase, but it still maintains interaction with CREPT in other phases (Fig. 6A). This result indicates that CREPT may dissociate from the MCM hexamer during the G1/S transition period, which may be due to the degradation of CREPT.
MCM六聚体包含6个MCM,包括MCM2至MCM7。为了验证CREPT和MCM六聚体的相互作用,在293T细胞中过表达了Myc-CREPT和Flag-MCM5。IP实验表明,Myc-CREPT和Flag-MCM5在未同步的293T细胞中相互作用强烈(图6B)。还观察到Myc-CREPT与Flag-MCM7相互作用(图14A),Myc-CREPT与Flag-MCM2相互作用(图14B),这表明CREPT与MCM六聚体结合。为了确认内源性相互作用,发明人在不同的交联条件下使用针对CREPT的抗体进行了co-IP实验(图14C)。结果表明,CREPT和MCM5在10分钟的交联下相互作用(图6C)。还观察到当将同步化的细胞释放0到8小时时,CREPT和MCM5强烈相互作用,但它们的相互作用看上去 在释放后10小时减弱(图6D)。值得注意的是,在释放细胞后的第10小时,CREPT维持在最低水平,对应于正在出现的SKP2和Cyclin E1(图6D,裂解物)。这些观察结果表明,由于CREPT降解,CREPT/MCM相互作用在G1/S阶段受损。发明人进一步使用未降解的突变CREPT进行IP实验,结果表明,Myc-CREPT(S134A/S166A)与Flag-MCM5的相互作用比WT蛋白和Myc-CREPT(S133E/S166E)与Flag-MCM5的相互作用更强(图6E)。该结果表明CREPT的降解导致其与MCM六聚体解离。The MCM hexamer contains 6 MCMs, including MCM2 to MCM7. To verify the interaction between CREPT and MCM hexamer, Myc-CREPT and Flag-MCM5 were overexpressed in 293T cells. IP experiments showed that Myc-CREPT and Flag-MCM5 interacted strongly in unsynchronized 293T cells (Figure 6B). Myc-CREPT was also observed to interact with Flag-MCM7 (Fig. 14A) and Myc-CREPT with Flag-MCM2 (Fig. 14B), indicating that CREPT binds to the MCM hexamer. To confirm the endogenous interaction, the inventors performed co-IP experiments using antibodies against CREPT under different cross-linking conditions (Figure 14C). The results showed that CREPT and MCM5 interacted under 10 min of cross-linking (Figure 6C). It was also observed that CREPT and MCM5 interact strongly when synchronized cells were released for 0 to 8 h, but their interaction appeared Attenuated 10 hours after release (Fig. 6D). Notably, CREPT was maintained at minimal levels at 10 hours after releasing cells, corresponding to the emerging SKP2 and Cyclin E1 (Fig. 6D, lysates). These observations suggest that the CREPT/MCM interaction is impaired during the G1/S phase due to CREPT degradation. The inventor further used undegraded mutant CREPT to conduct IP experiments, and the results showed that the interaction between Myc-CREPT (S134A/S166A) and Flag-MCM5 was greater than the interaction between WT protein and Myc-CREPT (S133E/S166E) and Flag-MCM5. stronger (Fig. 6E). This result indicates that the degradation of CREPT leads to its dissociation from the MCM hexamer.
为了破译CREPT/MCM相互作用和解离,发明人检查了MCM六聚体在染色质DNA上的占有率。为此,使用了不同的固定策略对MCM5和CREPT蛋白进行染色。结果表明,使用1%多聚甲醛直接固定时,MCM5在细胞中均匀染色(图14D,上图),但在固定前使用TritonX100预提取时,在某些细胞中变为阴性(图14D,下图)。而CREPT未在预提取的细胞中染色,这意味着CREPT不直接与DNA结合。为了查明CREPT的降解是否调节MCM六聚体在染色质DNA上的占有率,过表达了未降解的CREPT突变蛋白Myc-CREPT(S134A/S166A)并在预提取的条件下染色MCM5。结果表明,当Myc-CREPT(S134A/S166A)过表达时,MCM5被强烈染色(图6F和图6G),这意味着双MCM六聚体加载到染色质DNA上,且细胞周期停滞于G1末期或G1/S期。FACS分析证实CREPT(S134A/S166A)的表达导致S期细胞显著减少(图14E)。值得注意的是,所有MCM5阳性细胞的核都变大,并且在Myc-CREPT(S134A/S166A)过表达的细胞中显示出EdU阴性染色(图6G,见EdU染色)。这些结果表明,所有具有Myc-CREPT(S134A/S166A)过表达的MCM5强阳性细胞在进入S期之前细胞周期就阻滞了。综上,所有结果表明CREPT的降解使得MCM六聚体解离,并且这种解离的失败使细胞周期在进入S期之前停滞。To decipher CREPT/MCM interactions and dissociation, the inventors examined the occupancy of MCM hexamers on chromatin DNA. To this end, different fixation strategies were used to stain MCM5 and CREPT proteins. The results showed that MCM5 stained uniformly in cells when directly fixed with 1% paraformaldehyde (Figure 14D, top panel), but became negative in some cells when pre-extracted with TritonX100 before fixation (Figure 14D, bottom panel). picture). CREPT was not stained in pre-extracted cells, which means that CREPT does not bind directly to DNA. To find out whether the degradation of CREPT regulates the occupancy of MCM hexamers on chromatin DNA, the non-degraded CREPT mutant protein Myc-CREPT (S134A/S166A) was overexpressed and MCM5 was stained under pre-extraction conditions. The results show that when Myc-CREPT (S134A/S166A) is overexpressed, MCM5 is strongly stained (Figure 6F and Figure 6G), which means that double MCM hexamers are loaded onto chromatin DNA and the cell cycle is arrested at the end of G1 phase or G1/S phase. FACS analysis confirmed that expression of CREPT (S134A/S166A) resulted in a significant reduction of cells in S phase (Fig. 14E). Notably, the nuclei of all MCM5-positive cells were enlarged and showed EdU-negative staining in cells overexpressing Myc-CREPT (S134A/S166A) (Fig. 6G, see EdU staining). These results indicate that all MCM5 strongly positive cells with Myc-CREPT (S134A/S166A) overexpression were cell cycle arrested before entering the S phase. Taken together, all results indicate that degradation of CREPT dissociates MCM hexamers and that failure of this dissociation arrests the cell cycle before entering S phase.
实施例6.未降解的CREPT变体使DNA复制叉中止Example 6. Undegraded CREPT variants abort DNA replication forks
未降解的CREPT突变体通过与MCM六聚体保持结合而导致细胞死亡并使细胞周期停滞,因此发明人验证了CREPT-MCM复合物是否在G1/S转变期诱导基因组胁迫。由于基因组胁迫导致DNA损伤,发明人进行了TUNEL染色实验来检查DNA链断裂。结果表明,CREPT(S134A/S166A)的过表达导致WT细胞和CREPT缺失型细胞中存在显著的TUNEL信号,而WT蛋白和CREPT(S134E/S166E)的过表达显示出阴性信号(图15A)。值得注意的是,CREPT(S134A/S166A)的过表达在CREPT缺失型细胞中产生的TUNEL信号比在WT细胞中强得多(图15A,比较KO与Mock)。相 应地,还观察到γH2AX焦点出现在CREPT(S134A/S166A)过表达的细胞中(图15B)。这些结果表明未降解的CREPT变体引起了DNA损伤。Undegraded CREPT mutants cause cell death and cell cycle arrest by remaining bound to MCM hexamers, so the inventors verified whether the CREPT-MCM complex induces genomic stress during the G1/S transition phase. Since genomic stress causes DNA damage, the inventors performed TUNEL staining experiments to examine DNA strand breaks. The results showed that overexpression of CREPT (S134A/S166A) resulted in significant TUNEL signals in WT cells and CREPT-deficient cells, while overexpression of WT protein and CREPT (S134E/S166E) showed negative signals (Fig. 15A). Notably, overexpression of CREPT (S134A/S166A) produced a much stronger TUNEL signal in CREPT-deficient cells than in WT cells (Fig. 15A, compare KO with Mock). Mutually Correspondingly, γH2AX foci were also observed to appear in CREPT (S134A/S166A) overexpressing cells (Fig. 15B). These results indicate that undegraded CREPT variants cause DNA damage.
为了确认该DNA损伤是否是由于未降解的CREPT变体引起的基因组胁迫,发明人检测了由复制蛋白A2(RPA2)形成的焦点,RPA2与单链DNA(ssDNA)结合并响应于复制压力而磷酸化。用针对总RPA2的抗体进行的IF染色显示,CREPT(S134A/S166A)在HeLa细胞中的过表达导致RPA2焦点增多,且蛋白水平增加(图7A,参见ctrl)。当用HU(一种dNTP合成抑制剂,其阻断DNA复制)处理细胞时,RPR2焦点大大增加(图7A,比较HU和ctrl)。进一步检查磷酸化的RPA2(p-RPA2)时观察到,CREPT(S134A/S166A)过表达的细胞中出现了阳性焦点,并且在HU处理下大大增加了(图7B)。这些结果表明未降解的CREPT蛋白变体导致了基因组胁迫。To confirm whether this DNA damage was due to genomic stress caused by undegraded CREPT variants, the inventors examined foci formed by replication protein A2 (RPA2), which binds to single-stranded DNA (ssDNA) and phosphorylates in response to replication stress. change. IF staining with an antibody against total RPA2 showed that overexpression of CREPT (S134A/S166A) in HeLa cells resulted in increased RPA2 foci and increased protein levels (Fig. 7A, see ctrl). When cells were treated with HU, an inhibitor of dNTP synthesis that blocks DNA replication, RPR2 foci were greatly increased (Fig. 7A, compare HU and ctrl). Upon further examination of phosphorylated RPA2 (p-RPA2), it was observed that positive foci appeared in cells overexpressing CREPT (S134A/S166A) and were greatly increased under HU treatment (Fig. 7B). These results indicate that undegraded CREPT protein variants contribute to genomic stress.
由于在表达CREPT(S134A/S166A)的细胞中MCM-DNA解离失败,我们调查了未降解的CREPT变体诱导的细胞死亡是否是由DNA复制中断的基因组胁迫引起的。为此检查了新合成的DNA纤维。在HU阻断后释放细胞,并允许CIdU并入。IF染色结果显示,当CREPT(S134A/S166A)表达了12小时时,CIdU标记的DNA纤维轨迹的长度缩短(图7C)。为了进一步证实这一结果,进行了双重IdU和CIdU标记实验。结果表明,与WT细胞相比,在CREPT(S134A/S166A)细胞中,IdU标记的纤维的长度减少了(图7D)。与此一致地,代表正常条件下复制速率的CIdU标记的纤维在CREPT(S134A/S166A)细胞中变短(图7D)。定量分析表明,与WT细胞相比,在CREPT(S134A/S166A)过表达的细胞中诱导了更多的复制停滞纤维(CIdU+IdU-轨迹)(图7D)。这些结果表明,未降解的CREPT变体可能通过影响停滞的复制叉而导致DNA合成缺陷。Since MCM-DNA dissociation failed in cells expressing CREPT(S134A/S166A), we investigated whether cell death induced by undegraded CREPT variants was caused by genomic stress with disruption of DNA replication. Newly synthesized DNA fibers were examined for this purpose. Cells were released after HU blockade and CIdU incorporation was allowed. IF staining results showed that when CREPT (S134A/S166A) was expressed for 12 hours, the length of CIdU-labeled DNA fiber tracks was shortened (Figure 7C). To further confirm this result, double IdU and CIdU labeling experiments were performed. The results showed that the length of IdU-labeled fibers was reduced in CREPT (S134A/S166A) cells compared with WT cells (Fig. 7D). Consistently, CIdU-labeled fibers, which represent replication rates under normal conditions, were shorter in CREPT (S134A/S166A) cells (Fig. 7D). Quantitative analysis showed that more replication-arrested fibers (CIdU + IdU - trajectories) were induced in CREPT(S134A/S166A)-overexpressing cells compared with WT cells (Fig. 7D). These results suggest that non-degraded CREPT variants may cause DNA synthesis defects by affecting stalled replication forks.
材料和方法Materials and methods
以上实施例中使用的材料和方法如下所述。The materials and methods used in the above examples are described below.
质粒和siRNAPlasmids and siRNA
HA-CREPT、Flag-CREPT、Myc-CREPT、Myc-CREPT-CID、Myc-CREPT-CCT、GSTCREPT、GFP-P15RS和Myc-P15RS质粒由发明人的实验室自己构建。Flag-MCM2质粒由孔道春博士(北京大学生命科学学院)馈赠。pRK5-HA-UBI(#17608)、pRK5-HA-UBI-K11(#22901)、pRK5-HA-UBI-K48(#17605)、pRK5-HA-UBI-K63(#17606)和pSpCas9(BB)-2AGFP(PX458,#48138)购自Addgene。Flag-SKP1、 Flag-SKP2、Flag-CUL1、Flag-MCM5和Flag-MCM7由cDNA产生。CREPT突变的质粒通过定点诱变(Muta-directTM,SBS Genetech)产生。SKP2和CUL1 siRNA双链体由Lipofectamine RNAi MAX(Invitrogen)转染,寡核苷酸序列分别为AAUCUAAGCCUGGAAGGCCUGdTdT和UAGACAUUGGGUUCGCCGUdTdT。HA-CREPT, Flag-CREPT, Myc-CREPT, Myc-CREPT-CID, Myc-CREPT-CCT, GSTCREPT, GFP-P15RS and Myc-P15RS plasmids were constructed by the inventor's laboratory. Flag-MCM2 plasmid was a gift from Dr. Kong Daochun (School of Life Sciences, Peking University). pRK5-HA-UBI(#17608), pRK5-HA-UBI-K11(#22901), pRK5-HA-UBI-K48(#17605), pRK5-HA-UBI-K63(#17606) and pSpCas9(BB) -2AGFP (PX458, #48138) was purchased from Addgene. Flag-SKP1、 Flag-SKP2, Flag-CUL1, Flag-MCM5 and Flag-MCM7 were generated from cDNA. CREPT mutated plasmids were generated by site-directed mutagenesis (Muta-direct , SBS Genetech). SKP2 and CUL1 siRNA duplexes were transfected by Lipofectamine RNAi MAX (Invitrogen), and the oligonucleotide sequences were AAUCUAAGCCUGGAAGGCCUGdTdT and UAGACAUUGGGUUCGCCGUdTdT, respectively.
CRISPR-Cas9敲入细胞系CRISPR-Cas9 knock-in cell lines
HeLa细胞用于构建CRISPR-Cas9敲入细胞系。根据张锋的实验室指导(//crispr.mit.edu/)设计和优化了短向导RNA(sgRNA)寡核苷酸。sgRNA序列为CTCCTTCTCTGAGTCGGCGC。退火的sgRNA和BbsI消化的Px458载体通过溶液I(Takara)连接以构建Cas9 DNA剪切质粒。另一方面,将GFP的编码序列克隆并连接到PCDNA 3.1-HA载体上以构建GFP转录质粒。HeLa细胞与Cas9 DNA剪切和GFP转录质粒共转染。流式细胞仪对GFP阳性HeLa细胞进行分选,然后将单个细胞接种在96孔板中以选择DNA重组克隆。一周后,利用基因组插入的GFP表达来筛选细胞。HeLa cells were used to construct CRISPR-Cas9 knock-in cell lines. Short guide RNA (sgRNA) oligonucleotides were designed and optimized according to Zhang Feng's laboratory guidance (http://crispr.mit.edu/). The sgRNA sequence is CTCCTTCTCTGAGTCGGCGC. Annealed sgRNA and BbsI-digested Px458 vector were ligated by solution I (Takara) to construct Cas9 DNA shearing plasmid. On the other hand, the coding sequence of GFP was cloned and ligated into the PCDNA 3.1-HA vector to construct a GFP transcription plasmid. HeLa cells were co-transfected with Cas9 DNA cleavage and GFP transcription plasmids. GFP-positive HeLa cells were sorted by flow cytometry, and individual cells were seeded in 96-well plates to select DNA recombinant clones. One week later, cells were selected for expression of the genomically inserted GFP.
细胞周期同步cell cycle synchronization
通过双胸苷阻滞(DTB)将细胞同步在G1/S期。细胞用2mM胸苷处理至少18小时,在新鲜培养基中释放8小时,然后用2mM胸苷再处理至少16小时。通过胸苷-诺考达唑阻滞使细胞同步在G2/M期。细胞用2mM胸苷处理至少24小时,释放3小时,然后用340nM诺考达唑处理至少16小时。在所示时间收获细胞。通过流式细胞仪分析验证收获细胞的细胞周期阶段。对于MG132处理,在收获前4小时将MG132添加到DLD1细胞中。Cells were synchronized in G1/S phase by double thymidine block (DTB). Cells were treated with 2mM thymidine for at least 18 hours, released in fresh medium for 8 hours, and then treated with 2mM thymidine for at least another 16 hours. Cells were synchronized in G2/M phase by thymidine-nocodazole block. Cells were treated with 2mM thymidine for at least 24 hours, released for 3 hours, and then treated with 340 nM nocodazole for at least 16 hours. Cells were harvested at the times indicated. Verify the cell cycle stage of harvested cells by flow cytometric analysis. For MG132 treatment, MG132 was added to DLD1 cells 4 hours before harvest.
体外蛋白测定In vitro protein assay
由于SKP2蛋白在原核表达系统中不稳定。利用pET22b-SKP1、pET30A-SKP2和GST-CREPT蛋白进行体外蛋白相互作用测定。免疫沉淀测定验证了GST-CREPT和pET30A-SKP2之间的相互作用。Because SKP2 protein is unstable in prokaryotic expression system. In vitro protein interaction assay was performed using pET22b-SKP1, pET30A-SKP2 and GST-CREPT proteins. Immunoprecipitation assay verified the interaction between GST-CREPT and pET30A-SKP2.
利用纯化的GST、GST-CREPT(野生型)或GST-CREPT(S134A/S166A)进行体外激酶测定,在30℃下将蛋白在激酶缓冲液(10mM HEPES(pH7.5),50mM NaCl,2mM MgCl2,1mM二硫苏糖醇,1mM EGTA和0.1mM ATP)中与Myc-Cyclin E/Myc-CDK2蛋白一起温育30分钟。利用SDS上样缓冲液停止反应。通过蛋白质印迹检测CREPT的磷酸化。 Purified GST, GST-CREPT (wild type) or GST-CREPT (S134A/S166A) were used for in vitro kinase assay. The protein was dissolved in kinase buffer (10mM HEPES (pH7.5), 50mM NaCl, 2mM MgCl) at 30°C. 2 , 1mM dithiothreitol, 1mM EGTA and 0.1mM ATP) and incubated with Myc-Cyclin E/Myc-CDK2 protein for 30 minutes. Stop the reaction using SDS loading buffer. Phosphorylation of CREPT was detected by Western blotting.
DNA纤维分析DNA fiber analysis
DNA纤维实验如之前所述进行(Genois et al.,2021)。简言之,首先用50mM CldU标记DLD1细胞,用PBS洗涤两次并用250mM IdU标记。收获细胞并悬浮在冷PBS中,至浓度为1~1.5×106个细胞/ml,然后将3ul细胞液与7ul铺展缓冲液(0.5%SDS、200mM Tris-HCl pH7.4、50mM EDTA)混合,并涂抹在硅烷化的载玻片上。将载玻片倾斜成30-60°角以铺展纤维,并在室温下放置15分钟。DNA纤维在甲醇:乙酸(3:1)中固定20分钟。干燥后,载玻片在4℃下保存过夜。DNA纤维在2.5M HCl中变性30分钟,并用3%BSA封闭60分钟。CldU和IdU用大鼠抗BrdU和小鼠抗BrdU在室温下检测2小时,然后用Alexa488抗小鼠和Cy3抗大鼠二抗在室温下结合1小时。载玻片用Prolong Gold Antifade Reagent固定。在Olympus FV3000共聚焦显微镜上使用60X物镜对纤维进行成像。DNA fiber experiments were performed as previously described (Genois et al., 2021). Briefly, DLD1 cells were first labeled with 50mM CldU, washed twice with PBS and labeled with 250mM IdU. Harvest the cells and suspend them in cold PBS to a concentration of 1 to 1.5 × 10 cells/ml, then mix 3ul of cell solution with 7ul of spreading buffer (0.5% SDS, 200mM Tris-HCl pH7.4, 50mM EDTA) , and spread on silanized glass slides. Tilt the slide at a 30-60° angle to spread the fibers and leave at room temperature for 15 minutes. DNA fibers were fixed in methanol:acetic acid (3:1) for 20 minutes. After drying, the slides were stored at 4°C overnight. DNA fibers were denatured in 2.5 M HCl for 30 min and blocked with 3% BSA for 60 min. CldU and IdU were detected with rat anti-BrdU and mouse anti-BrdU for 2 hours at room temperature, followed by conjugation with Alexa488 anti-mouse and Cy3 anti-rat secondary antibodies for 1 hour at room temperature. Slides were fixed with Prolong Gold Antifade Reagent. Fibers were imaged on an Olympus FV3000 confocal microscope using a 60X objective.
免疫荧光测定Immunofluorescence assay
在预提取方法中,为了提取可溶性蛋白质,首先在冰上用1mL透化缓冲液(含有0.2%TritonX-100、20mM HEPES pH=7.4、100mM NaCl和300mM蔗糖)处理活细胞5分钟。除去透化缓冲液后,将细胞用2%多聚甲醛在室温下固定10分钟。在直接固定法中,细胞首先在室温下用2%多聚甲醛固定10分钟。然后,用PBS中的0.3%Triton X100透化细胞15分钟,并用PBST中的10%BSA封闭。固定的细胞与指定的一抗在室温下温育2小时或在4℃下温育过夜。在与荧光二抗温育并在Prolong Gold中封片后,在Olympus FV3000共聚焦显微镜上用60X物镜对细胞进行成像。In the pre-extraction method, to extract soluble proteins, live cells were first treated with 1 mL of permeabilization buffer (containing 0.2% TritonX-100, 20mM HEPES pH=7.4, 100mM NaCl, and 300mM sucrose) for 5 minutes on ice. After removing the permeabilization buffer, cells were fixed with 2% paraformaldehyde for 10 min at room temperature. In the direct fixation method, cells are first fixed with 2% paraformaldehyde for 10 min at room temperature. Then, cells were permeabilized with 0.3% Triton X100 in PBS for 15 min and blocked with 10% BSA in PBST. Fixed cells were incubated with the indicated primary antibodies for 2 h at room temperature or overnight at 4°C. After incubation with fluorescent secondary antibodies and mounting in Prolong Gold, cells were imaged on an Olympus FV3000 confocal microscope using a 60X objective.
实施例7.CREPT磷酸化泛素化抑制剂的筛选Example 7. Screening of CREPT phosphorylation ubiquitination inhibitors
7.1作为潜在CREPT磷酸化泛素化抑制剂的小分子化合物的预测7.1 Prediction of small molecule compounds as potential CREPT phosphorylation ubiquitination inhibitors
利用预测工具SwissTargetPrediction(http://www.swisstargetprediction.ch/)和SEA(Similarity ensemble approach;https://sea.bkslab.org/)共同预测CREPT的小分子磷酸化抑制剂,得到并合成了5种候选小分子化合物#1至#5。由于CREPT的磷酸化是其泛素化的前提,因此这5种候选小分子化合物#1至#5是潜在的CREPT磷酸化泛素化抑制剂。The prediction tool SwissTargetPrediction (http://www.swisstargetprediction.ch/) and SEA (Similarity ensemble approach; https://sea.bkslab.org/) were used to jointly predict the small molecule phosphorylation inhibitor of CREPT, and 5 was obtained and synthesized. candidate small molecule compounds #1 to #5. Since the phosphorylation of CREPT is a prerequisite for its ubiquitination, these five candidate small molecule compounds #1 to #5 are potential inhibitors of CREPT phosphorylation and ubiquitination.
7.2候选小分子化合物对CREPT泛素化的影响7.2 Effect of candidate small molecule compounds on CREPT ubiquitination
1)将293T细胞分到6cm的培养皿中,37℃培养24小时。转染图16中指定的质粒(HA-Ub和/或Myc-CREPT)至细胞中,5小时后换入新鲜培养基。 1) Divide 293T cells into 6cm culture dishes and culture them at 37°C for 24 hours. The plasmids specified in Figure 16 (HA-Ub and/or Myc-CREPT) were transfected into the cells and replaced with fresh medium after 5 hours.
2)转染24小时后使用1ml RIPA裂解液收集细胞,4℃裂解1小时。13000rpm 4℃离心10分钟。取800μl上清加入50μl protein plus beads和5μl抗myc抗体,标记为IP样品,4℃旋转温育过夜。取50μl上清加入等量的2x上样缓冲液,标记为裂解液样品。2) 24 hours after transfection, use 1ml RIPA lysis buffer to collect cells and lyse them at 4°C for 1 hour. Centrifuge at 13000rpm and 4℃ for 10 minutes. Take 800μl supernatant, add 50μl protein plus beads and 5μl anti-myc antibody, label it as IP sample, and incubate overnight at 4°C with rotation. Add 50 μl of the supernatant to an equal volume of 2x loading buffer and label it as a lysate sample.
3)IP样品使用细胞裂解缓冲液离心洗脱4次,每次10分钟。洗脱后的IP样品加入50μl的2x上样缓冲液。IP与裂解液样品100℃煮10分钟。SDS PAGE跑胶检测。3) IP samples were centrifuged and eluted using cell lysis buffer 4 times, 10 minutes each time. Add 50 μl of 2x loading buffer to the eluted IP sample. Boil IP and lysate samples at 100°C for 10 minutes. SDS PAGE gel running test.
结果如图16所示,未经候选抑制剂处理的CREPT野生型蛋白能够被泛素(Ub)泛素化(图16的第三道),阴性对照CREPT的突变体SA对照组检测不到泛素化。在添加候选化合物#1至#5的条带中,#4的泛素化明显减弱(相对于未经处理的野生型蛋白的泛素化水平下降了约20%)。The results are shown in Figure 16. The CREPT wild-type protein without candidate inhibitor treatment can be ubiquitinated by ubiquitin (Ub) (the third lane of Figure 16). The negative control CREPT mutant SA control group cannot detect ubiquitin. Vegetarianization. In the bands to which candidate compounds #1 to #5 were added, ubiquitination of #4 was significantly attenuated (approximately 20% decrease in ubiquitination level relative to the untreated wild-type protein).
上述结果证明化合物#4是有效的CREPT磷酸化泛素化抑制剂。化合物#4的结构式如下:
The above results demonstrate that compound #4 is a potent inhibitor of CREPT phosphorylation and ubiquitination. The structural formula of compound #4 is as follows:
7.3候选小分子化合物对细胞增殖的影响7.3 Effect of candidate small molecule compounds on cell proliferation
1)取对数生长期的DLD1(人结直肠腺癌上皮细胞)或MGC803(人胃癌细胞),用0.25%胰蛋白酶消化并轻轻吹打,使之成为单细胞,对活细胞进行计数,用含10%胎牛血清的DMEM培养液调整细胞密度至1×104细胞/L。1) Take DLD1 (human colorectal adenocarcinoma epithelial cells) or MGC803 (human gastric cancer cells) in the logarithmic growth phase, digest it with 0.25% trypsin and pipet gently to turn it into single cells, count the viable cells, and use The cell density was adjusted to 1×10 4 cells/L in DMEM culture medium containing 10% fetal calf serum.
2)按1:1比例加10mL培养液和10mL细胞稀释液混合后,取0.2mL混合液加入96孔板的每个孔中,共有3个重复孔。置于37℃、5%CO2温箱中培养12小时。2) After adding 10 mL of culture medium and 10 mL of cell diluent in a 1:1 ratio, add 0.2 mL of the mixed solution to each well of the 96-well plate, with a total of 3 replicate wells. Place in a 37°C, 5% CO2 incubator for 12 hours.
3)取上述5种候选小分子化合物#1至#5并用DMSO溶解,每种化合物的初筛浓度为10μM(DLD1细胞)或5μM(MGC803细胞)。每种化合物做3个重复;每种化合物都是以10μM(DLD1细胞)或5μM(MGC803细胞)的浓度温育3天,然后利用CCK测定细胞增殖情况。测量前,每孔换成混匀的10μl CCK-8溶液和90μl完全培养基(加了相应量CCK-8溶液及细胞培养液的孔作为空白对照)。37℃温育3小时。测 定450nm波长下的吸光度。计算和统计结果并画图,结果如图17的A(DLD1细胞)和B(MGC803细胞)所示。3) Take the above five candidate small molecule compounds #1 to #5 and dissolve them in DMSO. The initial screening concentration of each compound is 10 μM (DLD1 cells) or 5 μM (MGC803 cells). Each compound was run in triplicate; each compound was incubated for 3 days at a concentration of 10 μM (DLD1 cells) or 5 μM (MGC803 cells), and cell proliferation was measured using CCK. Before measurement, each well was replaced with 10 μl of mixed CCK-8 solution and 90 μl of complete culture medium (the wells with corresponding amounts of CCK-8 solution and cell culture medium were added as blank controls). Incubate at 37°C for 3 hours. Measurement Determine the absorbance at a wavelength of 450nm. The results were calculated and statistically drawn and graphed. The results are shown in Figure 17 A (DLD1 cells) and B (MGC803 cells).
可以看出,作为CREPT磷酸化泛素化抑制剂的#4化合物明显抑制了细胞增殖,这说明测试浓度的化合物#4在一定程度上抑制了CREPT的降解,导致部分细胞凋亡,该结果与实施例2-4的结果一致。It can be seen that compound #4, which is an inhibitor of CREPT phosphorylation and ubiquitination, significantly inhibited cell proliferation, which shows that the test concentration of compound #4 inhibited the degradation of CREPT to a certain extent, leading to apoptosis in some cells. This result is consistent with The results of Examples 2-4 are consistent.
以上描述了本发明的技术思想和具体实施方式,但应当理解,上述具体实施方式不以任何方式限制本发明的范围。本领域技术人员可以理解,在不脱离本发明的实质的情况下,可以对具体实施方式中所显示的发明进行多种修改和/或变化,修改和/或变化后的实施方式也涵盖在本发明的范围内。因此,本发明的实施方式仅是说明性的且非限制性的。The technical ideas and specific implementations of the present invention have been described above, but it should be understood that the above specific implementations do not limit the scope of the present invention in any way. Those skilled in the art can understand that various modifications and/or changes can be made to the invention shown in the specific embodiments without departing from the essence of the invention, and the modified and/or changed embodiments are also covered by this invention. within the scope of the invention. Accordingly, the embodiments of the present invention are illustrative only and not restrictive.
参考文献references
Anzalone,A.V.,Randolph,P.B.,Davis,J.R.et al.(2019)Search-and-replace genome editing without double-strand breaks or donor DNA.Nature 576,149–157.Anzalone,A.V.,Randolph,P.B.,Davis,J.R.et al.(2019)Search-and-replace genome editing without double-strand breaks or donor DNA.Nature 576,149–157.
Berthet,C.,Aleem,E.,Coppola,V.,Tessarollo,L.,and Kaldis,P.(2003).Cdk2 knockout mice are viable.Curr Biol 13,1775-1785.Berthet,C.,Aleem,E.,Coppola,V.,Tessarollo,L.,and Kaldis,P.(2003).Cdk2 knockout mice are viable.Curr Biol 13,1775-1785.
Chan,C.H.,Morrow,J.K.,Li,C.F.,Gao,Y.,Jin,G.,Moten,A.,Stagg,L.J.,Ladbury,J.E.,Cai,Z.,Xu,D.,et al.(2013).Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression.Cell 154,556-568.Chan, C.H., Morrow, J.K., Li, C.F., Gao, Y., Jin, G., Moten, A., Stagg, L.J., Ladbury, J.E., Cai, Z., Xu, D., et al. (2013 ).Pharmacological inactivation of Skp2 SCF ubiquitin ligase restricts cancer stem cell traits and cancer progression.Cell 154,556-568.
Genois,M.M.,Gagne,J.P.,Yasuhara,T.,Jackson,J.,Saxena,S.,Langelier,M.F.,Ahel,I.,Bedford,M.T.,Pascal,J.M.,Vindigni,A.,et al.(2021).CARM1 regulates replication fork speed and stress response by stimulating PARP1.Molecular Cell 81.Genois,M.M.,Gagne,J.P.,Yasuhara,T.,Jackson,J.,Saxena,S.,Langelier,M.F.,Ahel,I.,Bedford,M.T.,Pascal,J.M.,Vindigni,A.,et al.(2021 ).CARM1 regulates replication fork speed and stress response by stimulating PARP1.Molecular Cell 81.
Li,M.D.,Ma,D.H.,and Chang,Z.J.(2021).Current understanding of CREPT and p15RS,carboxyterminal domain(CTD)-interacting proteins,in human cancers.Oncogene 40,705-716.Li,M.D.,Ma,D.H.,and Chang,Z.J.(2021).Current understanding of CREPT and p15RS,carboxyterminal domain(CTD)-interacting proteins,in human cancers.Oncogene 40,705-716.
Lin,H.K.,Chen,Z.,Wang,G.,Nardella,C.,Lee,S.W.,Chan,C.H.,Yang,W.L.,Wang,J.,Egia,A.,Nakayama,K.I.,et al.(2010).Skp2 targeting suppresses tumorigenesis by Arf-p53-independent cellular senescence.Nature 464,374-379.Lin,H.K.,Chen,Z.,Wang,G.,Nardella,C.,Lee,S.W.,Chan,C.H.,Yang,W.L.,Wang,J.,Egia,A.,Nakayama,K.I.,et al.(2010 ).Skp2 targeting suppresses tumorigenesis by Arf-p53-independent cellular senescence.Nature 464,374-379.
Lu,D.,Wu,Y.,Wang,Y.,Ren,F.,Wang,D.,Su,F.,Zhang,Y.,Yang,X.,Jin,G.,Hao,X.,et al.(2012).CREPT accelerates tumorigenesis by regulating the transcription of  cell-cycle-related genes.Cancer Cell 21,92-104.Lu,D.,Wu,Y.,Wang,Y.,Ren,F.,Wang,D.,Su,F.,Zhang,Y.,Yang,X.,Jin,G.,Hao,X., et al.(2012).CREPT accelerates tumorigenesis by regulating the transcription of cell-cycle-related genes. Cancer Cell 21,92-104.
Maiani,E.,Milletti,G.,Nazio,F.,Holdgaard,S.G.,Bartkova,J.,Rizza,S.,Cianfanelli,V.,Lorente,M.,Simoneschi,D.,Di Marco,M.,et al.(2021).AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity.Nature 592,799-+.Maiani,E.,Milletti,G.,Nazio,F.,Holdgaard,S.G.,Bartkova,J.,Rizza,S.,Cianfanelli,V.,Lorente,M.,Simoneschi,D.,Di Marco,M., et al.(2021).AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity.Nature 592,799-+.
Schulman,B.A.,Carrano,A.C.,Jeffrey,P.D.,Bowen,Z.,Kinnucan,E.R.E.,Finnin,M.S.,Elledge,S.J.,Harper,J.W.,Pagano,M.,and Pavietich,N.P.(2000).Insights into SCF ubiquitin ligases from the structure of the Skp1-Skp2 complex.Nature 408,381-386.Schulman, B.A., Carrano, A.C., Jeffrey, P.D., Bowen, Z., Kinnucan, E.R.E., Finnin, M.S., Elledge, S.J., Harper, J.W., Pagano, M., and Pavietich, N.P. (2000). Insights into SCF ubiquitin ligases from the structure of the Skp1-Skp2 complex.Nature 408,381-386.
Simoneschi,D.,Rona,G.,Zhou,N.,Jeong,Y.T.,Jiang,S.W.,Milletti,G.,Arbini,A.A.,O'Sullivan,A.,Wang,A.A.,Nithikasem,S.,et al.(2021).CRL4(AMBRA1)is a master regulator of D-type cyclins.Nature 592,789-+.Simoneschi,D.,Rona,G.,Zhou,N.,Jeong,Y.T.,Jiang,S.W.,Milletti,G.,Arbini,A.A.,O'Sullivan,A.,Wang,A.A.,Nithikasem,S.,et al .(2021).CRL4(AMBRA1)is a master regulator of D-type cyclins.Nature 592,789-+.
Tadesse,S.,Caldon,E.C.,Tilley,W.,and Wang,S.(2019).Cyclin-Dependent Kinase 2 Inhibitors in Cancer Therapy:An Update.Journal of Medicinal Chemistry 62,4233-4251.Tadesse, S., Caldon, E.C., Tilley, W., and Wang, S. (2019). Cyclin-Dependent Kinase 2 Inhibitors in Cancer Therapy: An Update. Journal of Medicinal Chemistry 62, 4233-4251.
Zhu,L.(2010).Skp2 knockout reduces cell proliferation and mouse body size:and prevents cancer?Cell Res 20,605-607. Zhu,L.(2010).Skp2 knockout reduces cell proliferation and mouse body size:and prevents cancer? Cell Res 20,605-607.

Claims (41)

  1. 一种蛋白,其通过对SEQ ID No:4的134和166位点进行磷酸化失活修饰而得到,所述磷酸化失活修饰使得当所述蛋白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述蛋白保持非泛素化状态,从而使所述蛋白在所述真核细胞中不被降解,进而导致细胞周期阻滞和细胞凋亡。A protein obtained by phosphorylation inactivation modification at positions 134 and 166 of SEQ ID No: 4. The phosphorylation inactivation modification enables the true expression of the protein when it is at the end of G1 phase or the G1/S transition phase. In nuclear cells, the 134 and 166 positions remain in a non-phosphorylated state and the protein remains in a non-ubiquitinated state, thereby preventing the protein from being degraded in the eukaryotic cells, thereby causing cell cycle arrest. and apoptosis.
  2. 如权利要求1所述的蛋白,其中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。The protein of claim 1, wherein the eukaryotic cell is a human, yeast, mouse, dog, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell, preferably a human cell, Human cancer cells are more preferred.
  3. 如权利要求1所述的蛋白,其中,所述134和166位点上的所述磷酸化失活修饰是氨基酸突变和/或化学修饰。The protein of claim 1, wherein the phosphorylation inactivating modifications at positions 134 and 166 are amino acid mutations and/or chemical modifications.
  4. 如权利要求1所述的蛋白,其中,所述134和166位点上的所述磷酸化失活修饰是将丝氨酸(S)突变为丙氨酸(A)。The protein of claim 1, wherein the phosphorylation-inactivating modification at the 134 and 166 positions is to mutate serine (S) to alanine (A).
  5. 一种蛋白,其与权利要求1至4中任一项所述的蛋白具有(i)90%以上的序列同一性和(ii)相同的134和166位点上的磷酸化失活修饰。A protein having (i) more than 90% sequence identity and (ii) the same phosphorylation inactivation modifications at positions 134 and 166 as the protein according to any one of claims 1 to 4.
  6. 在权利要求1至5中任一项所述的蛋白的N端和/或C端连接有标签序列或引导序列的蛋白。A protein with a tag sequence or a guide sequence connected to the N-terminus and/or C-terminus of the protein according to any one of claims 1 to 5.
  7. 编码权利要求4所述的蛋白的核酸。Nucleic acid encoding the protein of claim 4.
  8. 包含权利要求7所述的核酸的载体。A vector comprising the nucleic acid of claim 7.
  9. 包含权利要求8所述的载体的细胞。A cell comprising the vector of claim 8.
  10. 权利要求1-6中任一项所述的蛋白、权利要求7所述的核酸或权利要求8所述的载体在制备抑制真核细胞增殖、抑制真核细胞的DNA复制、调控真核细胞的细胞周期或杀灭真核细胞的试剂中的应用。The protein according to any one of claims 1 to 6, the nucleic acid according to claim 7 or the vector according to claim 8 can be used in the preparation of inhibiting eukaryotic cell proliferation, inhibiting DNA replication of eukaryotic cells and regulating eukaryotic cells. Applications in the cell cycle or as reagents to kill eukaryotic cells.
  11. 权利要求1-6中任一项所述的蛋白、权利要求7所述的核酸或权利要求8所述的载体在制备抗癌药中的应用。Use of the protein according to any one of claims 1 to 6, the nucleic acid according to claim 7 or the vector according to claim 8 in the preparation of anti-cancer drugs.
  12. 一种治疗癌症的方法,所述方法包括:A method of treating cancer, the method comprising:
    i)向受试者施用有效量的权利要求1-6中任一项所述的蛋白、权利要求7所述的核酸或权利要求8所述的载体;或者i) administering to a subject an effective amount of the protein of any one of claims 1-6, the nucleic acid of claim 7, or the vector of claim 8; or
    ii)利用基于CRISPR/Cas9的基因编辑技术编辑受试者癌细胞基因组中的CRPET 基因,以使所述癌细胞表达权利要求4所述的蛋白。ii) Use CRISPR/Cas9-based gene editing technology to edit CRPET in the genome of cancer cells of subjects Gene, so that the cancer cell expresses the protein of claim 4.
  13. 如权利要求12所述的方法,其中,所述受试者是哺乳动物,优选是人。The method of claim 12, wherein the subject is a mammal, preferably a human.
  14. 如权利要求12所述的方法,其中,所述i)还包括:在向受试者施用有效量的所述蛋白、核酸或载体之前、过程中或之后,降低或消除受试者的癌细胞中的野生型CREPT的表达。The method of claim 12, wherein i) further comprises reducing or eliminating cancer cells in the subject before, during or after administering an effective amount of the protein, nucleic acid or vector to the subject Expression of wild-type CREPT.
  15. 源自非人真核生物的人CREPT的同源蛋白,其在对应于SEQ ID No:4的134和166位点的同源性位点处具有磷酸化失活修饰,所述磷酸化失活修饰使得当所述同源蛋白位于所述真核生物的G1末期或G1/S转换期的细胞中时,所述对应于SEQ ID No:4的134和166位点的同源性位点保持非磷酸化状态且所述同源蛋白保持非泛素化状态,从而使所述同源蛋白在所述细胞中不会被降解,进而导致细胞周期阻滞和细胞凋亡。Homologous proteins of human CREPT derived from non-human eukaryotes, which have phosphorylation-inactivating modifications at homology sites corresponding to positions 134 and 166 of SEQ ID No: 4, said phosphorylation-inactivating The modification is such that when the homologous protein is located in cells in the G1 end phase or G1/S transition phase of the eukaryotic organism, the homology sites corresponding to positions 134 and 166 of SEQ ID No: 4 are maintained. Non-phosphorylated state and the homologous protein remains in a non-ubiquitinated state, so that the homologous protein will not be degraded in the cell, thereby leading to cell cycle arrest and apoptosis.
  16. 如权利要求15所述的同源蛋白,其中,所述真核生物是酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥。The homologous protein of claim 15, wherein the eukaryotic organism is yeast, mouse, dog, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana.
  17. 如权利要求15所述的同源蛋白,其中,所述对应于SEQ ID No:4的134和166位点的同源性位点上的所述磷酸化失活修饰是氨基酸突变和/或化学修饰。The homologous protein of claim 15, wherein the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 is an amino acid mutation and/or chemical Grooming.
  18. 如权利要求15所述的同源蛋白,其中,所述对应于SEQ ID No:4的134和166位点的同源性位点上的所述磷酸化失活修饰是将丝氨酸突变为丙氨酸。The homologous protein of claim 15, wherein the phosphorylation inactivation modification on the homology site corresponding to positions 134 and 166 of SEQ ID No: 4 is to mutate serine to alanine acid.
  19. 如权利要求15所述的同源蛋白,其氨基酸序列为SEQ ID No:6。The homologous protein as claimed in claim 15, its amino acid sequence is SEQ ID No: 6.
  20. 一种蛋白,其选自:A protein selected from:
    i)在权利要求15至19中任一项所述的蛋白的N端和/或C端连接有标签序列或引导序列的蛋白;或i) A protein with a tag sequence or guide sequence connected to the N-terminus and/or C-terminus of the protein according to any one of claims 15 to 19; or
    ii)与权利要求15至19中任一项所述的蛋白具有90%以上的序列同一性且在所述同源性位点上具有相同的磷酸化失活修饰的蛋白。ii) A protein that has more than 90% sequence identity with the protein of any one of claims 15 to 19 and has the same phosphorylation inactivation modification at the homology site.
  21. 编码权利要求18或19所述的同源蛋白的核酸。Nucleic acid encoding the homologous protein of claim 18 or 19.
  22. 包含权利要求21所述的核酸的载体。A vector comprising the nucleic acid of claim 21.
  23. 包含权利要求22所述的载体的细胞。A cell comprising the vector of claim 22.
  24. 一种鉴别处于G1末期或G1/S转换期的真核细胞的方法,所述方法包括:A method for identifying eukaryotic cells in late G1 phase or G1/S transition phase, the method comprising:
    1)制备能够内源性表达带可检测标记的人CREPT蛋白或其在非人真核生物中的同源蛋白的真核细胞,并在允许细胞周期进行的条件下培养该真核细胞; 1) Prepare eukaryotic cells capable of endogenously expressing a detectably labeled human CREPT protein or its homologous protein in non-human eukaryotes, and culture the eukaryotic cells under conditions that allow the cell cycle to proceed;
    2)利用所述可检测标记观察或测量所述真核细胞中的所述人CREPT蛋白或所述同源蛋白的表达水平;2) using the detectable marker to observe or measure the expression level of the human CREPT protein or the homologous protein in the eukaryotic cell;
    3)将所述人CREPT蛋白或所述同源蛋白的表达水平最低的细胞鉴别为处于G1末期或G1/S转换期的细胞;3) Identify cells with the lowest expression level of the human CREPT protein or the homologous protein as cells in the late G1 phase or the G1/S transition phase;
    其中,所述人CREPT蛋白的序列为SEQ ID No:4。Wherein, the sequence of the human CREPT protein is SEQ ID No: 4.
  25. 如权利要求24所述的方法,其中,所述可检测标记是同位素标记、荧光标记或量子点标记或者是能够进一步与同位素标记、荧光标记或量子点标记结合的标记,优选为GFP。The method of claim 24, wherein the detectable label is an isotope label, a fluorescent label or a quantum dot label or a label that can further be combined with an isotope label, a fluorescent label or a quantum dot label, preferably GFP.
  26. 如权利要求24所述的方法,其中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞。The method of claim 24, wherein the eukaryotic cell is a human, yeast, mouse, canine, cat, chicken, toad, zebrafish, Drosophila, nematode or Arabidopsis thaliana cell.
  27. 一种筛选CREPT蛋白的非磷酸化非泛素化修饰剂的方法,其中,所述修饰剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在真核细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列为SEQ ID No:4,所述方法包括:A method for screening non-phosphorylation and non-ubiquitination modifiers of CREPT protein, wherein the modifier keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby allowing the CREPT protein to function in eukaryotic cells It maintains a non-ubiquitinated state and will not be degraded; the amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes:
    i)将模拟非磷酸化状态的候选修饰剂加入表达CREPT蛋白的被同步至G1期的真核细胞中,然后释放并培养所述真核细胞,并在所述真核细胞存活时检查所述CREPT蛋白的S134和S166位点的磷酸化水平;i) Add candidate modifiers that mimic the non-phosphorylated state to eukaryotic cells expressing CREPT protein that are synchronized to G1 phase, then release and culture the eukaryotic cells, and examine the eukaryotic cells while they are alive Phosphorylation levels of S134 and S166 of CREPT protein;
    或者or
    ii)在体外将模拟非磷酸化状态的候选修饰剂与CREPT蛋白温育,并在Cyclin E/CDK2激酶的催化条件下检查所述CREPT蛋白的S134和S166位点的磷酸化水平,ii) incubate the candidate modifier that simulates the non-phosphorylated state with the CREPT protein in vitro, and check the phosphorylation levels of the S134 and S166 sites of the CREPT protein under the catalytic conditions of Cyclin E/CDK2 kinase,
    如果经步骤i)或ii)处理的CREPT蛋白的S134和S166位点的磷酸化水平相对于未经处理的对照下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述候选修饰剂筛选为CREPT蛋白的非磷酸化非泛素化修饰剂。If the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The candidate modifiers are screened as non-phosphorylated, non-ubiquitinated modifiers of the CREPT protein.
  28. 一种鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列为SEQ ID No:4,所述方法包括:A method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby maintaining the non-ubiquitin of the CREPT protein in cells. The amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes:
    i)将待鉴定的物质加入表达CREPT蛋白的被同步至G1期的真核细胞中,然后释放并培养所述真核细胞,并在所述真核细胞存活时检查所述CREPT蛋白的S134 和S166位点的磷酸化水平;或者i) Add the substance to be identified to eukaryotic cells expressing CREPT protein that are synchronized to the G1 phase, then release and culture the eukaryotic cells, and examine the S134 of the CREPT protein while the eukaryotic cells are alive and the phosphorylation level of S166; or
    ii)在体外将待鉴定的物质与CREPT蛋白温育,并在Cyclin E/CDK2激酶的催化条件下检查所述CREPT蛋白的S134和S166位点的磷酸化水平;ii) Incubate the substance to be identified with the CREPT protein in vitro, and check the phosphorylation levels of the S134 and S166 sites of the CREPT protein under the catalytic conditions of Cyclin E/CDK2 kinase;
    如果经步骤i)或ii)处理的CREPT蛋白的S134和S166位点的磷酸化水平相对于未经处理的对照下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述物质鉴定为CREPT蛋白的磷酸化抑制剂,否则所述物质不是CREPT蛋白的磷酸化抑制剂。If the phosphorylation levels of the S134 and S166 sites of the CREPT protein treated in step i) or ii) decrease relative to the untreated control, for example, by more than 10%, more than 20%, more than 30% or more than 40%, then The substance is identified as a phosphorylation inhibitor of CREPT protein, which is otherwise not a phosphorylation inhibitor of CREPT protein.
  29. 如权利要求27或28所述的方法,其中,检查所述CREPT蛋白的S134和S166位点的磷酸化水平的步骤采用质谱法或免疫沉淀法来进行。The method of claim 27 or 28, wherein the step of checking the phosphorylation levels of S134 and S166 sites of the CREPT protein is performed using mass spectrometry or immunoprecipitation.
  30. 如权利要求29所述的方法,其中,所述免疫沉淀法包括:使用识别CREPT蛋白的S134和S166位点的磷酸化的抗磷酸化抗体来进行免疫沉淀。The method of claim 29, wherein the immunoprecipitation method includes performing immunoprecipitation using an anti-phosphorylated antibody that recognizes the phosphorylation of S134 and S166 sites of the CREPT protein.
  31. 一种鉴定物质是否为CREPT蛋白的磷酸化抑制剂的方法,其中,所述抑制剂使CREPT蛋白的S134和S166位点保持持续非磷酸化状态,从而使所述CREPT蛋白在细胞中保持非泛素化状态且不会降解;所述CREPT蛋白的氨基酸序列为SEQ ID No:4,所述方法包括:A method for identifying whether a substance is a phosphorylation inhibitor of CREPT protein, wherein the inhibitor keeps the S134 and S166 sites of the CREPT protein in a continuous non-phosphorylated state, thereby maintaining the non-ubiquitin of the CREPT protein in cells. The amino acid sequence of the CREPT protein is SEQ ID No: 4, and the method includes:
    i)将待鉴定的物质加入表达CREPT蛋白的真核细胞中并培养所述真核细胞,和i) adding the substance to be identified to eukaryotic cells expressing the CREPT protein and culturing the eukaryotic cells, and
    ii)使用免疫沉淀法检查所述真核细胞的CREPT蛋白的泛素化水平;ii) using an immunoprecipitation method to examine the ubiquitination level of the CREPT protein of the eukaryotic cells;
    与未经所述物质处理的对照细胞中CREPT蛋白的泛素化水平相比,如果经处理的细胞中的CREPT蛋白的泛素化水平下降,例如下降10%以上、20%以上、30%以上或40%以上,则将所述物质鉴定为CREPT蛋白的磷酸化抑制剂,否则所述物质不是CREPT蛋白的磷酸化抑制剂。Compared with the ubiquitination level of CREPT protein in control cells that have not been treated with the substance, if the ubiquitination level of CREPT protein in the treated cells decreases, for example, decreases by more than 10%, more than 20%, or more than 30% or more than 40%, then the substance is identified as a phosphorylation inhibitor of CREPT protein, otherwise the substance is not a phosphorylation inhibitor of CREPT protein.
  32. 如权利要求30所述的方法,其中,在步骤i)之前,所述方法还包括:使用预测工具SwissTargetPrediction和SEA针对CREPT来设计所述待鉴定的物质。The method of claim 30, wherein before step i), the method further comprises: using prediction tools SwissTargetPrediction and SEA to design the substance to be identified for CREPT.
  33. 如权利要求30所述的方法,其中,步骤ii)包括用识别CREPT蛋白的抗CREPT抗体和识别泛素的泛素抗体对CREPT蛋白的泛素化水平进行定量。The method of claim 30, wherein step ii) includes quantifying the ubiquitination level of the CREPT protein using an anti-CREPT antibody that recognizes the CREPT protein and an ubiquitin antibody that recognizes ubiquitin.
  34. 一种抑制CREPT蛋白在真核细胞中的降解的方法,所述方法包括:A method for inhibiting the degradation of CREPT protein in eukaryotic cells, the method comprising:
    1)使选自SKP2抑制剂、CUL1抑制剂、neddylation抑制剂和CDK2抑制剂的抑制剂进入表达CREPT的真核细胞中;和/或1) Allowing an inhibitor selected from the group consisting of SKP2 inhibitors, CUL1 inhibitors, neddylation inhibitors and CDK2 inhibitors to enter eukaryotic cells expressing CREPT; and/or
    2)对CREPT蛋白的第134和166位点进行磷酸化失活修饰,使得当经修饰的蛋 白位于G1末期或G1/S转换期的真核细胞中时,所述134和166位点保持非磷酸化状态且所述经修饰的蛋白保持非泛素化状态,从而使所述经修饰的蛋白不被降解。2) Phosphorylation and inactivation modification of positions 134 and 166 of the CREPT protein, so that when the modified protein When the protein is located in eukaryotic cells at the end of G1 phase or G1/S transition phase, the 134 and 166 sites remain in an unphosphorylated state and the modified protein remains in a non-ubiquitinated state, thereby making the modified protein The protein is not degraded.
  35. 如权利要求34所述的方法,其中,所述真核细胞是人、酵母、小鼠、犬、猫、鸡、蟾蜍、斑马鱼、果蝇、线虫或拟南芥的细胞,优选人细胞,更优选人癌细胞。The method of claim 34, wherein the eukaryotic cell is a human, yeast, mouse, dog, cat, chicken, toad, zebrafish, fruit fly, nematode or Arabidopsis thaliana cell, preferably a human cell, Human cancer cells are more preferred.
  36. 如权利要求34所述的方法,其中,所述134和166位点上的所述磷酸化失活修饰是氨基酸突变和/或化学修饰。The method of claim 34, wherein the phosphorylation inactivating modifications at positions 134 and 166 are amino acid mutations and/or chemical modifications.
  37. 如权利要求34所述的方法,其中,所述134和166位点上的所述磷酸化失活修饰是将丝氨酸(S)突变为丙氨酸(A)。The method of claim 34, wherein the phosphorylation-inactivating modifications at positions 134 and 166 are mutations of serine (S) to alanine (A).
  38. 如权利要求34所述的方法,其中,所述SKP2抑制剂是针对SKP2的双链siRNA,其序列为AAUCUAAGCCUGGAAGGCCUGdTdT;所述CUL1抑制剂是针对CUL1的双链siRNA,其序列为UAGACAUUGGGUUCGCCGUdTdT;所述neddylation抑制剂是MLN4924。The method of claim 34, wherein the SKP2 inhibitor is a double-stranded siRNA directed against SKP2 and its sequence is AAUCUAAGCCUGGAAGGCCUGdTdT; the CUL1 inhibitor is a double-stranded siRNA directed against CUL1 and its sequence is UAGACAUUGGGUUCGCCGUdTdT; the neddylation The inhibitor is MLN4924.
  39. 一种蛋白,其选自:A protein selected from:
    1)通过将SEQ ID No:4的166位丝氨酸突变为丙氨酸而得到的蛋白;1) Protein obtained by mutating serine 166 of SEQ ID No: 4 to alanine;
    2)通过将SEQ ID No:8的136位丝氨酸突变为丙氨酸而得到的蛋白;2) Protein obtained by mutating serine at position 136 of SEQ ID No:8 to alanine;
    3)通过将SEQ ID No:8的174位丝氨酸突变为丙氨酸而得到的蛋白;3) Protein obtained by mutating serine 174 of SEQ ID No: 8 to alanine;
    4)通过将SEQ ID No:4的134和166位丝氨酸均突变为半胱氨酸而得到的蛋白;4) Protein obtained by mutating serine 134 and serine 166 of SEQ ID No: 4 to cysteine;
    5)通过将SEQ ID No:4的166位丝氨酸突变为半胱氨酸而得到的蛋白;5) Protein obtained by mutating serine 166 of SEQ ID No: 4 to cysteine;
    6)通过将SEQ ID No:8的136位丝氨酸突变为半胱氨酸而得到的蛋白;6) Protein obtained by mutating serine at position 136 of SEQ ID No:8 to cysteine;
    7)通过将SEQ ID No:8的174位丝氨酸突变为半胱氨酸而得到的蛋白;7) Protein obtained by mutating serine at position 174 of SEQ ID No:8 to cysteine;
    8)通过将SEQ ID No:8的136和174位丝氨酸均突变为半胱氨酸而得到的蛋白;和8) A protein obtained by mutating both serine 136 and serine 174 of SEQ ID No: 8 to cysteine; and
    9)与1)至8)中的任一种蛋白具有90%以上的序列同一性且具有相同的丙氨酸或半胱氨酸突变的蛋白。9) A protein that has more than 90% sequence identity with any protein in 1) to 8) and has the same alanine or cysteine mutation.
  40. 编码权利要求39所述的蛋白的核酸。Nucleic acid encoding the protein of claim 39.
  41. 包含权利要求40所述的核酸的载体。 A vector comprising the nucleic acid of claim 40.
PCT/CN2023/111172 2022-08-12 2023-08-04 Non-phosphorylated and non-ubiquitinated crept protein and use thereof WO2024032485A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210970052.6A CN116143907A (en) 2022-08-12 2022-08-12 Non-phosphorylated and non-ubiquitinated CREPT proteins and uses thereof
CN202210970052.6 2022-08-12

Publications (1)

Publication Number Publication Date
WO2024032485A1 true WO2024032485A1 (en) 2024-02-15

Family

ID=86351289

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/111172 WO2024032485A1 (en) 2022-08-12 2023-08-04 Non-phosphorylated and non-ubiquitinated crept protein and use thereof

Country Status (2)

Country Link
CN (1) CN116143907A (en)
WO (1) WO2024032485A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116143907A (en) * 2022-08-12 2023-05-23 清华大学 Non-phosphorylated and non-ubiquitinated CREPT proteins and uses thereof
CN116003566A (en) * 2022-08-12 2023-04-25 清华大学 CREPT mutant and application thereof in inhibiting tumor growth

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1807623A (en) * 2005-12-28 2006-07-26 清华大学 Positive regulation gene for cell cycle and its coded protein and uses
CN108949718A (en) * 2018-07-25 2018-12-07 清华大学 CREPT(S145A) mutant and its application in inhibition tumour growth
CN109295000A (en) * 2018-10-15 2019-02-01 清华大学 A kind of multipotential stem cell forms the application in inducing multi-potent stem cell of indispensable protein CREPT
CN116143907A (en) * 2022-08-12 2023-05-23 清华大学 Non-phosphorylated and non-ubiquitinated CREPT proteins and uses thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1807623A (en) * 2005-12-28 2006-07-26 清华大学 Positive regulation gene for cell cycle and its coded protein and uses
CN108949718A (en) * 2018-07-25 2018-12-07 清华大学 CREPT(S145A) mutant and its application in inhibition tumour growth
CN109295000A (en) * 2018-10-15 2019-02-01 清华大学 A kind of multipotential stem cell forms the application in inducing multi-potent stem cell of indispensable protein CREPT
CN116143907A (en) * 2022-08-12 2023-05-23 清华大学 Non-phosphorylated and non-ubiquitinated CREPT proteins and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DING LIDAN, YANG LIU, HE YUQI, ZHU BINGTAO, REN FANGLI, FAN XUANZI, WANG YINYIN, LI MENGDI, LI JUN, KUANG YANSHEN, LIU SIHAN, ZHAI: "CREPT/RPRD1B associates with Aurora B to regulate Cyclin B1 expression for accelerating the G2/M transition in gastric cancer", CELL DEATH & DISEASE, NATURE PUBLISHING GROUP, GB, vol. 9, no. 12, 5 December 2018 (2018-12-05), GB , pages 1172, XP093137885, ISSN: 2041-4889, DOI: 10.1038/s41419-018-1211-8 *

Also Published As

Publication number Publication date
CN116143907A (en) 2023-05-23

Similar Documents

Publication Publication Date Title
Hangen et al. Interaction between AIF and CHCHD4 regulates respiratory chain biogenesis
Villa et al. Parkin-independent mitophagy controls chemotherapeutic response in cancer cells
WO2024032485A1 (en) Non-phosphorylated and non-ubiquitinated crept protein and use thereof
Vlach et al. Phosphorylation‐dependent degradation of the cyclin‐dependent kinase inhibitor p27Kip1
Mailand et al. Regulation of G2/M events by Cdc25A through phosphorylation‐dependent modulation of its stability
Beli et al. Proteomic investigations reveal a role for RNA processing factor THRAP3 in the DNA damage response
Fu et al. Set7 mediated Gli3 methylation plays a positive role in the activation of Sonic Hedgehog pathway in mammals
Bigot et al. Phosphorylation-mediated interactions with TOPBP1 couple 53BP1 and 9-1-1 to control the G1 DNA damage checkpoint
Chen et al. ADP‐ribosylation of histone variant H2AX promotes base excision repair
Calvayrac et al. Cytoplasmic p27Kip1 promotes tumorigenesis via suppression of RhoB activity
Zhu et al. SUMOylation of HNRNPA2B1 modulates RPA dynamics during unperturbed replication and genotoxic stress responses
Watanabe et al. FBXO 21 mediates the ubiquitylation and proteasomal degradation of EID 1
AU2001265947B2 (en) Enzymatic assays for screening anti-cancer agents
US7846909B2 (en) Method and compositions for inhibiting MAGE protein interaction with KAP-1
AU2001265947A1 (en) Enzymatic assays for screening anti-cancer agents
Malcles et al. Regulation of bovine papillomavirus replicative helicase E1 by the ubiquitin-proteasome pathway
Masilamani et al. Calpain-mediated cleavage generates a ZBTB18 N-terminal product that regulates HIF1A signaling and glioblastoma metabolism
Gao et al. NDP52 SUMOylation contributes to low‐dose X‐rays‐induced cardiac hypertrophy through PINK1/Parkin‐mediated mitophagy via MUL1/SUMO2 signalling
US20070072210A1 (en) Use of brca1-associated protein to treat and screen for dna damage and to identify therapeutics that promote a dna damage response
Lin et al. β-8-Oxoguanine DNA Glycosylase Overexpression Reduces Oxidative Stress-Induced Mitochondrial Dysfunction and Apoptosis Through the JNK Signaling Pathway in Human Bronchial Epithelial Cells
KR102258977B1 (en) A method for screening a therapeutic agent for cancer using binding inhibitor of Cyclin-dependent kinase 1(CDK1) - Cyclin B1
KR102138229B1 (en) A method for screening a therapeutic agent for cancer using binding inhibitor of Cyclin-dependent kinase 1(CDK1) - Cyclin B1
Chacin A New Mechanism of Activation of the Anaphase-Promoting Complex/Cyclosome
Sarwar et al. LKB1 negatively regulates AKT1 signaling via DBC1 and TRB3
de la Peña Avalos et al. The protein phosphatase EYA4 promotes homologous recombination (HR) through dephosphorylation of tyrosine 315 on RAD51

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: 23851706

Country of ref document: EP

Kind code of ref document: A1