WO2024113302A1 - Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use - Google Patents

Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use Download PDF

Info

Publication number
WO2024113302A1
WO2024113302A1 PCT/CN2022/135820 CN2022135820W WO2024113302A1 WO 2024113302 A1 WO2024113302 A1 WO 2024113302A1 CN 2022135820 W CN2022135820 W CN 2022135820W WO 2024113302 A1 WO2024113302 A1 WO 2024113302A1
Authority
WO
WIPO (PCT)
Prior art keywords
grna
hsv
vector
viral vector
virus
Prior art date
Application number
PCT/CN2022/135820
Other languages
French (fr)
Chinese (zh)
Inventor
王华东
徐富强
应敏
刘同钽
史青
Original Assignee
深圳先进技术研究院
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 深圳先进技术研究院 filed Critical 深圳先进技术研究院
Priority to PCT/CN2022/135820 priority Critical patent/WO2024113302A1/en
Publication of WO2024113302A1 publication Critical patent/WO2024113302A1/en

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • 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
    • 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
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome

Definitions

  • the patent of this invention relates to a method of using CRISPR/Cas gene editing for gene therapy, and in particular to gRNA targeting HSV essential genes, a CRISPR/Cas gene editing system, a delivery system and applications.
  • Herpes simplex virus 1 is the prototype member of the human herpesviridae family. Epidemiological surveys have shown that herpes simplex virus type 1 (HSV-1) is very common and prevalent worldwide, with a global prevalence of about 70%, and most infections occur in childhood. HSV-1 is a highly contagious virus that is mainly transmitted through oral-oral contact. Autopsy studies have shown that the HSV-1 viral genome is present in the trigeminal ganglion of 85-90% of individuals, and is the most common cause of cold sores, viral encephalitis, and infectious blindness. HSV-1 usually causes mild or even asymptomatic infections in healthy or immunocompetent individuals, but can cause severe corneal infections and neuroencephalitis caused by severe central nervous system (CNS) infections in newborns or immunocompromised patients.
  • CNS central nervous system
  • HSV infection Clinical symptoms caused by HSV infection include latent infection without clinical symptoms, oral ulcers and corneal inflammation, and severe central nervous system infection causing neurological encephalitis.
  • HSV-1 is the most common cause of encephalitis in children and adults.
  • HSV-1 is a common neurotropic pathogen that can reach the central nervous system without obvious clinical symptoms.
  • HSV-1 can infect the terminals of olfactory neurons and enter the CNS through retrograde axonal transport of neurons, eventually reaching the olfactory bulb in the brain.
  • HSV-1 can also be reactivated from neurons in the trigeminal ganglion and reach the skin or CNS through anterograde transport; after entering the CNS, the virus can either remain dormant in this tissue or be activated to cause severe acute necrotizing encephalitis.
  • Herpes simplex encephalitis is one of the most serious manifestations of HSV-1 infection, with a mortality rate of up to 97%.
  • HSV-1 infection can cause a chronic immune inflammatory disease in the cornea, herpes simplex keratitis (HSK), which can cause irreversible damage and blindness in infected people.
  • HSK herpes simplex keratitis
  • the virus enters the corneal epithelium from the trigeminal ganglion latently and causes an inflammatory response through reactivation. Patients are characterized by corneal opacity and new blood vessel formation on the surface of the eye. Recurrent inflammation caused by periodic reactivation of the virus may even lead to blindness.
  • HVEM is an important host cell protein receptor in the immune pathogenesis of HSV-1 infection in the eye.
  • Herpes simplex virus type 1 (HSV-1) is a major cause of neuroencephalitis and infectious blindness.
  • current drugs for the treatment of HSV cannot eliminate latent HSV virus or reactivated virus in the trigeminal ganglion. Therefore, the development of new therapeutic strategies against latent HSV has become a priority.
  • CRISPR-based genome editing technology represents a promising approach to combat viral infection and replication by modifying or destroying the viral genetic material.
  • the present invention aims to provide gRNA, CRISPR/Cas gene editing system, delivery system and application targeting HSV essential genes.
  • the present invention designs and screens a series of gRNAs that directly target HSV essential genes VP16, ICP27, ICP4 or gD, VP16 (2gRNA), ICP27 (2gRNA), ICP4 (2gRNA) or gD (4gRNA), and specifically provides new technologies, methods and delivery strategies based on CRISPR-Cas gene editing to resist HSV infection.
  • the first aspect of the present invention provides a gRNA targeting an essential gene of the HSV virus, wherein the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
  • the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
  • a second aspect of the present invention provides a gRNA composition targeting essential genes of the HSV virus, wherein the gRNA composition is selected from two or more of the nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
  • the gRNA composition is a composition of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
  • the third aspect of the present invention provides a gRNA expression vector targeting essential genes of the HSV virus, which comprises the gRNA, or the gRNA composition.
  • the expression vector is a viral vector or a non-viral vector
  • the viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
  • the non-viral vector is a plasmid or mRNA.
  • a fourth aspect of the present invention provides a CRISPR/Cas gene editing system, which comprises the gRNA, or the gRNA composition, and a Cas protein.
  • the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, Cas9 from Neisseria meningitidis, Cas9 from Streptococcus thermophilus, Cas9 from Staphylococcus aureus, Cpf1 from Acidaminococcus, Cpf1 from Corynebacterium glutamicum, micro-SaCas9 from Staphylococcus aureus, and C2c2 from Oral Cibotium;
  • the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
  • a fifth aspect of the present invention provides a delivery system for the CRISPR/Cas gene editing system, the delivery system comprising:
  • the Cas protein expression vector is a viral vector or a non-viral vector
  • the viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
  • the non-viral vector is a plasmid or mRNA.
  • a sixth aspect of the present invention provides a kit comprising:
  • the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, Cas9 from Neisseria meningitidis, Cas9 from Streptococcus thermophilus, Cas9 from Staphylococcus aureus, Cpf1 from Acidaminococcus, Cpf1 from Corynebacterium glutamicum, mini-SaCas9 from Staphylococcus aureus, and C2c2 from Oral Cibotium;
  • the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
  • the Cas protein expression vector is a viral vector or a non-viral vector
  • the viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
  • the non-viral vector is a plasmid or mRNA.
  • the seventh aspect of the present invention provides the use of the gRNA, or the gRNA composition, or the gRNA expression vector, or the CRISPR/Cas gene editing system, or the delivery system, or the kit in the preparation of drugs for preventing and/or treating HSV infection-related diseases.
  • the HSV infection-related disease is an HSV-1 or HSV-2-related disease
  • the HSV infection-related diseases include viral encephalitis, herpes simplex virus keratitis and infectious blindness.
  • the eighth aspect of the present invention provides the use of the gRNA, or the gRNA composition, or the gRNA expression vector, or the CRISPR/Cas gene editing system, or the delivery system, or the kit in the preparation of drugs for preventing and/or treating anti-HSV.
  • the HSV is HSV-1 or HSV-2.
  • the ninth aspect of the present invention provides a pharmaceutical composition comprising a delivery system for the CRISPR/Cas gene editing system.
  • the tenth aspect of the present invention provides a method for inhibiting HSV replication and/or expression of HSV essential genes, the method comprising: before or after HSV infection, transfecting the target cells with the delivery system of the CRISPR/Cas gene editing system, and the ribonucleoprotein complex formed by the Cas-gRNA expressed by the cells targetedly and precisely shears and destroys the HSV genome, thereby blocking viral replication and transmission.
  • the present invention provides a new technology for resisting herpes simplex virus infection by using CRISPR-Cas genome editing technology.
  • a series of gRNAs directly targeting HSV essential genes VP16, ICP27, ICP4 or gD are designed and screened for the genome sequence of HSV clinical strains, and a gene therapy viral vector or a non-viral vector is used to deliver nuclease Cas9 and gRNA targeting viral genes.
  • Gene editing targeted to HSV key genes can effectively inhibit HSV replication and spread infection, and each gRNA can effectively inhibit HSV viral replication when mediated alone.
  • the gRNAs of aVP16-2, aICP27-2, aICP4-2 or agD-1 can inhibit HSV viral replication better than the same group, and the gene editing of gRNAs in pairs can more effectively inhibit HSV viral replication.
  • the mixed gene editing (named Cocktail) of four gRNAs of aVP16-2, aICP27-2, aICP4-2 and agD-1 inhibits HSV viral replication better than the gene editing mediated by a single gRNA, and the cocktail strategy of gRNA combination shows an effect better than single target editing.
  • in vivo gene editing using CRISPR-Cas9/gRNA delivered by viral vectors can effectively block HSV replication and transmission.
  • the present invention uses the method of gRNA mixed gene editing (Cocktail), whether it is a prevention strategy or a treatment strategy Cocktail, which can effectively inhibit HSV virus replication and inter-tissue transmission.
  • gRNA mixed gene editing Cocktail
  • the current drugs for treating HSV cannot eliminate the HSV virus lurking in the trigeminal ganglion or the virus relapses.
  • the gene therapy mediated by the method of the present invention and viral vectors can effectively eliminate latently infected or recurrently infected HSV viruses, providing a potential important means for the treatment of refractory HSV-1 related diseases.
  • FIG1 is a diagram showing the screening of HSV target genes for CRISPR gene editing and the design of gRNA
  • Figure 2 is an identification diagram of CRISPR Cas9/gRNA plasmid
  • Figure 3 is a diagram showing the effect of Cas9/gRNA single plasmid transfection and anti-HSV replication
  • Figure 4 is the determination of virus titer in the cell supernatant after Cas9/gRNA single plasmid transfection editing
  • Figure 5 is a diagram showing the design of the Cas9/gRNA plasmid optimization dual-plasmid combination strategy and the anti-HSV replication effect
  • Figure 6 is a diagram showing the cocktail strategy of four types of gRNAs of YouScreen and the anti-HSV replication effect at the cellular level;
  • FIG7 is a determination of virus titer in the cell supernatant after editing using the cocktail combination strategy
  • Figure 8 is a comparison of the preventive and therapeutic effects of Cocktail multi-target editing
  • FIG9 is a diagram showing the packaging preparation of gene delivery Cas9/gRNA lentivirus and its anti-HSV effect
  • Figure 10 shows that the LV-Cas9/gRNA control virus cannot inhibit HSV replication
  • Figure 11 is a diagram showing the effect of LV-Cas9/gRNA single virus on anti-HSV replication in vivo;
  • Figure 12 shows the LV-Cas9/gRNA virus cocktail strategy against HSV replication
  • FIG13 shows the dose effect of LV-Cas9/gRNA virus combination (Cocktail).
  • Figure 14 is a diagram showing the effect of LV-Cas9/gRNA virus combination (Cocktail) in treating HSV infection.
  • the present invention designs and screens a series of gRNAs that directly target HSV essential genes VP16, ICP27, ICP4 or gD, VP16 (2 gRNAs), ICP27 (2 gRNAs), ICP4 (2 gRNAs) or gD (4 gRNAs).
  • the present invention uses vectors to deliver Cas9/gRNA to target cells to achieve editing of targeted genes.
  • the vectors include commonly used viral vectors and non-viral vectors, wherein viral vectors include lentiviral vectors, retroviral vectors, adeno-associated viral vectors, herpes simplex virus vectors, adenoviral vectors, virus-like particles (VLPs), etc.; non-viral vectors include plasmids, mRNA, etc.
  • the present invention constructs 10 lentiviral vector core plasmids carrying SpCas9 and corresponding gRNA, and then packages and prepares lentiviral vectors of gene delivery CRISPR editing system. First, the inhibitory effects of 10 SpCas9/gRNAs were screened in cell experiments.
  • the virus in the control group (NC and Vec) rapidly proliferated, and almost all cells expressed green fluorescent protein, while the fluorescence ratios of the 10 gRNA groups targeting the cutting of 4 genes decreased to varying degrees; by detecting the HSV virus titer of the cell supernatant, the virus titer of the experimental group was significantly lower than that of the control group, and the inhibitory effects of the four gRNA groups of agD-1, aICP4-2, aVP16-2, and aICP27-2 were screened out as the most obvious, thus serving as an alternative for packaging lentiviral vectors.
  • the combined gRNA strategy (“cocktail” strategy) clearly showed superior effects over single gRNA gene editing.
  • the present invention tests whether the combination of Cas9/gRNA for targeted editing of 4 genes can achieve better HSV inhibition effects.
  • the Cocktail group showed a significant inhibitory effect on HSV replication at low doses, and there was an obvious dose effect.
  • the amount of Cas9/gRNA plasmids increased, the stronger the effect of inhibiting viral proliferation.
  • the present invention conducted an experiment of first infecting the HSV virus and then treating it with the CRISPR editing system.
  • the HSV-GFP virus was infected 12 hours, 6 hours and 1 hour in advance, respectively, and then the Cas9/gRNA-cocktail plasmid was transfected. It was found that during the high-speed proliferation stage of acute HSV virus infection (12 hours of viral proliferation), Cas9/gRNA intervention had a certain inhibitory effect but it was not obvious. If intervention was given earlier after HSV infection - CRISPR treatment in the 6-hour viral proliferation group and the 1-hour viral proliferation group could significantly reduce the proliferation of HSV virus, and the earlier the intervention, the better.
  • the present invention uses packaged lentivirus to carry out animal experiments in vivo.
  • the Cas9/gRNA combination (Cocktail) strategy can effectively inhibit virus proliferation and spread.
  • the control group (NC) without any intervention and the lentivirus empty control group (Vec)
  • the virus has been efficiently proliferated 5 days after H129-GFP was injected into the mouse V1 brain region, and spread to brain regions such as LGd along the neural circuit.
  • the infected neurons are highlighted by fluorescent protein, and the lentivirus empty administration has no therapeutic effect.
  • the effects of inhibiting viral replication are shown, and the effects of the two groups of LV-aICP4 and LV-aICP27 are better than LV-gD and LV-aVP16.
  • the Cas9/gRNA combination (Cocktail) treatment effect is relatively the best, and no or only a small number of cells are observed to be infected and marked at the injection site V1, and it also shows a dose effect.
  • Another important thing about the present invention is that it also verifies the "infection first, then treatment” experiment, where the mouse V1 brain region is first infected with H129-GFP, and then lentivirus cocktail treatment is administered one day later. It is found that the replication and spread of HSV can be effectively inhibited, and the effect is better than single lentivirus administration.
  • CRISPR gene editing targeting the four key genes of HSV, VP16, ICP27, ICP4, and gD can effectively inhibit the replication and proliferation of HSV in the lytic replication cycle, and the Cas9/gRNA combination (Cocktail) strategy is more efficient. It can be imagined that the use of CRISPR genome editing technology to eliminate latent HSV viruses in a latent infection state with much lower viral titers may have a more significant inhibitory effect.
  • the Cas9/gRNA genome editing of the present invention can effectively inhibit the infection and spread of HSV at the prevention level and the treatment level, especially in the in vivo animal experiment.
  • the proliferation of HSV is significantly inhibited, and the number of labeled nerve cells decreases exponentially compared with the control group.
  • no related toxic side effects were observed in the Cas9/gRNA combination (Cocktail) treatment, indicating the safety of this strategy.
  • This method of eliminating HSV infection using CRISPR genome editing provides a new means for the potential treatment of refractory HSV-1 related diseases, especially when traditional treatments are ineffective.
  • Example 1 Screening of HSV target genes for CRISPR gene editing and design and synthesis of gRNA
  • HSV essential genes VP16, ICP27, ICP4 and gD were selected as potential targets for CRISPR/Cas9 strategy editing.
  • four essential viral genes were designed and targeted, and guide RNAs combined with Cas9 were developed to test their ability to inhibit HSV-1 replication.
  • sgRNA was designed based on the target site sequence of the selected gene and screened by CRISPR DESIGN system (http://crispr.mit.edu/).
  • the sgRNA oligonucleotide sequences are shown in Table 1.
  • a total of 10 gRNAs were designed: VP16 targeting (2 gRNAs, sequences see SEQ ID NO.1 and 2), ICP27 targeting (2 gRNAs, sequences see SEQ ID NO.3 and 4), ICP4 targeting (2 gRNAs, sequences see SEQ ID NO.5 and 6) and gD targeting (4 gRNAs, sequences see SEQ ID NO.7-10) ( Figure 1, Table 1).
  • the corresponding 10 core plasmids of lentiviral vectors carrying SpCas9 and gRNA were constructed and verified ( Figure 1) and the corresponding lentivirus was packaged.
  • sgRNA was designed according to the target sequence of the selected gene and screened by the CRISPR design system (http://crispr.mit.edu/).
  • the sgRNA oligonucleotide sequences are shown in Table 1.
  • the empty vector lentiCRISPR v2 vector (addgene, #52961, sequence see SEQ ID NO.11) was digested with BsmBI, and the annealed oligonucleotides were cloned into the single guide RNA scaffold to obtain the CRISPR Cas9/gRNA lentiviral core plasmid.
  • the packaging plasmid was then used to produce lentiCRISPR-sgRNA viruses in HEK293T cells as previously described (Cold Spring Harb Protoc.
  • the above lentivirus constructs were co-transfected into 293T cells with three lentiviral packaging vectors (pGAG/POL, pREV, and pMD2.G), and the supernatant was collected 2 to 3 days later to obtain the targeted virus.
  • the titers of the virus stocks of the five lentiviruses produced were all 1 ⁇ 10 8 PFU/ml.
  • the plasmid was constructed, it was sent to a sequencing company for sequencing, and the sequencing result was successful. At the same time, the plasmid was used as a template to amplify the target fragment.
  • the reaction system and PCR program were as follows:
  • Example 4 Cas9/gRNA single plasmid transfection and anti-HSV replication effect
  • BHK cells in the logarithmic growth phase were collected and inoculated into six-well plates at a density of 1 ⁇ 10 6 cells/mL. After the cells adhered to the wall, the cell culture medium was replaced with 400 ⁇ L Opti-MEM serum-free medium.
  • the virus supernatant was collected and the virus titer was measured by plaque assay.
  • BHK cells in the logarithmic growth phase were taken and inoculated into 12-well plates at a density of 2 ⁇ 10 5 /mL, and the cells were allowed to adhere to the wall.
  • the virus supernatant was diluted 10 times 6 times, and 100 ⁇ L of the diluted virus supernatant was successively drawn into the well plate. After adsorption for 1 hour, the virus liquid was discarded, and an equal volume of 4% DMEM culture medium and 1% agarose solution were added. After standing, the agarose concentration was placed in a 35°C, 3% CO 2 cell culture incubator for 24 hours, and the spots were observed and counted under a fluorescence microscope. The results are shown in Figure 3. Compared with the control group, the expression of green fluorescent protein was reduced after Cas9/gRNA single plasmid transfection.
  • the titer of HSV is another measure of the effectiveness of Cas9/gRNA compared with the control group, as shown in Figure 4, which shows a significant reduction in the HSV titer in the Cas9/gRNA transfection group.
  • agD-1, aICP4-2, aICP27-2, and aVP16-2 had the best inhibitory effect on the proliferation of HSV, so these Cas9/gRNA plasmids were selected for subsequent experiments and lentiviral packaging.
  • Example 5 Cas9/gRNA plasmid optimization dual plasmid combination strategy design and anti-HSV replication effect
  • the four Cas9/gRNA plasmids agD-1, aICP4-2, aICP27-2, and aVP16-g2 with the highest inhibition efficiency on HSV replication in Case 4 were selected and combined in pairs (agD-g1+aICP4-g2; agD-g1+aICP27-g2; agD-g1+aVP16-g2; aICP27-g2+aICP4-g2; aVP16-g2+aICP4-g2; aICP27-g2+aVP16-g2;) and transfected into BHK cells together. After 24 hours, the cells were infected at an MOI of 0.1.
  • the virus solution was discarded, 2 mL of DMEM medium containing 2% serum was added, and the cells were cultured in a cell culture incubator at 35°C and 3% CO2 for 48 hours. The cells were observed and imaged using a fluorescence microscope. As shown in Figure 5, the expression of green fluorescent protein was reduced after the optimized double-plasmid group was combined and transfected compared with the control group.
  • Example 6 Cocktail strategy of four types of gRNAs of YouScreen and anti-HSV replication effect at the cellular level
  • Example 8 Packaging and preparation of gene delivery Cas9/gRNA lentivirus
  • Each prepared DNA solution (20 ⁇ g of pGC-LV vector, 15 ⁇ g of pHelper 1.0 vector, and 10 ⁇ g of pHelper 2.0 vector) was mixed evenly with the corresponding volume of Opti-MEM, and the total volume was adjusted to 2.5 ml. The solution was incubated at room temperature for 5 minutes.
  • 293T cells human kidney epithelial cell line
  • Cas9/gRNA human kidney epithelial cell line
  • the results were consistent with the plasmid effect on BHK cells. The results not only showed that Cas9/gRNA editing can effectively inhibit HSV replication in different cell lines, but also that the packaged lentivirus has a good effect. See Figure 9.
  • Example 9 LV-Cas9/gRNA control virus cannot inhibit HSV replication
  • a microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel should be applied to the sutured wound to prevent inflammation and relieve pain in the mice. On day 5, 500 H129-EGFP viral particles were injected into the same brain region.
  • Example 10 Effect of LV-Cas9/gRNA single virus on HSV replication in vivo
  • a microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel was applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
  • Lentivirus expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into the primary visual cortex V1 in advance, and then H129-EGFP was injected 5 days later. It was observed that there was no EGFP expression in the downstream output area LGD of the H129-EGFP injection site V1 and the contralateral V1, indicating that the infection and replication of the virus were significantly inhibited (see Figure 11).
  • Example 11 LV-Cas9/gRNA virus cocktail strategy to prevent HSV replication
  • a microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel were applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
  • Example 12 LV-Cas9/gRNA virus combination (Cocktail) dose effect
  • a microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel were applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
  • Example 13 Effect of LV-Cas9/gRNA virus cocktail in treating HSV infection
  • a microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and virus injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel should be applied to the sutured wound to prevent inflammation and relieve pain in the mice. On day 5, 5 ⁇ 10 4 TU of lentivirus mixtures expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into the same brain region.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed are gRNA targeting HSV essential genes, a CRISPR/Cas gene editing system, a delivery system, and use. Aiming at an HSV clinical strain genome sequence, the present invention designs and screens a gRNA series that directly targets the HSV essential genes VP16, ICP27, ICP4, or gD, and uses a viral vector or a non-viral vector to deliver nuclease Cas9 and gRNA targeting a viral gene. The HSV key gene-targeted gene editing can effectively inhibit HSV replication and spread of infection, and the gRNA combination cocktail strategy shows an effect superior to that of single-target editing. The method of the present invention and the gene therapy mediated by the viral vector can effectively eliminate HSV viruses with latent infections or recurrent infections, thus providing a potentially important means for the treatment of refractory HSV-1-related diseases such as viral encephalitis, herpes simplex keratitis, and infectious blindness.

Description

靶向HSV必需基因的gRNA、CRISPR/Cas基因编辑系统、递送系统及应用gRNA targeting HSV essential genes, CRISPR/Cas gene editing system, delivery system and application 技术领域Technical Field
本发明专利涉及CRISPR/Cas基因编辑用于基因治疗的方法,特别涉及靶向HSV必需基因的gRNA、CRISPR/Cas基因编辑系统、递送系统及应用。The patent of this invention relates to a method of using CRISPR/Cas gene editing for gene therapy, and in particular to gRNA targeting HSV essential genes, a CRISPR/Cas gene editing system, a delivery system and applications.
背景技术Background technique
单纯疱疹病毒1(HSV-1)是人类疱疹病毒科的原型成员。流行病学调查表明,单纯疱疹病毒Ⅰ型(herpes simplexvirus 1,HSV-1)在世界范围内非常普遍和流行,流行病学调查显示,全球流行率约为70%,且大多数感染发生在儿童时期。HSV-1是一种具有高度传染性的病毒,主要通过口-口接触传播。尸检研究表明85-90%的个体三叉神经节中存在HSV-1病毒基因组,是唇疱疹、病毒性脑炎和感染性失明的最常见病因。HSV-1通常会在健康或免疫能力正常的个体中引起轻度甚至无症状感染,但在新生儿或免疫功能受损的患者中会导致严重的眼角膜感染和由严重的中枢神经系统(CNS)感染引起的神经脑炎。Herpes simplex virus 1 (HSV-1) is the prototype member of the human herpesviridae family. Epidemiological surveys have shown that herpes simplex virus type 1 (HSV-1) is very common and prevalent worldwide, with a global prevalence of about 70%, and most infections occur in childhood. HSV-1 is a highly contagious virus that is mainly transmitted through oral-oral contact. Autopsy studies have shown that the HSV-1 viral genome is present in the trigeminal ganglion of 85-90% of individuals, and is the most common cause of cold sores, viral encephalitis, and infectious blindness. HSV-1 usually causes mild or even asymptomatic infections in healthy or immunocompetent individuals, but can cause severe corneal infections and neuroencephalitis caused by severe central nervous system (CNS) infections in newborns or immunocompromised patients.
HSV感染导致的临床症状包括无临床症状的潜伏感染、口腔溃疡和眼角膜炎症以及严重的中枢神经系统感染造成的神经性脑炎。在北美,HSV-1是引起儿童和成人脑炎最常见的病因。HSV-1是常见的嗜神经性病原体,可以在没有明显临床症状的情况下到达中枢神经系统。HSV-1可感染嗅神经元的末端,并通过神经元的逆行轴突运输进入CNS,最终到达脑中的嗅球。HSV-1也可以从三叉神经节中的神经元重新激活,并通过顺行运输到达皮肤或CNS;进入CNS后,病毒既可以在该组织中处于静止的潜伏状态,也可以激活导致严重的急性坏死性脑炎。单纯疱疹病毒性脑炎(herpes simplex encephalitis,HSE)是HSV-1感染产生的最严重的表现之一,死亡率可高达97%。研究表明,虽然使用抗病毒药物阿昔洛韦(acyclovir,ACV)治疗可降低HSE患者的死亡率,但大部分患者康复后存在永久性神经系统后遗症,包括认知、记忆和行为障碍的症状,并且会增加癫痫的发病率。Clinical symptoms caused by HSV infection include latent infection without clinical symptoms, oral ulcers and corneal inflammation, and severe central nervous system infection causing neurological encephalitis. In North America, HSV-1 is the most common cause of encephalitis in children and adults. HSV-1 is a common neurotropic pathogen that can reach the central nervous system without obvious clinical symptoms. HSV-1 can infect the terminals of olfactory neurons and enter the CNS through retrograde axonal transport of neurons, eventually reaching the olfactory bulb in the brain. HSV-1 can also be reactivated from neurons in the trigeminal ganglion and reach the skin or CNS through anterograde transport; after entering the CNS, the virus can either remain dormant in this tissue or be activated to cause severe acute necrotizing encephalitis. Herpes simplex encephalitis (HSE) is one of the most serious manifestations of HSV-1 infection, with a mortality rate of up to 97%. Studies have shown that although the use of the antiviral drug acyclovir (ACV) can reduce the mortality rate of HSE patients, most patients have permanent neurological sequelae after recovery, including symptoms of cognitive, memory and behavioral disorders, and an increased incidence of epilepsy.
HSV-1感染在眼角膜中可引起慢性免疫性炎症性疾病,即单纯疱疹性角膜炎(herpes simplexkeratitis,HSK),该疾病会导致感染者出现不可逆的损伤和失明。在眼部感染期间,病毒从三叉神经节潜伏进入到角膜上皮,通过再激活作用引起炎症反应,患者特征在于角膜混浊和眼球表面新血管形成,由病毒的周期性再激活引起的复发性炎症甚至可能导致失明。研究显示,HVEM是眼部HSV-1感染的免疫发病机制中的重要宿主细胞蛋白质受体。HSV-1 infection can cause a chronic immune inflammatory disease in the cornea, herpes simplex keratitis (HSK), which can cause irreversible damage and blindness in infected people. During ocular infection, the virus enters the corneal epithelium from the trigeminal ganglion latently and causes an inflammatory response through reactivation. Patients are characterized by corneal opacity and new blood vessel formation on the surface of the eye. Recurrent inflammation caused by periodic reactivation of the virus may even lead to blindness. Studies have shown that HVEM is an important host cell protein receptor in the immune pathogenesis of HSV-1 infection in the eye.
目前没有用于预防HSV感染的疫苗。原发性的病毒感染和潜伏感染后的再激活都有可能引起程度不同的慢性炎症过程。临床常用的治疗药物是以口服阿昔洛韦 为代表的核苷类似物。自20世纪80年代以来,ACV及其衍生物就成为预防和治疗疱疹病毒感染的首选药物。耐药毒株病例的不断出现,也为抗病毒治疗提出了更高的要求。由于HSV-1流行基数大,潜伏感染问题严重,这使得人们不得不重视耐药性问题,同时也提示新型药物的研发迫在眉睫。There is currently no vaccine for the prevention of HSV infection. Both primary viral infection and reactivation after latent infection may cause chronic inflammatory processes of varying degrees. Commonly used clinical therapeutic drugs are nucleoside analogs represented by oral acyclovir. Since the 1980s, ACV and its derivatives have become the first choice for the prevention and treatment of herpes virus infections. The continuous emergence of drug-resistant strains has also put forward higher requirements for antiviral treatment. Due to the large prevalence of HSV-1 and the serious problem of latent infection, people have to pay attention to the problem of drug resistance, and it also suggests that the development of new drugs is imminent.
单纯疱疹病毒1型(HSV-1)是神经脑炎和感染性失明的主要原因。然而,目前治疗HSV的药物不能消除三叉神经节潜伏HSV病毒或再激活的病毒。因此,开发针对潜在HSV的新治疗策略已成为当务之急。以CRISPR为主的基因组编辑技术代表了一种通过修改或破坏病毒的遗传物质来对抗病毒感染和复制的有前景的方法。Herpes simplex virus type 1 (HSV-1) is a major cause of neuroencephalitis and infectious blindness. However, current drugs for the treatment of HSV cannot eliminate latent HSV virus or reactivated virus in the trigeminal ganglion. Therefore, the development of new therapeutic strategies against latent HSV has become a priority. CRISPR-based genome editing technology represents a promising approach to combat viral infection and replication by modifying or destroying the viral genetic material.
发明内容Summary of the invention
为了解决现有技术中的不足,本发明旨在提供靶向HSV必需基因的gRNA、CRISPR/Cas基因编辑系统、递送系统及应用。In order to address the deficiencies in the prior art, the present invention aims to provide gRNA, CRISPR/Cas gene editing system, delivery system and application targeting HSV essential genes.
本发明设计并筛选了一系列直接靶向HSV必需基因VP16、ICP27、ICP4或gD的gRNA,VP16(2gRNA),ICP27(2gRNA),ICP4(2gRNA)or gD(4gRNA),并具体提供基于CRISPR-Cas基因编辑抗HSV感染的新技术、方法和递送策略。The present invention designs and screens a series of gRNAs that directly target HSV essential genes VP16, ICP27, ICP4 or gD, VP16 (2gRNA), ICP27 (2gRNA), ICP4 (2gRNA) or gD (4gRNA), and specifically provides new technologies, methods and delivery strategies based on CRISPR-Cas gene editing to resist HSV infection.
本发明的技术方案如下:The technical solution of the present invention is as follows:
本发明第一方面提供一种靶向HSV病毒必需基因的gRNA,所述gRNA选自SEQ ID NO.1-SEQ ID NO.10所示核苷酸序列中的一种。The first aspect of the present invention provides a gRNA targeting an essential gene of the HSV virus, wherein the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
进一步地,所述gRNA选自SEQ ID NO.2、SEQ ID NO.4、SEQ ID NO.6和SEQ ID NO.7所示核苷酸序列中的一种。Furthermore, the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
本发明第二方面提供一种靶向HSV病毒必需基因的gRNA组合物,所述gRNA组合物选自SEQ ID NO.1-SEQ ID NO.10所示核苷酸序列中的两种或以上。A second aspect of the present invention provides a gRNA composition targeting essential genes of the HSV virus, wherein the gRNA composition is selected from two or more of the nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
进一步地,所述gRNA组合物为SEQ ID NO.2、SEQ ID NO.4、SEQ ID NO.6和SEQ ID NO.7所示核苷酸序列的组合物。Furthermore, the gRNA composition is a composition of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
本发明第三方面提供一种靶向HSV病毒必需基因的gRNA表达载体,其包含所述gRNA,或所述gRNA组合物。The third aspect of the present invention provides a gRNA expression vector targeting essential genes of the HSV virus, which comprises the gRNA, or the gRNA composition.
进一步地,所述表达载体为病毒载体或非病毒载体;Furthermore, the expression vector is a viral vector or a non-viral vector;
所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒(VLP);The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
本发明第四方面提供一种CRISPR/Cas基因编辑系统,其包含所述gRNA,或所述gRNA组合物,以及Cas蛋白。A fourth aspect of the present invention provides a CRISPR/Cas gene editing system, which comprises the gRNA, or the gRNA composition, and a Cas protein.
进一步地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白、脑炎双球菌 Cas9、嗜热链球菌Cas9、金黄色葡萄球菌Cas9、氨基酸球菌Cpf1、谷氨酸棒杆菌Cpf1、金黄色葡萄球菌微型SaCas9、口腔纤毛菌C2c2的一种或多种;Further, the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, Cas9 from Neisseria meningitidis, Cas9 from Streptococcus thermophilus, Cas9 from Staphylococcus aureus, Cpf1 from Acidaminococcus, Cpf1 from Corynebacterium glutamicum, micro-SaCas9 from Staphylococcus aureus, and C2c2 from Oral Cibotium;
优选地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白。Preferably, the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
本发明第五方面提供所述CRISPR/Cas基因编辑系统的递送系统,所述递送系统包括:A fifth aspect of the present invention provides a delivery system for the CRISPR/Cas gene editing system, the delivery system comprising:
1)所述的gRNA表达载体;1) the gRNA expression vector;
2)Cas蛋白表达载体。2) Cas protein expression vector.
进一步地,所述Cas蛋白表达载体为病毒载体或非病毒载体;Furthermore, the Cas protein expression vector is a viral vector or a non-viral vector;
所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒(VLP);The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
本发明第六方面提供一种试剂盒,其包括:A sixth aspect of the present invention provides a kit comprising:
1)所述gRNA,或所述gRNA组合物,或所述的gRNA表达载体;1) the gRNA, or the gRNA composition, or the gRNA expression vector;
2)Cas蛋白表达载体。2) Cas protein expression vector.
进一步地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白、脑炎双球菌Cas9、嗜热链球菌Cas9、金黄色葡萄球菌Cas9、氨基酸球菌Cpf1、谷氨酸棒杆菌Cpf1、金黄色葡萄球菌微型SaCas9、口腔纤毛菌C2c2的一种或多种;Further, the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, Cas9 from Neisseria meningitidis, Cas9 from Streptococcus thermophilus, Cas9 from Staphylococcus aureus, Cpf1 from Acidaminococcus, Cpf1 from Corynebacterium glutamicum, mini-SaCas9 from Staphylococcus aureus, and C2c2 from Oral Cibotium;
优选地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白。Preferably, the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
进一步地,所述Cas蛋白表达载体为病毒载体或非病毒载体;Furthermore, the Cas protein expression vector is a viral vector or a non-viral vector;
所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒(VLP);The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle (VLP);
所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
本发明第七方面提供所述gRNA,或所述gRNA组合物,或所述的gRNA表达载体,或所述CRISPR/Cas基因编辑系统,或所述递送系统,或所述试剂盒在制备预防和/或治疗HSV感染相关疾病的药物中的应用。The seventh aspect of the present invention provides the use of the gRNA, or the gRNA composition, or the gRNA expression vector, or the CRISPR/Cas gene editing system, or the delivery system, or the kit in the preparation of drugs for preventing and/or treating HSV infection-related diseases.
进一步地,所述HSV感染相关疾病为HSV-1型或HSV-2型相关疾病;Furthermore, the HSV infection-related disease is an HSV-1 or HSV-2-related disease;
优选地,所述HSV感染相关疾病包括病毒性脑炎、单纯疱疹病毒性角膜炎和感染性失明。Preferably, the HSV infection-related diseases include viral encephalitis, herpes simplex virus keratitis and infectious blindness.
本发明第八方面提供所述gRNA,或所述gRNA组合物,或所述的gRNA表达载体,或所述CRISPR/Cas基因编辑系统,或所述递送系统,或所述试剂盒在制备预防和/或治疗抗HSV药物中的应用。The eighth aspect of the present invention provides the use of the gRNA, or the gRNA composition, or the gRNA expression vector, or the CRISPR/Cas gene editing system, or the delivery system, or the kit in the preparation of drugs for preventing and/or treating anti-HSV.
进一步地,所述HSV为HSV-1或HSV-2。Furthermore, the HSV is HSV-1 or HSV-2.
本发明第九方面提供一种药物组合物,其包括所述CRISPR/Cas基因编辑系统的递送系统。The ninth aspect of the present invention provides a pharmaceutical composition comprising a delivery system for the CRISPR/Cas gene editing system.
本发明第十方面提供一种抑制HSV复制和/或HSV病毒必需基因表达的方法,所述方法包括:在HSV感染前或感染后,将所述CRISPR/Cas基因编辑系统的递送系统转染目的细胞,细胞表达的Cas-gRNA形成的核糖核蛋白复合物靶向的精准剪切破坏HSV基因组,阻断病毒复制和传播。The tenth aspect of the present invention provides a method for inhibiting HSV replication and/or expression of HSV essential genes, the method comprising: before or after HSV infection, transfecting the target cells with the delivery system of the CRISPR/Cas gene editing system, and the ribonucleoprotein complex formed by the Cas-gRNA expressed by the cells targetedly and precisely shears and destroys the HSV genome, thereby blocking viral replication and transmission.
本发明的有益效果为:The beneficial effects of the present invention are:
本发明提供一种利用CRISPR-Cas基因组编辑技术抗单纯疱疹病毒感染的新技术,针对HSV临床株基因组序列,设计并筛选了一系列直接靶向HSV必需基因VP16、ICP27、ICP4或gD的gRNA,利用基因治疗病毒载体或非病毒载体递送核酸酶Cas9和靶向病毒基因的gRNA。HSV关键基因靶向的基因编辑可以有效抑制HSV复制和传播感染,每一个gRNA单独介导均能有效的抑制HSV病毒复制,其中的aVP16-2、aICP27-2、aICP4-2或agD-1的gRNA单独编辑抑制HSV病毒复制较同组效果更好,gRNA两两组合的基因编辑能更有效的抑制HSV病毒复制,aVP16-2、aICP27-2、aICP4-2和agD-1四种gRNA混合基因编辑(命名Cocktail)抑制HSV病毒复制效果优于单gRNA介导的基因编辑,gRNA联用鸡尾酒策略显示出优于单靶点编辑的效果。动物实验中病毒载体递送的CRISPR-Cas9/gRNA在体基因编辑可有效阻断HSV复制和传播。The present invention provides a new technology for resisting herpes simplex virus infection by using CRISPR-Cas genome editing technology. A series of gRNAs directly targeting HSV essential genes VP16, ICP27, ICP4 or gD are designed and screened for the genome sequence of HSV clinical strains, and a gene therapy viral vector or a non-viral vector is used to deliver nuclease Cas9 and gRNA targeting viral genes. Gene editing targeted to HSV key genes can effectively inhibit HSV replication and spread infection, and each gRNA can effectively inhibit HSV viral replication when mediated alone. Among them, the gRNAs of aVP16-2, aICP27-2, aICP4-2 or agD-1 can inhibit HSV viral replication better than the same group, and the gene editing of gRNAs in pairs can more effectively inhibit HSV viral replication. The mixed gene editing (named Cocktail) of four gRNAs of aVP16-2, aICP27-2, aICP4-2 and agD-1 inhibits HSV viral replication better than the gene editing mediated by a single gRNA, and the cocktail strategy of gRNA combination shows an effect better than single target editing. In animal experiments, in vivo gene editing using CRISPR-Cas9/gRNA delivered by viral vectors can effectively block HSV replication and transmission.
本发明应用gRNA混合基因编辑(Cocktail)的方法,不论是预防策略,或治疗策略Cocktail均能有效的抑制HSV病毒复制,以及组织间传播。HSV感染后gRNA混合基因编辑(Cocktail)治疗的时间越早,抑制效果越明显。目前治疗HSV的药物不能消除三叉神经节潜伏的HSV病毒或病毒复发,通过本发明方法和病毒载体介导的基因治疗可以有效消除潜伏感染或复发感染的HSV病毒,为难治性HSV-1相关疾病的治疗提供潜在重要手段。The present invention uses the method of gRNA mixed gene editing (Cocktail), whether it is a prevention strategy or a treatment strategy Cocktail, which can effectively inhibit HSV virus replication and inter-tissue transmission. The earlier the gRNA mixed gene editing (Cocktail) treatment is after HSV infection, the more obvious the inhibitory effect. The current drugs for treating HSV cannot eliminate the HSV virus lurking in the trigeminal ganglion or the virus relapses. The gene therapy mediated by the method of the present invention and viral vectors can effectively eliminate latently infected or recurrently infected HSV viruses, providing a potential important means for the treatment of refractory HSV-1 related diseases.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1为CRISPR基因编辑的HSV靶基因的筛选及gRNA的设计图;FIG1 is a diagram showing the screening of HSV target genes for CRISPR gene editing and the design of gRNA;
图2为CRISPR Cas9/gRNA质粒的鉴定图;Figure 2 is an identification diagram of CRISPR Cas9/gRNA plasmid;
图3为Cas9/gRNA单质粒转染及抗HSV复制效果图;Figure 3 is a diagram showing the effect of Cas9/gRNA single plasmid transfection and anti-HSV replication;
图4为Cas9/gRNA单质粒转染编辑后的细胞上清病毒滴度测定;Figure 4 is the determination of virus titer in the cell supernatant after Cas9/gRNA single plasmid transfection editing;
图5为Cas9/gRNA质粒优化双质粒联用策略设计及抗HSV复制效果图;Figure 5 is a diagram showing the design of the Cas9/gRNA plasmid optimization dual-plasmid combination strategy and the anti-HSV replication effect;
图6为优筛的四类gRNA联用(Cocktail)策略及细胞水平抗HSV复制效果图;Figure 6 is a diagram showing the cocktail strategy of four types of gRNAs of YouScreen and the anti-HSV replication effect at the cellular level;
图7为Cocktail联用策略编辑后的细胞上清病毒滴度测定;FIG7 is a determination of virus titer in the cell supernatant after editing using the cocktail combination strategy;
图8为Cocktail多靶点编辑预防和治疗效果比较;Figure 8 is a comparison of the preventive and therapeutic effects of Cocktail multi-target editing;
图9为基因递送Cas9/gRNA慢病毒的包装制备及抗HSV效果图;FIG9 is a diagram showing the packaging preparation of gene delivery Cas9/gRNA lentivirus and its anti-HSV effect;
图10为LV-Cas9/gRNA对照病毒不能抑制HSV复制;Figure 10 shows that the LV-Cas9/gRNA control virus cannot inhibit HSV replication;
图11为LV-Cas9/gRNA单病毒在体抗HSV复制效果图;Figure 11 is a diagram showing the effect of LV-Cas9/gRNA single virus on anti-HSV replication in vivo;
图12为LV-Cas9/gRNA病毒联用(Cocktail)策略抗HSV复制;Figure 12 shows the LV-Cas9/gRNA virus cocktail strategy against HSV replication;
图13为LV-Cas9/gRNA病毒联用(Cocktail)剂量效应;FIG13 shows the dose effect of LV-Cas9/gRNA virus combination (Cocktail);
图14为LV-Cas9/gRNA病毒联用(Cocktail)治疗HSV感染效果图。Figure 14 is a diagram showing the effect of LV-Cas9/gRNA virus combination (Cocktail) in treating HSV infection.
具体实施方式Detailed ways
下面结合附图和实施方式对本发明进行详细说明。如未特别说明,均为本领域的常规技术。所述试剂或材料,如未特别说明,均来源于商业渠道。The present invention is described in detail below in conjunction with the accompanying drawings and embodiments. Unless otherwise specified, all are conventional techniques in the art. The reagents or materials, unless otherwise specified, are derived from commercial channels.
本发明设计并筛选了一系列直接靶向HSV必需基因VP16、ICP27、ICP4或gD的gRNA,VP16(2种gRNA),ICP27(2种gRNA),ICP4(2种gRNA)or gD(4种gRNA)。基于CRISPR/Cas9基因编辑技术,本发明应用载体递送Cas9/gRNA至目的细胞,实现对靶向基因的编辑,载体包括常用的病毒载体和非病毒载体,其中病毒载体包括慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体、病毒样颗粒(VLP)等;非病毒载体包括质粒、mRNA等。The present invention designs and screens a series of gRNAs that directly target HSV essential genes VP16, ICP27, ICP4 or gD, VP16 (2 gRNAs), ICP27 (2 gRNAs), ICP4 (2 gRNAs) or gD (4 gRNAs). Based on CRISPR/Cas9 gene editing technology, the present invention uses vectors to deliver Cas9/gRNA to target cells to achieve editing of targeted genes. The vectors include commonly used viral vectors and non-viral vectors, wherein viral vectors include lentiviral vectors, retroviral vectors, adeno-associated viral vectors, herpes simplex virus vectors, adenoviral vectors, virus-like particles (VLPs), etc.; non-viral vectors include plasmids, mRNA, etc.
在一个具体实施方案中,本发明构建了10种携带SpCas9和相应gRNA的慢病毒载体核心质粒,接着包装制备了基因递送CRISPR编辑系统的慢病毒载体。首先在细胞实验上筛选了10种SpCas9/gRNA的抑制HSV效果,感染H129-GFP病毒后2天,对照组(NC和Vec)组病毒迅速增殖、几乎所有的细胞均表达绿色荧光蛋白,而靶向切割4种基因的10种gRNA组荧光比例均有不同程度的下降;通过检测细胞上清HSV病毒滴度,实验组病毒滴度均显著低于对照组,筛选出agD-1,aICP4-2,aVP16-2,aICP27-2四种gRNA组的抑制效果最明显,从而作为包装慢病毒载体的备选。In a specific embodiment, the present invention constructs 10 lentiviral vector core plasmids carrying SpCas9 and corresponding gRNA, and then packages and prepares lentiviral vectors of gene delivery CRISPR editing system. First, the inhibitory effects of 10 SpCas9/gRNAs were screened in cell experiments. Two days after infection with H129-GFP virus, the virus in the control group (NC and Vec) rapidly proliferated, and almost all cells expressed green fluorescent protein, while the fluorescence ratios of the 10 gRNA groups targeting the cutting of 4 genes decreased to varying degrees; by detecting the HSV virus titer of the cell supernatant, the virus titer of the experimental group was significantly lower than that of the control group, and the inhibitory effects of the four gRNA groups of agD-1, aICP4-2, aVP16-2, and aICP27-2 were screened out as the most obvious, thus serving as an alternative for packaging lentiviral vectors.
此外,联合gRNA策略(“鸡尾酒”策略)明显显示出优于单gRNA基因编辑的效果。本发明测试了靶向编辑4种基因的Cas9/gRNA联用能否达到更好的抑制HSV效果。将不同浓度(0.5μg,1μg,2μg)的gRNA质粒联用(Cocktail)转染细胞,然后HSV-GFP病毒按MOI=0.01和MOI=0.001分别感染转染后的细胞,发现Cocktail组在低剂量即表现出明显的抑制HSV复制效果,而且存在明显的剂量效应,随着Cas9/gRNA质粒的量增多,抑制病毒增殖的效力越强这些结果表明靶向编辑4种基因的Cas9/gRNA联合策略能高效地抑制HSV病毒复制和增殖,而且表现出明显的剂量效应。In addition, the combined gRNA strategy ("cocktail" strategy) clearly showed superior effects over single gRNA gene editing. The present invention tests whether the combination of Cas9/gRNA for targeted editing of 4 genes can achieve better HSV inhibition effects. Cells were transfected with gRNA plasmids of different concentrations (0.5μg, 1μg, 2μg) in combination (Cocktail), and then the HSV-GFP virus was infected with the transfected cells at MOI=0.01 and MOI=0.001, respectively. It was found that the Cocktail group showed a significant inhibitory effect on HSV replication at low doses, and there was an obvious dose effect. As the amount of Cas9/gRNA plasmids increased, the stronger the effect of inhibiting viral proliferation. These results show that the combined Cas9/gRNA strategy for targeted editing of 4 genes can effectively inhibit HSV viral replication and proliferation, and show a significant dose effect.
针对CRISPR编辑系统的治疗效果如何问题,本发明做了先感染HSV病毒后用CRISPR编辑系统治疗的实验,分别在提前12小时、6小时和1小时感染HSV- GFP病毒,然后转染Cas9/gRNA-cocktail质粒,发现在HSV病毒急性感染的高速增殖阶段(病毒增殖12小时)Cas9/gRNA干预有一定抑制效果但不明显,如果HSV感染后更早干预-病毒增殖6小时组和病毒增殖1小时组CRISPR治疗均能显著的降低HSV病毒增殖,而且越早干预越好。In response to the question of the therapeutic effect of the CRISPR editing system, the present invention conducted an experiment of first infecting the HSV virus and then treating it with the CRISPR editing system. The HSV-GFP virus was infected 12 hours, 6 hours and 1 hour in advance, respectively, and then the Cas9/gRNA-cocktail plasmid was transfected. It was found that during the high-speed proliferation stage of acute HSV virus infection (12 hours of viral proliferation), Cas9/gRNA intervention had a certain inhibitory effect but it was not obvious. If intervention was given earlier after HSV infection - CRISPR treatment in the 6-hour viral proliferation group and the 1-hour viral proliferation group could significantly reduce the proliferation of HSV virus, and the earlier the intervention, the better.
本发明利用包装的慢病毒在体开展了动物实验,在体实验中Cas9/gRNA联用(Cocktail)策略能高效地抑制病毒增殖和传播。在未做任何干预的对照组(NC),以及慢病毒空载对照组(Vec)中在小鼠V1脑区注射H129-GFP后5天病毒已高效的增殖,并沿神经环路传播到LGd等脑区,感染的神经元被荧光蛋白高亮标记,慢病毒空载给药无治疗效果。当用携带表达Cas9/gRNA的慢病毒单独治疗时均表现出抑制病毒复制(注射部位荧光细胞少)和病毒传播(LGd脑区无标记)的效果,相较LV-aICP4和LV-aICP27两组的效果优于LV-gD和LV-aVP16。Cas9/gRNA联用(Cocktail)治疗效果相对最好,在注射部位V1观察不到或仅有少量细胞被感染标记,而且也呈现剂量效应。另外重要的本发明也验证了“先感染后治疗”的实验,在小鼠V1脑区先感染H129-GFP,1天后给药慢病毒Cocktail治疗发现能有效地抑制HSV复制传播,效果优于单慢病毒给药。The present invention uses packaged lentivirus to carry out animal experiments in vivo. In the in vivo experiment, the Cas9/gRNA combination (Cocktail) strategy can effectively inhibit virus proliferation and spread. In the control group (NC) without any intervention, and the lentivirus empty control group (Vec), the virus has been efficiently proliferated 5 days after H129-GFP was injected into the mouse V1 brain region, and spread to brain regions such as LGd along the neural circuit. The infected neurons are highlighted by fluorescent protein, and the lentivirus empty administration has no therapeutic effect. When the lentivirus carrying the expression of Cas9/gRNA is used for treatment alone, the effects of inhibiting viral replication (few fluorescent cells at the injection site) and viral spread (no mark in the LGd brain region) are shown, and the effects of the two groups of LV-aICP4 and LV-aICP27 are better than LV-gD and LV-aVP16. The Cas9/gRNA combination (Cocktail) treatment effect is relatively the best, and no or only a small number of cells are observed to be infected and marked at the injection site V1, and it also shows a dose effect. Another important thing about the present invention is that it also verifies the "infection first, then treatment" experiment, where the mouse V1 brain region is first infected with H129-GFP, and then lentivirus cocktail treatment is administered one day later. It is found that the replication and spread of HSV can be effectively inhibited, and the effect is better than single lentivirus administration.
靶向HSV四个关键基因VP16,ICP27,ICP4,gD的CRISPR基因编辑能有效地抑制HSV在裂解性复制周期的复制和增殖,而且Cas9/gRNA联用(Cocktail)策略更高效,可以想见利用CRISPR基因组编辑技术清除潜伏感染状态、病毒滴度低得多的latent HSV病毒,抑制作用可能更显著。CRISPR gene editing targeting the four key genes of HSV, VP16, ICP27, ICP4, and gD, can effectively inhibit the replication and proliferation of HSV in the lytic replication cycle, and the Cas9/gRNA combination (Cocktail) strategy is more efficient. It can be imagined that the use of CRISPR genome editing technology to eliminate latent HSV viruses in a latent infection state with much lower viral titers may have a more significant inhibitory effect.
综上,本发明的Cas9/gRNA基因组编辑能高效的抑制HSV在预防水平和治疗水平的感染和传播,特别在动物活体实验中作用明显,相较对照组Cas9/gRNA不管是单独给药,或是Cocktail联合给药HSV的增殖显著被抑制,标记的神经细胞数量相较对照组存在指数级的下降。另外Cas9/gRNA联用(Cocktail)治疗手段并未观察到相关的毒副作用,说明该策略的安全性。这项使用CRISPR基因组编辑来消除HSV感染的方法为难治性HSV-1相关疾病的潜在治疗提供新的手段,尤其是当传统治疗方法无效时。In summary, the Cas9/gRNA genome editing of the present invention can effectively inhibit the infection and spread of HSV at the prevention level and the treatment level, especially in the in vivo animal experiment. Compared with the control group, whether Cas9/gRNA is administered alone or in combination with Cocktail, the proliferation of HSV is significantly inhibited, and the number of labeled nerve cells decreases exponentially compared with the control group. In addition, no related toxic side effects were observed in the Cas9/gRNA combination (Cocktail) treatment, indicating the safety of this strategy. This method of eliminating HSV infection using CRISPR genome editing provides a new means for the potential treatment of refractory HSV-1 related diseases, especially when traditional treatments are ineffective.
下面结合实施例对本发明进行详细说明。The present invention is described in detail below with reference to the embodiments.
实施例1:CRISPR基因编辑的HSV靶基因的筛选及gRNA的设计合成Example 1: Screening of HSV target genes for CRISPR gene editing and design and synthesis of gRNA
HSV病毒必需基因VP16、ICP27、ICP4和gD被选择为CRISPR/Cas9策略编辑的潜在靶点。本实施例设计并靶向了4个病毒必需基因,并开发了与Cas9结合的引导RNA,以测试它们抑制HSV-1复制的能力。HSV essential genes VP16, ICP27, ICP4 and gD were selected as potential targets for CRISPR/Cas9 strategy editing. In this example, four essential viral genes were designed and targeted, and guide RNAs combined with Cas9 were developed to test their ability to inhibit HSV-1 replication.
基于选定基因的靶位点序列设计sgRNA,并通过CRISPR DESIGN系统筛选(http://crispr.mit.edu/).sgRNA寡核苷酸序列如表1所示,共设计了10个gRNA:VP16靶向(2gRNA,序列见SEQ ID NO.1和2)、ICP27靶向(2gRNA,序列见 SEQ ID NO.3和4),ICP4靶向(2gRNA,序列见SEQ ID NO.5和6)和gD靶向(4gRNA,序列见SEQ ID NO.7-10)(图1,表1)。构建并验证了携带SpCas9和gRNA的慢病毒载体的相应10个核心质粒(图1)并包装了相应的慢病毒。sgRNA was designed based on the target site sequence of the selected gene and screened by CRISPR DESIGN system (http://crispr.mit.edu/). The sgRNA oligonucleotide sequences are shown in Table 1. A total of 10 gRNAs were designed: VP16 targeting (2 gRNAs, sequences see SEQ ID NO.1 and 2), ICP27 targeting (2 gRNAs, sequences see SEQ ID NO.3 and 4), ICP4 targeting (2 gRNAs, sequences see SEQ ID NO.5 and 6) and gD targeting (4 gRNAs, sequences see SEQ ID NO.7-10) (Figure 1, Table 1). The corresponding 10 core plasmids of lentiviral vectors carrying SpCas9 and gRNA were constructed and verified (Figure 1) and the corresponding lentivirus was packaged.
表1 CRISPR/Cas9编辑的HSV1基因和设计的sgRNA序列Table 1 CRISPR/Cas9-edited HSV1 gene and designed sgRNA sequences
Figure PCTCN2022135820-appb-000001
Figure PCTCN2022135820-appb-000001
实施例2:CRISPR Cas9/gRNA慢病毒核心质粒的构建Example 2: Construction of CRISPR Cas9/gRNA lentiviral core plasmid
sgRNA是根据所选基因的靶点序列设计的,并通过CRISPR设计系统(http://crispr.mit.edu/)进行筛选。sgRNA寡核苷酸序列如表1。使用BsmBI对空载体lentiCRISPR v2载体(addgene,#52961,序列见SEQ ID NO.11)进行消化,并将退火的寡核苷酸克隆到单导RNA支架上,获得CRISPR Cas9/gRNA慢病毒核心质粒。然后在HEK293T细胞中用包装质粒生产lentiCRISPR-sgRNA病毒,如以前描述的方法(Cold Spring Harb Protoc.2018 Apr 2;2018(4))。简而言之,将上述len-tivirus构建体与三种慢病毒包装载体(pGAG/POL、pREV和pMD2.G)共同转染到293T细胞中,2至3天后收集上清液,获得靶向病毒。生产的五种慢病毒(LV-vec,LV-agD-1,LV-aVP16-2,LV-aICP4-2,LV-aICP27-2)病毒储备的滴度均为1×10 8PFU/ml。 sgRNA was designed according to the target sequence of the selected gene and screened by the CRISPR design system (http://crispr.mit.edu/). The sgRNA oligonucleotide sequences are shown in Table 1. The empty vector lentiCRISPR v2 vector (addgene, #52961, sequence see SEQ ID NO.11) was digested with BsmBI, and the annealed oligonucleotides were cloned into the single guide RNA scaffold to obtain the CRISPR Cas9/gRNA lentiviral core plasmid. The packaging plasmid was then used to produce lentiCRISPR-sgRNA viruses in HEK293T cells as previously described (Cold Spring Harb Protoc. 2018 Apr 2; 2018 (4)). In brief, the above lentivirus constructs were co-transfected into 293T cells with three lentiviral packaging vectors (pGAG/POL, pREV, and pMD2.G), and the supernatant was collected 2 to 3 days later to obtain the targeted virus. The titers of the virus stocks of the five lentiviruses produced (LV-vec, LV-agD-1, LV-aVP16-2, LV-aICP4-2, and LV-aICP27-2) were all 1×10 8 PFU/ml.
实施例3:CRISPR Cas9/gRNA质粒的鉴定Example 3: Identification of CRISPR Cas9/gRNA plasmid
质粒构建好后,送测序公司测序,测序结果成功。同时,用质粒作为模板扩增 目的片段,反应体系和PCR程序如下:After the plasmid was constructed, it was sent to a sequencing company for sequencing, and the sequencing result was successful. At the same time, the plasmid was used as a template to amplify the target fragment. The reaction system and PCR program were as follows:
Figure PCTCN2022135820-appb-000002
Figure PCTCN2022135820-appb-000002
PCR程序:PCR procedure:
Figure PCTCN2022135820-appb-000003
Figure PCTCN2022135820-appb-000003
PCR程序结束后,配制1%琼脂糖凝胶,以180V在TAE电泳液中跑胶23min,结果见图2。十个质粒的目的片段大小一致,鉴定正确。After the PCR program was completed, 1% agarose gel was prepared and run in TAE electrophoresis buffer at 180V for 23 minutes. The results are shown in Figure 2. The target fragments of the ten plasmids were consistent in size and were identified correctly.
实施例4:Cas9/gRNA单质粒转染及抗HSV复制效果Example 4: Cas9/gRNA single plasmid transfection and anti-HSV replication effect
收集对数生长期的BHK细胞,以密度为1×10 6个/mL接种六孔板。待细胞贴壁后,将细胞培养基更换为400μL Opti-MEM无血清培养基。准备1.5mL无菌EP管,分别加入2μg Cas9/gRNA单质粒,50μL Opti-MEM培养基轻柔摇匀混合,在室温下温育5min。再向无菌EP管中分别加入4μL Lipofectamine 2000试剂,50μL Opti-MEM培养基,轻柔摇匀混合,在室温下静置15min。吸取质粒与Lipofectamine 2000混合液100μL至板孔中,混匀,于37℃,5%CO 2细胞培养箱中培养。转染24h后,弃细胞上清。每孔以MOI=0.1加100μL HSV 129-EGFP感染,吸附1h后弃病毒液,加2mL含2%血清的DMEM培养基,于35℃,3%CO 2细胞培养箱中培养48h。用荧光显微镜观察细胞,并拍照。 BHK cells in the logarithmic growth phase were collected and inoculated into six-well plates at a density of 1×10 6 cells/mL. After the cells adhered to the wall, the cell culture medium was replaced with 400μL Opti-MEM serum-free medium. Prepare a 1.5mL sterile EP tube, add 2μg Cas9/gRNA single plasmid, 50μL Opti-MEM medium, gently shake and mix, and incubate at room temperature for 5min. Then add 4μL Lipofectamine 2000 reagent and 50μL Opti-MEM medium to the sterile EP tube, shake and mix gently, and let stand at room temperature for 15min. Pipette 100μL of the mixture of plasmid and Lipofectamine 2000 into the plate wells, mix well, and culture in a cell culture incubator at 37℃, 5% CO 2. After transfection for 24h, discard the cell supernatant. Each well was infected with 100 μL HSV 129-EGFP at MOI = 0.1, and the virus solution was discarded after adsorption for 1 hour, and 2 mL of DMEM medium containing 2% serum was added, and cultured in a cell culture incubator at 35°C, 3% CO 2 for 48 hours. The cells were observed under a fluorescence microscope and photographed.
收集病毒上清,通过蚀斑法测病毒滴度。取对数生长期的BHK细胞,以密度为2×10 5个/mL接种12孔板,待细胞贴壁。将病毒上清以10倍稀释6次,依次吸取100μL稀释的病毒上清加入孔板中,吸附1h后弃病毒液,加入等体积混合的4%DMEM培养基和1%的琼脂糖溶液。静置,待琼脂糖浓度后置于35℃,3%CO 2细胞培养箱中培养24h,于荧光显微镜观察并数斑。结果如图3所示,与对照组相比Cas9/gRNA单质粒转染后绿色荧光蛋白的表达量降低。 The virus supernatant was collected and the virus titer was measured by plaque assay. BHK cells in the logarithmic growth phase were taken and inoculated into 12-well plates at a density of 2×10 5 /mL, and the cells were allowed to adhere to the wall. The virus supernatant was diluted 10 times 6 times, and 100 μL of the diluted virus supernatant was successively drawn into the well plate. After adsorption for 1 hour, the virus liquid was discarded, and an equal volume of 4% DMEM culture medium and 1% agarose solution were added. After standing, the agarose concentration was placed in a 35°C, 3% CO 2 cell culture incubator for 24 hours, and the spots were observed and counted under a fluorescence microscope. The results are shown in Figure 3. Compared with the control group, the expression of green fluorescent protein was reduced after Cas9/gRNA single plasmid transfection.
此外,与对照组相比,HSV的滴度是Cas9/gRNA有效性的另一种量度,如图4,在Cas9/gRNA转染组中显示了HSV滴度的显着降低。其中,agD-1,aICP4-2, aICP27-2和aVP16-2对HSV的增殖具有最佳的抑制作用,因此选择了这些Cas9/gRNA质粒进行后续实验和慢病毒包装。In addition, the titer of HSV is another measure of the effectiveness of Cas9/gRNA compared with the control group, as shown in Figure 4, which shows a significant reduction in the HSV titer in the Cas9/gRNA transfection group. Among them, agD-1, aICP4-2, aICP27-2, and aVP16-2 had the best inhibitory effect on the proliferation of HSV, so these Cas9/gRNA plasmids were selected for subsequent experiments and lentiviral packaging.
实施例5:Cas9/gRNA质粒优化双质粒联用策略设计及抗HSV复制效果Example 5: Cas9/gRNA plasmid optimization dual plasmid combination strategy design and anti-HSV replication effect
选取案例4中对HSV复制效果抑制效率最高的四种Cas9/gRNA质粒agD-1,aICP4-2,aICP27-2,aVP16-g2两两组合(agD-g1+aICP4-g2;agD-g1+aICP27-g2;agD-g1+aVP16-g2;aICP27-g2+aICP4-g2;aVP16-g2+aICP4-g2;aICP27-g2+aVP16-g2;)后一起转染BHK细胞,24小时后以MOI=0.1进行感染,吸附1h后弃病毒液,加2mL含2%血清的DMEM培养基,于35℃,3%CO 2细胞培养箱中培养48h。用荧光显微镜观察细胞,并成像。结果如图5所示,与对照组相比优化的双质粒组联合转染后绿色荧光蛋白的表达量降低。 The four Cas9/gRNA plasmids agD-1, aICP4-2, aICP27-2, and aVP16-g2 with the highest inhibition efficiency on HSV replication in Case 4 were selected and combined in pairs (agD-g1+aICP4-g2; agD-g1+aICP27-g2; agD-g1+aVP16-g2; aICP27-g2+aICP4-g2; aVP16-g2+aICP4-g2; aICP27-g2+aVP16-g2;) and transfected into BHK cells together. After 24 hours, the cells were infected at an MOI of 0.1. After adsorption for 1 hour, the virus solution was discarded, 2 mL of DMEM medium containing 2% serum was added, and the cells were cultured in a cell culture incubator at 35°C and 3% CO2 for 48 hours. The cells were observed and imaged using a fluorescence microscope. As shown in Figure 5, the expression of green fluorescent protein was reduced after the optimized double-plasmid group was combined and transfected compared with the control group.
实施例6:优筛的四类gRNA联用(Cocktail)策略及细胞水平抗HSV复制效果Example 6: Cocktail strategy of four types of gRNAs of YouScreen and anti-HSV replication effect at the cellular level
将四种质粒混合后按浓度梯度转染BHK细胞,每种质粒各转染0.5μg、1μg和2μg,24H后分别以MOI=0.001和MOI=0.01感染HSV H129-EGFP,在48小时后成像细胞荧光表达情况,并测定上清病毒滴度。结果如图6和7。从HSV滴度或报告基因绿色荧光蛋白的表达丰度的来看,Cocktail策略都可以有效抑制HSV的增殖,HSV感染MOI=0.01或0.001,在2d.p.i可以看到明显的趋势。此外,不同剂量组之间没有检测到病毒滴度的明显的统计差异,表明在这组实验中,Cocktail策略没有剂量作用。The four plasmids were mixed and transfected into BHK cells according to a concentration gradient. Each plasmid was transfected with 0.5μg, 1μg and 2μg. After 24 hours, HSV H129-EGFP was infected at MOI=0.001 and MOI=0.01, respectively. Cell fluorescence expression was imaged after 48 hours, and the supernatant virus titer was measured. The results are shown in Figures 6 and 7. From the perspective of HSV titer or the expression abundance of the reporter gene green fluorescent protein, the cocktail strategy can effectively inhibit the proliferation of HSV. HSV infection MOI=0.01 or 0.001, a clear trend can be seen at 2d.p.i. In addition, no significant statistical difference in virus titer was detected between different dose groups, indicating that the cocktail strategy had no dose effect in this set of experiments.
实施例7:Cocktail多靶点编辑预防和治疗效果比较Example 7: Comparison of preventive and therapeutic effects of cocktail multi-target editing
为了模仿HSV感染的患者的治疗,首先将细胞感染HSV H129-GFP(MOI=0.01),然后分别在HSV感染后1H,6H和12H与四个gRNA质粒混合共转染。与6H.P.I和12H.P.I.相比,在1H.P.I中施用质粒表达Cas9/gRNA显示出最佳的抑制作用。而Cocktail多靶点编辑预防组在病毒感染前24小时表达Cas9/gRNA同样显示出较好的抑制作用。如图8。To mimic the treatment of HSV-infected patients, cells were first infected with HSV H129-GFP (MOI=0.01), and then co-transfected with four gRNA plasmids at 1H, 6H, and 12H after HSV infection. Compared with 6H.P.I and 12H.P.I., the best inhibitory effect was shown by the plasmid expression of Cas9/gRNA in 1H.P.I. The Cocktail multi-target editing prevention group expressed Cas9/gRNA 24 hours before viral infection and also showed good inhibitory effect. As shown in Figure 8.
实施例8:基因递送Cas9/gRNA慢病毒的包装制备Example 8: Packaging and preparation of gene delivery Cas9/gRNA lentivirus
1)转染前24h,用胰蛋白酶消化对数生长期的293T细胞,以含10%血清的培养基调整细胞密度为1.2 x 10 7细胞/20ml,重新接种于15cm细胞培养皿,37℃、5%CO 2培养箱内培养。24h待细胞密度达70%~80%时即可用于转染。细胞状态对于病毒包装至关重要,因此需要保证良好的细胞状态和较少的传代次数。 1) 24 hours before transfection, digest the 293T cells in the logarithmic growth phase with trypsin, adjust the cell density to 1.2 x 107 cells/20ml with a medium containing 10% serum, re-inoculate in a 15cm cell culture dish, and culture in a 37℃, 5% CO2 incubator. When the cell density reaches 70% to 80% after 24 hours, it can be used for transfection. The cell state is crucial for virus packaging, so it is necessary to ensure a good cell state and a low number of passages.
2)转染前2h将细胞培养基更换为无血清培养基。2) 2 h before transfection, the cell culture medium was replaced with serum-free medium.
3)向一灭菌离心管中加入:3) Add to a sterile centrifuge tube:
所制备的各DNA溶液(pGC-LV载体20μg,pHelper 1.0载体15μg,pHelper2.0载体10μg),与相应体积的Opti-MEM混合均匀,调整总体积为2.5ml,在室温下温育5分钟。Each prepared DNA solution (20 μg of pGC-LV vector, 15 μg of pHelper 1.0 vector, and 10 μg of pHelper 2.0 vector) was mixed evenly with the corresponding volume of Opti-MEM, and the total volume was adjusted to 2.5 ml. The solution was incubated at room temperature for 5 minutes.
4)将Lipofectamine 2000试剂轻柔摇匀,取100μl Lipofectamine 2000试剂在另一管中与2.4ml Opti-MEM混合,在室温下温育5分钟。4) Gently shake the Lipofectamine 2000 reagent, take 100 μl of Lipofectamine 2000 reagent and mix it with 2.4 ml Opti-MEM in another tube, and incubate at room temperature for 5 minutes.
5)把稀释后的DNA与稀释后的Lipofectamine 2000进行混合,轻轻地颠倒混匀,不要振荡。必须在5分钟之内混合。5) Mix the diluted DNA with the diluted Lipofectamine 2000 and gently invert to mix without shaking. Mixing must be done within 5 minutes.
6)混合后,在室温下温育【静置】15~20分钟,以便形成DNA与Lipofectamine 2000稀释液的转染复合物。6) After mixing, incubate at room temperature for 15 to 20 minutes to form a transfection complex of DNA and Lipofectamine 2000 dilution solution.
7)将DNA与Lipofectamine 2000混合液转移至293T细胞的培养液中,混匀,于37℃,5%CO2细胞培养箱中培养。7) Transfer the mixture of DNA and Lipofectamine 2000 into the culture medium of 293T cells, mix well, and culture in a cell culture incubator at 37°C, 5% CO2.
8)培养4~6h后倒去含有转染混和物的培养基,每培养皿细胞中加入含10%血清的细胞培养基10ml,于37℃、5%CO 2培养箱内继续培养48小时。后收上清获得包装好的慢病毒。 8) After culturing for 4 to 6 hours, the culture medium containing the transfection mixture was discarded, 10 ml of cell culture medium containing 10% serum was added to each culture dish cell, and the cells were cultured in a 37°C, 5% CO 2 incubator for 48 hours. The supernatant was then collected to obtain the packaged lentivirus.
之后使用了293T细胞(人肾上皮细胞系)进一步验证设计靶向HSV复制的表达Cas9/gRNA的慢病毒的抑制作用。结果与BHK细胞上的质粒效果一致,结果不仅表明Cas9/gRNA编辑可以有效地抑制不同细胞系中的HSV复制,包装的慢病毒同样有较好效果。如图9。293T cells (human kidney epithelial cell line) were then used to further verify the inhibitory effect of the lentivirus expressing Cas9/gRNA designed to target HSV replication. The results were consistent with the plasmid effect on BHK cells. The results not only showed that Cas9/gRNA editing can effectively inhibit HSV replication in different cell lines, but also that the packaged lentivirus has a good effect. See Figure 9.
实施例9:LV-Cas9/gRNA对照病毒不能抑制HSV复制Example 9: LV-Cas9/gRNA control virus cannot inhibit HSV replication
在第0天,将5×10 4TU空载慢病毒和PBS注射到成年雄性C57BL/6小鼠(n=3)中。通过腹腔注射(80mg/kg)戊巴比妥钠麻醉动物,并放置在立体定位设备中。在手术和病毒注射过程中,所有动物都用异氟烷(1-1.5%)保持麻醉。用牙科钻头将目标区域坐标:V1(AP,-2.80mm;ML,-2.40mm;和DV,-0.90mm)上方的头骨磨薄,并小心地去除头骨碎片。将微量进样器连接到玻璃微管,尖端直径为10-15mm,用注射器泵以每分钟30nl的速度进行注射。注射完成后,将小鼠额外放置10分钟,然后慢慢退针。在手术后,应用盐酸林霉素和盐酸利多卡因凝胶涂抹缝合伤口以防止炎症并减轻小鼠的疼痛。在5天时,将500个H129-EGFP病毒粒子注射到相同脑区。 On day 0, 5 × 10 4 TU of empty lentivirus and PBS were injected into adult male C57BL/6 mice (n=3). Animals were anesthetized by intraperitoneal injection (80 mg/kg) of sodium pentobarbital and placed in a stereotaxic apparatus. All animals were kept anesthetized with isoflurane (1-1.5%) during surgery and virus injection. The skull above the target area coordinates: V1 (AP, -2.80 mm; ML, -2.40 mm; and DV, -0.90 mm) was thinned with a dental drill, and skull fragments were carefully removed. A microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel should be applied to the sutured wound to prevent inflammation and relieve pain in the mice. On day 5, 500 H129-EGFP viral particles were injected into the same brain region.
分别预先注射了PBS和LV-VEC在初级视觉皮层V1作为对照,然后在5天后注入H129-EGFP的实验结果显示,可以在观察到H129-EGFP注射位点和V1的下游输出区域,如LGD和对侧V1有大量的EGFP表达,说明病的的感染和复制都没有受到抑制。如图10。The results of the experiment in which PBS and LV-VEC were injected into the primary visual cortex V1 as controls and then H129-EGFP was injected 5 days later showed that a large amount of EGFP expression could be observed in the H129-EGFP injection site and the downstream output areas of V1, such as LGD and contralateral V1, indicating that the infection and replication of the disease were not inhibited (Figure 10).
实施例10:LV-Cas9/gRNA单病毒在体抗HSV复制效果Example 10: Effect of LV-Cas9/gRNA single virus on HSV replication in vivo
在第0天,将5×10 4TU实验组的分别表达agD-1,aICP4-2,aICP27-2,aVP16-g2的慢病毒注射到成年雄性C57BL/6小鼠(n=3)中。通过腹腔注射(80mg/kg)戊巴比妥钠麻醉动物,并放置在立体定位设备中。在手术和病毒注射过程中,所有动物都用异氟烷(1-1.5%)保持麻醉。用牙科钻头将目标区域坐标:V1(AP,-2.80mm;ML,-2.40mm;和DV,-0.90mm)上方的头骨磨薄,并小心地去除头骨碎片。将微量进样器连接到玻璃微管,尖端直径为10-15mm,用注射器泵以每分钟30nl的速度进行注射。注射完成后,将小鼠额外放置10分钟,然后慢慢退针。在手术后,应用盐酸林霉素和盐酸利多卡因凝胶涂抹缝合伤口以防止炎症并减轻小鼠的疼痛。在5天时,将500个H129-EGFP病毒粒子注射到相同脑区。 On day 0, 5×10 4 TU of experimental groups of lentivirus expressing agD-1, aICP4-2, aICP27-2, aVP16-g2, respectively, were injected into adult male C57BL/6 mice (n=3). Animals were anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg) and placed in a stereotaxic apparatus. All animals were kept anesthetized with isoflurane (1-1.5%) during surgery and virus injection. The skull above the target region coordinates: V1 (AP, -2.80 mm; ML, -2.40 mm; and DV, -0.90 mm) was thinned with a dental drill, and skull fragments were carefully removed. A microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel was applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
预先注射分别表达agD-1,aICP4-2,aICP27-2,aVP16-g2的慢病毒在初级视觉皮层V1,然后在5天后注入H129-EGFP,可以在观察到H129-EGFP注射位点V1的下游输出区域LGD和对侧V1都没有的EGFP表达,说明病的的感染和复制都受到明显抑制。如图11。Lentivirus expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into the primary visual cortex V1 in advance, and then H129-EGFP was injected 5 days later. It was observed that there was no EGFP expression in the downstream output area LGD of the H129-EGFP injection site V1 and the contralateral V1, indicating that the infection and replication of the virus were significantly inhibited (see Figure 11).
实施例11:LV-Cas9/gRNA病毒联用(Cocktail)策略抗HSV复制Example 11: LV-Cas9/gRNA virus cocktail strategy to prevent HSV replication
在第0天,将5×10 4TU实验组的分别表达agD-1,aICP4-2,aICP27-2,aVP16-g2的慢病毒均匀混合制剂注射到成年雄性C57BL/6小鼠(n=3)中。通过腹腔注射(80mg/kg)戊巴比妥钠麻醉动物,并放置在立体定位设备中。在手术和病毒注射过程中,所有动物都用异氟烷(1-1.5%)保持麻醉。用牙科钻头将目标区域坐标:V1(AP,-2.80mm;ML,-2.40mm;和DV,-0.90mm)上方的头骨磨薄,并小心地去除头骨碎片。将微量进样器连接到玻璃微管,尖端直径为10-15mm,用注射器泵以每分钟30nl的速度进行注射。注射完成后,将小鼠额外放置10分钟,然后慢慢退针。在手术后,应用盐酸林霉素和盐酸利多卡因凝胶涂抹缝合伤口以防止炎症并减轻小鼠的疼痛。在5天时,将500个H129-EGFP病毒粒子注射到相同脑区。 On day 0, 5×10 4 TU of a homogenous mixed preparation of lentivirus expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into adult male C57BL/6 mice (n=3). Animals were anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg) and placed in a stereotaxic apparatus. All animals were kept anesthetized with isoflurane (1-1.5%) during surgery and virus injection. The skull above the target area coordinates: V1 (AP, -2.80 mm; ML, -2.40 mm; and DV, -0.90 mm) was thinned with a dental drill, and skull fragments were carefully removed. A microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel were applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
可以在观察到H129-EGFP注射位点V1的下游输出区域LGD和对侧V1都没有的EGFP表达,说明病毒的的感染和复制都受到明显抑制。如图12。It can be observed that there is no EGFP expression in the downstream output region LGD of the H129-EGFP injection site V1 and the contralateral V1, indicating that the infection and replication of the virus are significantly inhibited (see Figure 12).
实施例12:LV-Cas9/gRNA病毒联用(Cocktail)剂量效应Example 12: LV-Cas9/gRNA virus combination (Cocktail) dose effect
在第0天,将1×10 5TU实验组的分别表达agD-1,aICP4-2,aICP27-2,aVP16-g2的慢病毒均匀混合制剂注射到成年雄性C57BL/6小鼠(n=3)中。通过腹腔注射(80mg/kg)戊巴比妥钠麻醉动物,并放置在立体定位设备中。在手术和病毒注射过程中,所有动物都用异氟烷(1-1.5%)保持麻醉。用牙科钻头将目标区 域坐标:V1(AP,-2.80mm;ML,-2.40mm;和DV,-0.90mm)上方的头骨磨薄,并小心地去除头骨碎片。将微量进样器连接到玻璃微管,尖端直径为10-15mm,用注射器泵以每分钟30nl的速度进行注射。注射完成后,将小鼠额外放置10分钟,然后慢慢退针。在手术后,应用盐酸林霉素和盐酸利多卡因凝胶涂抹缝合伤口以防止炎症并减轻小鼠的疼痛。在5天时,将500个H129-EGFP病毒粒子注射到相同脑区。 On day 0, 1×10 5 TU of a homogenous mixed preparation of lentivirus expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into adult male C57BL/6 mice (n=3). Animals were anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg) and placed in a stereotaxic apparatus. All animals were kept anesthetized with isoflurane (1-1.5%) during surgery and virus injection. The skull above the target area coordinates: V1 (AP, -2.80 mm; ML, -2.40 mm; and DV, -0.90 mm) was thinned with a dental drill, and skull fragments were carefully removed. A microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel were applied to the sutured wound to prevent inflammation and relieve pain in mice. On day 5, 500 H129-EGFP virus particles were injected into the same brain area.
可以在观察到H129-EGFP注射位点V1的下游输出区域LGD和对侧V1都没有的EGFP表达,说明病毒的的感染和复制都受到明显抑制。并且和只注射5×10 4TU的实验组相比,注射位点已观察不到神经元被病毒感染,说明LV-Cas9/gRNA病毒联用(Cocktail)具有剂量效应。如图13。 It can be observed that there is no EGFP expression in the downstream output area LGD and the contralateral V1 of the H129-EGFP injection site V1, indicating that the infection and replication of the virus are significantly inhibited. And compared with the experimental group injected with only 5×10 4 TU, no neurons were observed to be infected by the virus at the injection site, indicating that the LV-Cas9/gRNA virus combination (Cocktail) has a dose effect. As shown in Figure 13.
实施例13:LV-Cas9/gRNA病毒联用(Cocktail)治疗HSV感染效果Example 13: Effect of LV-Cas9/gRNA virus cocktail in treating HSV infection
在第0天,将500个H129-EGFP病毒粒子注射到相同脑区注射到成年雄性C57BL/6小鼠(n=3)中。通过腹腔注射(80mg/kg)戊巴比妥钠麻醉动物,并放置在立体定位设备中。在手术和病毒注射过程中,所有动物都用异氟烷(1-1.5%)保持麻醉。用牙科钻头将目标区域坐标:V1(AP,-2.80mm;ML,-2.40mm;和DV,-0.90mm)上方的头骨磨薄,并小心地去除头骨碎片。将微量进样器连接到玻璃微管,尖端直径为10-15mm,用注射器泵以每分钟30nl的速度进行病毒注射。注射完成后,将小鼠额外放置10分钟,然后慢慢退针。在手术后,应用盐酸林霉素和盐酸利多卡因凝胶涂抹缝合伤口以防止炎症并减轻小鼠的疼痛。在5天时,将5×10 4TU实验组的分别表达agD-1,aICP4-2,aICP27-2,aVP16-g2的慢病毒均匀混合制剂注射到相同脑区。 On day 0, 500 H129-EGFP viral particles were injected into the same brain region into adult male C57BL/6 mice (n=3). Animals were anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg) and placed in a stereotaxic apparatus. All animals were kept anesthetized with isoflurane (1-1.5%) during surgery and virus injection. The skull above the target area coordinates: V1 (AP, -2.80 mm; ML, -2.40 mm; and DV, -0.90 mm) was thinned with a dental drill, and skull fragments were carefully removed. A microinjector was connected to a glass micropipette with a tip diameter of 10-15 mm, and virus injections were performed at a rate of 30 nl per minute using a syringe pump. After the injection was completed, the mouse was left for an additional 10 minutes, and then the needle was slowly withdrawn. After surgery, lincomycin hydrochloride and lidocaine hydrochloride gel should be applied to the sutured wound to prevent inflammation and relieve pain in the mice. On day 5, 5×10 4 TU of lentivirus mixtures expressing agD-1, aICP4-2, aICP27-2, and aVP16-g2 were injected into the same brain region.
与两个对照组相比,可以在观察到H129-EGFP注射位点V1和对侧V1很少的EGFP表达,并且下游输出区域LGD没有EGFP表达,说明病毒的的感染和复制都受到较强抑制。如图14。Compared with the two control groups, very little EGFP expression can be observed in the H129-EGFP injection site V1 and the contralateral V1, and no EGFP expression in the downstream output area LGD, indicating that the infection and replication of the virus are strongly inhibited (see Figure 14).

Claims (19)

  1. 一种靶向HSV病毒必需基因的gRNA,其特征在于,所述gRNA选自SEQ ID NO.1-SEQ ID NO.10所示核苷酸序列中的一种。A gRNA targeting an essential gene of the HSV virus, characterized in that the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
  2. 根据权利要求1所述的gRNA,其特征在于,所述gRNA选自SEQ ID NO.2、SEQ ID NO.4、SEQ ID NO.6和SEQ ID NO.7所示核苷酸序列中的一种。The gRNA according to claim 1 is characterized in that the gRNA is selected from one of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
  3. 一种靶向HSV病毒必需基因的gRNA组合物,其特征在于,所述gRNA组合物选自SEQ ID NO.1-SEQ ID NO.10所示核苷酸序列中的两种或以上。A gRNA composition targeting essential genes of HSV virus, characterized in that the gRNA composition is selected from two or more nucleotide sequences shown in SEQ ID NO.1-SEQ ID NO.10.
  4. 根据权利要求3所述的gRNA组合物,其特征在于,所述gRNA组合物为SEQ ID NO.2、SEQ ID NO.4、SEQ ID NO.6和SEQ ID NO.7所示核苷酸序列的组合物。The gRNA composition according to claim 3 is characterized in that the gRNA composition is a composition of the nucleotide sequences shown in SEQ ID NO.2, SEQ ID NO.4, SEQ ID NO.6 and SEQ ID NO.7.
  5. 一种靶向HSV病毒必需基因的gRNA表达载体,其特征在于,其包含权利要求1或2所述gRNA,或权利要求3或4所述gRNA组合物。A gRNA expression vector targeting an essential gene of the HSV virus, characterized in that it comprises the gRNA described in claim 1 or 2, or the gRNA composition described in claim 3 or 4.
  6. 根据权利要求5所述的gRNA表达载体,其特征在于,所述表达载体为病毒载体或非病毒载体;The gRNA expression vector according to claim 5, characterized in that the expression vector is a viral vector or a non-viral vector;
    所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒;The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle;
    所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
  7. 一种CRISPR/Cas基因编辑系统,其特征在于,其包含权利要求1或2所述gRNA,或权利要求3或4所述gRNA组合物,以及Cas蛋白。A CRISPR/Cas gene editing system, characterized in that it comprises the gRNA described in claim 1 or 2, or the gRNA composition described in claim 3 or 4, and a Cas protein.
  8. 根据权利要求7所述的CRISPR/Cas基因编辑系统,其特征在于,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白、脑炎双球菌Cas9、嗜热链球菌Cas9、金黄色葡萄球菌Cas9、氨基酸球菌Cpf1、谷氨酸棒杆菌Cpf1、金黄色葡萄球菌微型SaCas9、口腔纤毛菌C2c2的一种或多种;The CRISPR/Cas gene editing system according to claim 7, characterized in that the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, encephalococcus Cas9, Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, Acidaminococcus Cpf1, Corynebacterium glutamicum Cpf1, Staphylococcus aureus micro-SaCas9, and oral filamentous fungi C2c2;
    优选地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白。Preferably, the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
  9. 权利要求7或8所述CRISPR/Cas基因编辑系统的递送系统,其特征在于,所述递送系统包括:The delivery system of the CRISPR/Cas gene editing system according to claim 7 or 8, characterized in that the delivery system comprises:
    1)权利要求5或6所述的gRNA表达载体;1) The gRNA expression vector according to claim 5 or 6;
    2)Cas蛋白表达载体。2) Cas protein expression vector.
  10. 根据权利要求9所述的递送系统,其特征在于,所述Cas蛋白表达载体为病毒载体或非病毒载体;The delivery system according to claim 9, characterized in that the Cas protein expression vector is a viral vector or a non-viral vector;
    所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒;The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle;
    所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
  11. 一种试剂盒,其特征在于,其包括:A kit, characterized in that it comprises:
    1)权利要求1或2所述gRNA,或权利要求3或4所述gRNA组合物,或权 利要求5或6所述的gRNA表达载体;1) The gRNA according to claim 1 or 2, or the gRNA composition according to claim 3 or 4, or the gRNA expression vector according to claim 5 or 6;
    2)Cas蛋白表达载体。2) Cas protein expression vector.
  12. 根据权利要求11所述的试剂盒,其特征在于,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白、脑炎双球菌Cas9、嗜热链球菌Cas9、金黄色葡萄球菌Cas9、氨基酸球菌Cpf1、谷氨酸棒杆菌Cpf1、金黄色葡萄球菌微型SaCas9、口腔纤毛菌C2c2的一种或多种;The kit according to claim 11, characterized in that the Cas protein is one or more of SpCas9 protein derived from Streptococcus pyogenes, encephalococcus Cas9, Streptococcus thermophilus Cas9, Staphylococcus aureus Cas9, Acidococcus ammoniae Cpf1, Corynebacterium glutamicum Cpf1, Staphylococcus aureus micro-SaCas9, and oral filamentous fungi C2c2;
    优选地,所述Cas蛋白为化脓性链球菌来源的SpCas9蛋白。Preferably, the Cas protein is SpCas9 protein derived from Streptococcus pyogenes.
  13. 根据权利要求11所述的试剂盒,其特征在于,所述Cas蛋白表达载体为病毒载体或非病毒载体;The kit according to claim 11, characterized in that the Cas protein expression vector is a viral vector or a non-viral vector;
    所述病毒载体为慢病毒载体、逆转录病毒载体、腺相关病毒载体、单纯疱疹病毒载体、腺病毒载体或病毒样颗粒;The viral vector is a lentiviral vector, a retroviral vector, an adeno-associated viral vector, a herpes simplex viral vector, an adenoviral vector or a virus-like particle;
    所述非病毒载体为质粒或mRNA。The non-viral vector is a plasmid or mRNA.
  14. 权利要求1或2所述gRNA,或权利要求3或4所述gRNA组合物,或权利要求5或6所述的gRNA表达载体,或权利要求7或8所述CRISPR/Cas基因编辑系统,或权利要求9或10所述递送系统,或权利要求11-13任一项所述试剂盒在制备预防和/或治疗HSV感染相关疾病的药物中的应用。Use of the gRNA described in claim 1 or 2, or the gRNA composition described in claim 3 or 4, or the gRNA expression vector described in claim 5 or 6, or the CRISPR/Cas gene editing system described in claim 7 or 8, or the delivery system described in claim 9 or 10, or the kit described in any one of claims 11-13 in the preparation of drugs for preventing and/or treating diseases related to HSV infection.
  15. 根据权利要求14所述的应用,其特征在于,所述HSV感染相关疾病为HSV-1型或HSV-2型相关疾病;The use according to claim 14, characterized in that the HSV infection-related disease is an HSV-1 or HSV-2-related disease;
    优选地,所述HSV感染相关疾病包括病毒性脑炎、单纯疱疹病毒性角膜炎和感染性失明。Preferably, the HSV infection-related diseases include viral encephalitis, herpes simplex virus keratitis and infectious blindness.
  16. 权利要求1或2所述gRNA,或权利要求3或4所述gRNA组合物,或权利要求5或6所述的gRNA表达载体,或权利要求7或8所述CRISPR/Cas基因编辑系统,或权利要求9或10所述递送系统,或权利要求11-13任一项所述试剂盒在制备预防和/或治疗抗HSV药物中的应用。Use of the gRNA of claim 1 or 2, or the gRNA composition of claim 3 or 4, or the gRNA expression vector of claim 5 or 6, or the CRISPR/Cas gene editing system of claim 7 or 8, or the delivery system of claim 9 or 10, or the kit of any one of claims 11 to 13 in the preparation of drugs for the prevention and/or treatment of anti-HSV.
  17. 根据权利要求16所述的应用,其特征在于,所述HSV为HSV-1或HSV-2。The use according to claim 16, characterized in that the HSV is HSV-1 or HSV-2.
  18. 一种药物组合物,其特征在于,其包括权利要求9或10所述CRISPR/Cas基因编辑系统的递送系统。A pharmaceutical composition, characterized in that it comprises the delivery system of the CRISPR/Cas gene editing system according to claim 9 or 10.
  19. 一种抑制HSV复制和/或HSV病毒必需基因表达的方法,其特征在于,所述方法包括:在HSV感染前或感染后,将权利要求9或10所述CRISPR/Cas基因编辑系统的递送系统转染目的细胞,细胞表达的Cas-gRNA形成的核糖核蛋白复合物靶向的精准剪切破坏HSV基因组,阻断病毒复制和传播。A method for inhibiting HSV replication and/or HSV virus essential gene expression, characterized in that the method comprises: before or after HSV infection, transfecting the target cells with the delivery system of the CRISPR/Cas gene editing system according to claim 9 or 10, and the ribonucleoprotein complex formed by the Cas-gRNA expressed by the cells targets and precisely shears and destroys the HSV genome, thereby blocking viral replication and transmission.
PCT/CN2022/135820 2022-12-01 2022-12-01 Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use WO2024113302A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/135820 WO2024113302A1 (en) 2022-12-01 2022-12-01 Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/135820 WO2024113302A1 (en) 2022-12-01 2022-12-01 Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use

Publications (1)

Publication Number Publication Date
WO2024113302A1 true WO2024113302A1 (en) 2024-06-06

Family

ID=91322750

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/135820 WO2024113302A1 (en) 2022-12-01 2022-12-01 Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use

Country Status (1)

Country Link
WO (1) WO2024113302A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992005284A1 (en) * 1990-09-21 1992-04-02 University Of Maryland Compositions and methods for inhibiting growth or replication of viruses
CN107580504A (en) * 2015-01-14 2018-01-12 天普大学-联邦高等教育系统 The I types herpes simplex virus of RNA guiding and the radical cure of other associated herpesvirus
WO2018017925A1 (en) * 2016-07-22 2018-01-25 President And Fellows Of Harvard College Targeting lytic and latent herpes simplex virus 1 infection with crispr/cas9
US20180251770A1 (en) * 2015-10-30 2018-09-06 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating herpes simplex virus
CN109468318A (en) * 2017-09-08 2019-03-15 中山大学 For inhibiting HSV-1 to replicate and/or CRISPR/Cas9 system, method, kit and its application of target sequence expression
WO2022006745A1 (en) * 2020-07-07 2022-01-13 Oriengene Biotechnology Ltd Guide rna for hsv-1 gene editing and method thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992005284A1 (en) * 1990-09-21 1992-04-02 University Of Maryland Compositions and methods for inhibiting growth or replication of viruses
CN107580504A (en) * 2015-01-14 2018-01-12 天普大学-联邦高等教育系统 The I types herpes simplex virus of RNA guiding and the radical cure of other associated herpesvirus
US20180251770A1 (en) * 2015-10-30 2018-09-06 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating herpes simplex virus
WO2018017925A1 (en) * 2016-07-22 2018-01-25 President And Fellows Of Harvard College Targeting lytic and latent herpes simplex virus 1 infection with crispr/cas9
CN109468318A (en) * 2017-09-08 2019-03-15 中山大学 For inhibiting HSV-1 to replicate and/or CRISPR/Cas9 system, method, kit and its application of target sequence expression
WO2022006745A1 (en) * 2020-07-07 2022-01-13 Oriengene Biotechnology Ltd Guide rna for hsv-1 gene editing and method thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KHODADAD NASTARAN, FANI MONA, JAMEHDOR SALEH, NAHIDSAMIEI RAHIL, MAKVANDI MANOOCHEHR, KABOLI SAEED, TEIMOORI ALI, THEKKINIATH JOSE: "A knockdown of the herpes simplex virus type-1 gene in all-in-one CRISPR vectors", FOLIA HISTOCHEMICA ET CYTOBIOLOGICA, VESALIUS UNIVERSITY MEDICAL PUBLISHER, KRAKOW,, PL, vol. 58, no. 3, 1 January 2020 (2020-01-01), PL , pages 174 - 181, XP093177742, ISSN: 0239-8508, DOI: 10.5603/FHC.a2020.0020 *
OH HYUNG SUK, NEUHAUSSER WERNER M, EGGAN PIERCE, ANGELOVA MAGDALENA, KIRCHNER RORY, EGGAN KEVIN C, KNIPE DAVID M: "Herpesviral lytic gene functions render the viral genome susceptible to novel editing by CRISPR/Cas9", ELIFE, ELIFE SCIENCES PUBLICATIONS LTD., GB, vol. 8, 2 December 2019 (2019-12-02), GB , XP093177737, ISSN: 2050-084X, DOI: 10.7554/eLife.51662 *

Similar Documents

Publication Publication Date Title
US20230167425A1 (en) Rna guided eradication of varicella zoster virus
US20230001016A1 (en) Rna guided eradication of human jc virus and other polyomaviruses
JP7176737B2 (en) Excision of retroviral nucleic acid sequences
WO2017213896A1 (en) Negative feedback regulation of hiv-1 by gene editing strategy
JP2019500909A (en) Oncolytic virus strain
US20210252135A1 (en) Treatment using oncolytic virus
JP6924487B2 (en) Non-embedded virus delivery system and how to use it
US20120027725A1 (en) Safe lentiviral vectors for targeted delivery of multiple therapeutic molecules to treat liver cancer
JP2019520814A (en) Non-integrated virus delivery system and related method
JP2022513639A (en) Low neurotoxic HSV vector
US20100041741A1 (en) Suppression of mitochondrial oxidative stress
WO2024011980A1 (en) Gene therapy drug for herpes simplex virus infectious diseases
JP5037775B2 (en) Use of parapoxvirus ovis for the manufacture of antiviral drugs and medicaments against cancer
WO2021175293A1 (en) Herpes simplex virus and use thereof
WO2020112471A1 (en) New generation regulatable fusogenic oncolytic herpes simplex virus type 1 virus and methods of use
WO2020238427A1 (en) Oncolytic virus system for specifically killing tumor cells, and application thereof
WO2024113302A1 (en) Grna targeting hsv essential genes, crispr/cas gene editing system, delivery system, and use
US20190085326A1 (en) Negative feedback regulation of HIV-1 by gene editing strategy
CN118127004A (en) GRNA of target HSV essential gene, CRISPR/Cas gene editing system, delivery system and application
CN116724110A (en) Recombinant oncolytic virus and construction method and application thereof
CN112972651B (en) Application of IFITMs in preparation of EBV (Epstein Barr virus) epithelial infection inhibitor and epithelial tumor prevention and treatment drugs
EP4335448A1 (en) Modified oncolytic parvovirus h-1pv with an enhanced fitness and superior anticancer activity
JP2024512053A (en) Oncolytic herpes simplex virus vectors subject to dual transcriptional and translational regulation
CN116323947A (en) Nucleic acid molecule for encoding Kras gene mutant
BRPI0112407B1 (en) USE OF OVIS PARAPOXVIRUS CEPAS TO PRODUCE ANTIVIAL DRUGS AND CANCER DRUGS